Sie sind auf Seite 1von 212

Volume 142

Number 5

September 3, 2010

www.cell.com

Reorienting
Planar Polarity
Minireview
Histone Crosstalk
bioreprogramming

Biopotential.
Unlock extraordinary potential with stem cell technologies from Sigma . ®

Stem cell biology offers astonishing


research potential; Sigma® Life Science has
the innovations you need to discover the
promise it holds. Access a world of RNAi with
the MISSION® RNAi Library, efficiently edit
genes of interest using advanced CompoZr®
ZFN technology, and characterize your stem
cells with our Prestige Antibodies®, powered
by Atlas Antibodies. The applications are
endless—and so are the possibilities.

wherebiobegins.com/bioreprogramming

Sigma, MISSION, CompoZr and Prestige Antibodies are registered trademarks of


Sigma-Aldrich and its affiliate Sigma-Aldrich Biotechnology, L.P.
.
E W Intelligent design

N +
inCuSaFe™ copper enriched
stainless steel interior

+ Single-beam, dual capture


infrared CO2 sensor

+ SafeCell UV
protection in situ

+ Hydrogen peroxide
vapor sterilization in situ

1
Good laboratory technique

Sterisonic™ GxP
Performance and Productivity
Delivers Best Efficiency Value:

2 Hours 14 Hours

SANYO Brand X
Sterisonic™

H2O2 sterilization vs. high heat sterilization


= Uptime (Hours) = Downtime (Hours)

The rapid in situ H2O2 sequence returns the


fully sterilized Sterisonic™ GxP to normal use quicker

Spot on results.
than any competitive incubator in the world.

The industry’s first in situ H2O2 sterilization with the fastest turnaround.
For maximum productivity in clinical, general purpose or the most highly compliant GMP applications,
the new SANYO Sterisonic™ GxP CO2 incubator offers an impressive return on investment. With multiple
contamination control safeguards, exclusive on-board H2O2 sterilization, FDA-21CFR data capture and
graphical LCD display, the Sterisonic™ GxP rewards good laboratory technique with performance
you can trust. Learn more, visit www.sterisonic.com or call 800-858-8442.

pictured above: Sterisonic™ GxP MCO-19AICUVH with rapid H2O2 vapor sterilization system.

FREEER
OFF
[ D E TA IL
S
!
O N L IN E ]
FREE!
FREE!
H2O2 sterilization system acessory, plus BD labware consumables kit.
Limited time upgrade offer with purchase of the Sterisonic™ GxP MCO 19AICUVH. ($900 Value)
BD stem cell starter kit with Sterisonic™ GxP quote.
No purchase necessary! Act now. Supplies are limited. ($150 Value)
www.sterisonic.com
©2009 Sanyo Biomedical OWS 1015 05/09
015.A1.0101.F.US © 2009 Eppendorf AG.
Background: electroporated L-mouse cells.
 Electroporation of
mammalian cells

 Electroporation
of bacteria

 Electroporation
of yeasts

 Cell fusion

Outstanding
transfection
rates!

One electroporator– many applications.


Our Multiporator ® is a compact, flexible system A safe and easy method for performing cell fusion:
designed for the electroporation of eukaryotic cells,  Microfusion chamber for viewing and optimization
bacteria and other microorganisms, as well as the of fusion parameters
electrofusion of cells.  Helix chamber for larger cell volumes
 Applications include: monoclonal antibodies, plant
Unique features include Soft Pulse microsecond pulses, protoplast fusion, tumor research, cancer vaccines,
patented* electronically regulated pulses, a gentle cloning mammals
hypoosmolar buffer system and a choice of
electroporation and electrofusion options. For more information visit www.eppendorf.com

*U.S. Pat. 6,008,038

www.eppendorf.com • Email: info@eppendorf.com • Application hotline: 516-515-2258


In the U.S.: Eppendorf North America, Inc. 800-645-3050 • In Canada: Eppendorf Canada Ltd. 800-263-8715
If your standards and your samples are the same size,
they must be the same mass, right?
That’s the problem with calibration-based measuring techniques. You don’t know—you can only assume. Which is why
every major pharmaceutical and biotechnology company, as well as most federal regulatory agencies are switching from relative
methods to Wyatt Technology’s absolute measurements. Our DAWN® multi-angle light scattering (MALS) instruments
allow you to determine absolute molecular weights and sizes without relying on so-called standards, or measurements made in
someone else’s lab. Wyatt instruments measure all of the quantities required for determining absolute molar masses directly.
If you’re ready to abandon the habit and hope of reference standards, call 805-681-9009 or visit wyatt.com to request our
free 28-page Ultimate Guide to Light Scattering. Our booklet will show you how
to make your reliance on standards a thing of the past. CORPORA
CORPORATION

DAWN HELEOS®. The most advanced Optilab rEX®. The refractometer with ViscoStar™. The viscometer with Eclipse. The ultimate system for the DynaPro Plate Reader. Automated
multi-angle light scattering instrument the greatest sensitivity and range. unparalleled signal-to-noise, stable separation of macromolecules and dynamic light scattering for 96 or
for macromolecular characterization. baselines and a 21st-century interface. nanoparticles in solution. 384 or 1536 well plates.

©2008 Wyatt Technology. DAWN, Optilab, DynaPro and the Wyatt Technology logo are registered trademarks, and ViscoStar and Eclipse are trademarks of Wyatt Technology Corporation.
BE THE FIRSTto read the latest issue of any Cell Press journal.

Register for Cell Press Email Alerts and get the complete table of contents as soon as
the issue publishes online — FREE!

Cell Press Email Alerts deliver the news, research, and commentaries featured in each
journal’s latest issue, including the full title of every article, direct links to the articles,
and the complete author list. Plus, to save you time, each research article has a brief
summary highlighting its significant findings.

You don’t have to be a subscriber to sign up for Cell Press Email Alerts. While subscribers have
instant access to the full text of all articles listed in the Email Alerts, non-subscribers can read
the abstracts of all articles as well as the full text of the issue’s Featured Article.

www.cellpress.com
Editor Editorial Board Douglas Green Dana Pe’er
Emilie Marcus Abul Abbas Leonard Guarente Kathrin Plath
C. David Allis Taekjip Ha Carol Prives
Senior Deputy Editor
Geneviève Almouzni Daniel Haber Klaus Rajewsky
Elena Porro
Uri Alon Ulrike Heberlein Venki Ramakrishnan
Scientific Editors Angelika Amon Nobutaka Hirokawa Rama Ranganathan
Karen Carniol Johan Auwerx Mark Hochstrasser Anne Ridley
Connie M. Lee Richard Axel Arthur Horwich James Roberts
Fabiola Rivas Cori Bargmann Tony Hunter Alexander Rudensky
Lara Szewczak Konrad Basler James Hurley Helen Saibil
Bonnie Bassler Richard Hynes Joshua Sanes
Leading Edge Editor
David Baulcombe Thomas Jessell Randy Schekman
Orla M. Smith
Jeffrey Benovic Narry Kim Ueli Schibler
Associate Editor Carolyn Bertozzi Mary-Claire King Joseph Schlessinger
Robert Kruger Wendy Bickmore David Kingsley Hans Schöler
Elizabeth Blackburn Frank Kirchhoff Trina Schroer
Assistant Editor Joan Brugge Richard Kolodner Geraldine Seydoux
Michaeleen Doucleff Lewis Cantley John Kuriyan Kevan Shokat
Senior Managing Editor Joanne Chory Robert Lamb Pamela Sklar
Meredith Adinolfi David Clapham Mark Lemmon Nahum Sonenberg
Andrew Clark Beth Levine James Spudich
Deputy Managing Editor Hans Clevers Wendell Lim Paul Sternberg
Jackie M. Smith Stephen Cohen Jennifer Lippincott-Schwartz Bruce Stillman
Illustration and Pascale Cossart Dan Littman Azim Surani
Design Supervisor Jeff Dangl Richard Losick Keiji Tanaka
Nathan E. Bress Ted Dawson Scott Lowe Craig Thompson
Pier Paolo di Fiore Tom Maniatis Robert Tjian
Lead Illustrator Marileen Dogterom Matthias Mann Jürg Tschopp
Andrew A. Tang Julian Downward Kelsey Martin Ulrich von Andrian
Bruce Edgar Joan Massagué Gerhard Wagner
Illustrators
Steve Elledge Iain Mattaj Detlef Weigel
Yvonne Blanco
Anne Ephrussi Satyajit Mayor Alan Weiner
Kate Mahan
Ronald Evans Ruslan Medzhitov Jonathan Weissman
Production Staff Witold Filipowicz Craig Mello Matthew Welch
Reyna Clancy Marco Foiani Tom Misteli Tian Xu
Elaine Fuchs Tim Mitchison Shinya Yamanaka
Editorial Assistant
Yukiko Goda Alex Mogilner Marino Zerial
Mary Beth O’Leary
Stephen Goff Paul Nurse Xiaowei Zhuang
Joe Goldstein Roy Parker Huda Zoghbi

Cell Office
Cell, Cell Press, 600 Technology Square, 5th Floor, Cambridge, Massachusetts 02139
Phone: (+1) 617 661 7057, Fax: (+1) 617 661 7061, E-mail: celleditor@cell.com
Online Publication: http://www.cell.com

Cell (ISSN 0092-8674) is published biweekly by Cell Press, 600 Technology Square, 5th Floor, Cambridge, Massachusetts 02139. The institutional subscription rate for
2010 is $1,360 (US and Canada) or $1,532 (elsewhere). The individual subscription rate is $202 (US and Canada) or $305 (elsewhere). The individual copy price is $50.
Periodicals postage paid at Boston, Massachusetts and additional mailing offices. Postmaster: send address changes to Elsevier Customer Service Americas,
Cell Press Journals, 11830 Westline Industrial Drive, St. Louis, MO 63146, USA.
The paper used in this publication meets the requirments of ANSI/NISO Z39.48-1992 (Permanence of Paper). Printed by Dartmouth Printing Company, Hanover, NH.
FINE SURGICAL INSTRUMENTS FOR RESEARCH TM

The Elements of Discovery


Scissors • Needle Holders • Forceps • Retractors • Clamps • Rongeurs • Probes
Scalpels • Surgical & Laboratory Accessories • Student Quality Instruments

R e q u e s t a c a t a l o g a t f i n e s c i e n c e. c o m o r c a l l 1 - 8 0 0 - 5 2 1 - 2 1 0 9

Scopus is the largest abstract and citation


database of peer-reviewed literature and
quality web sources with smart tools to
track, analyze and visualize research.

enrich
your
experience
www.scopus.com
Cell Press Display Advertising
Midwest/Southeast/Eastern Canada:
President & CEO Inez Herrero, ph: 585 678 4395; fax: 585 678 4722;
Lynne Herndon e-mail: i.herrero@elsevier.com

Editor in Chief, Vice President of Content Development Mid-Atlantic/New England: Vikki Macomber, ph: 508 928
Emilie Marcus 1255; fax: 508 928 1256; e-mail: v.macomber@elsevier.com

Vice President of Marketing and Publishing West Coast/Western Canada/Asia: Lynne Stickrod, ph: 415
Els Bosma 931 9782; fax: 415 520 6940; e-mail: l.stickrod@elsevier.com

Vice President of Web Development and Operations UK/Europe: James Kenney, ph: +44 20 7424 4216; fax: +44 18
Keith Wollman 6585 3136; e-mail: j.kenney@elsevier.com

Director of Marketing Classified Advertising


Jonathan Atkinson United States and Canada:
Gordon Sheffield, Key Account Manager, ph: 617 386 2189; fax:
Production Supervisor
617 397 2805; e-mail: g.sheffield@elsevier.com
Meredith Adinolfi
UK, Europe, and Asia:
Operations Supervisor
Sabrina Dodge, Key Account Manager, ph: +44 20 7424 4997;
Daniel Fauxsmith
fax: +44 18 6585 3136; e-mail: s.dodge@elsevier.com
Press Officer
Cathleen Genova

ª2010 Elsevier Inc. All rights reserved. or ideas contained in the material herein. Because of rapid advances in
This journal and the individual contributions contained in it are protected the medical sciences, in particular, independent verification of diagnoses
under copyright by Elsevier Inc., and the following terms and conditions and drug dosages should be made. Although all advertising material is
apply to their use: expected to conform to ethical (medical) standards, inclusion in this
Photocopying: publication does not constitute a guarantee or endorsement of the quality
Single photocopies of single articles may be made for personal use as al- or value of such product or of the claims made of it by its manufacturer.
lowed by national copyright laws. Permission of the Publisher and payment Reprints:
of a fee are required for all other photocopying, including multiple or system- Article reprints are available through Cell’s reprint service; for informa-
atic copying, copying for advertising or promotional purposes, resale, and tion, contact Nicholas Pavlow (e-mail: n.pavlow@elsevier.com; ph: (+1)
all forms of document delivery. Special rates are available for educational 212 633 3960).
institutions that wish to make photocopies for nonprofit educational class- Subscription Orders and Inquiries:
room use. For information on how to seek permission, visit www.elsevier. Mail, fax, or e-mail address changes to Elsevier Customer Service Amer-
com/permissions or call (+44) 1865 843830 (UK) / (+1) 215 239 3804 (US). icas, allowing 4–6 weeks for processing. Lost or damaged issues will be
Permissions: replaced, subject to availability, if Cell Press is notified within the claim
For information on how to seek permission, visit www.elsevier.com/ period (US and airmail delivery: 3 months from issue date; surface deliv-
permissions or call (+44) 1865 843830 (UK) / (+1) 215 239 3804 (US). ery: 4 months from issue date). Periodical delivery in the US can take up
Derivative Works: to 3 weeks. Airmail delivery can take 2–4 weeks.
Subscribers may reproduce tables of contents or prepare lists of articles The price of a single copy of Cell is $50 (excluding special issues). All
including summaries for internal circulation within their institutions. orders must be prepaid and in writing. Please include the volume and is-
Permission of the Publisher is required for resale or distribution outside sue number, payment (check or credit card, MasterCard, Visa, or Amer-
the institution. Permission of the Publisher is required for all other deriv- ican Express only), and a delivery address. Allow 4–6 weeks for delivery.
ative works, including compilations and translations (please consult Mailing address: Elsevier Customer Service Americas, Cell Press
www.elsevier.com/permissions). Journals, 11830 Westline Industrial Drive, St. Louis, MO 63146,
USA. Toll-free phone within USA/Canada: 866 314 2355; phone for
Electronic Storage or Usage:
outside US/Canada: (+1) 314 453 7038; fax: (+1) 314 523 5170; e-mail:
Permission of the Publisher is required to store or use electronically any
subs@cell.com; internet: www.cellpress.com or <www.cell.com>.
material contained in this journal, including any article or part of an article
(please consult www.elsevier.com/permissions). Except as outlined Funding Body Agreements and Policies:
above, no part of this publication may be reproduced, stored in a retrieval Elsevier has established agreements and developed policies to allow au-
system, or transmitted in any form or by any means, electronic, mechan- thors whose articles appear in journals published by Elsevier to comply
ical, photocopying, recording, or otherwise, without prior written permis- with potential manuscript archiving requirements as specified as condi-
sion of the Publisher. tions of their grant awards. To learn more about existing agreements and
Notice: policies, visit www.cellpress.com.
No responsibility is assumed by the Publisher for any injury and/or damage Guide for Authors:
to persons or property as a matter of products liability, negligence, or other- For a full and complete guide for authors, please go to www.cell.com/
wise, or from any use or operation of any methods, products, instructions, authors.
REMARKABLY FAST

Phusion® High-Fidelity DNA Polymerase


With Phusion® High-Fidelity DNA Polymerase, there is no need to compromise any aspect of your PCR performance.
A superior choice for cloning, this recombinant polymerase has an error rate 50-fold lower than Taq DNA Polymerase,
combining extreme precision with unparalleled speed and robustness.

Advantages:
Not all PCR polymerases are created equal
• Extreme fidelity • High yield
• High speed • Specificity
• Robustness

Go to www.neb.com/phusion to find out


how Phusion High-Fidelity DNA Polymerase can
improve your PCR performance.

Produced by Distributed by
Amplification of a 3.8 kb fragment from the human beta globin gene clearly illustrates the
extreme speed and robustness offered by using Phusion DNA Polymerase. Reactions were
performed according to the suppliers’ recommendations using varying extension times
(shown above gel).

Phusion is a registered trademark of Finnzymes Oy.

DNA AMPLIFICATION PROTEIN EXPRESSION GENE EXPRESSION


CLONING & MAPPING RNA ANALYSIS
& PCR & ANALYSIS & CELLULAR ANALYSIS
www.neb.com
Leading Edge
Cell Volume 142 Number 5, September 3, 2010

IN THIS ISSUE
MOLECULAR BIOLOGY SELECT

ESSAY
661 Phosphotyrosine Signaling: Evolving a New W.A. Lim and T. Pawson
Cellular Communication System

CORRESPONDENCE

668 What Controls T Cell Receptor R.A. Fernandes, C. Yu, A.M. Carmo, E.J. Evans,
Phosphorylation? P.A. van der Merwe, and S.J. Davis
669 Response: Multilayered Control of T Cell E. Gagnon, C. Xu, W. Yang, H.H. Chu, M.E. Call,
Receptor Phosphorylation J.J. Chou, and K.W. Wucherpfennig

PREVIEWS
672 Fishing Out a Sensor for Anti-inflammatory Oils A.R. Saltiel

674 A New Spin on Planar Cell Polarity P. Olguin and M. Mlodzik


676 Viable Rat-Mouse Chimeras: D. Solter
Where Do We Go from Here?

679 ‘Fore Brain: L.B. Sweeney and L. Luo


A Hint of the Ancestral Cortex

MINIREVIEW

682 The Language of Histone Crosstalk J.-S. Lee, E. Smith, and A. Shilatifard

SNAPSHOT

822 Nuclear Receptors I N.J. McKenna and B.W. O’Malley


www.phosphosite.org

A Comprehensive Online
Protein Modification Resource
provided by Cell Signaling Technology with grant support from the NIH

Cell Signaling Technology® and PhosphoSitePlus® are trademarks of Cell Signaling Technology, Inc.
© 2010 Cell Signaling Technology, Inc.
PhosphoSitePlus® v3.0 (PSP) is an open web • Expansive and continuously curated content
resource that integrates encyclopedic information • Molecular rendering to visualize the location of modification sites
on experimentally determined protein modification
• On-the-fly generation of kinase substrate sequence logos
sites, upstream and downstream regulation of
• Browsing of high-throughput content by disease, cell line, and tissue
these modifications, and powerful analytical tools
for investigating the structural and biological • New search interfaces that retrieve modification sites and proteins by
significance of protein modifications. subcellular locations, sequence and motifs, domains, responsiveness
to treatments, disease, tissue, and cell type

Unparalleled product quality, validation and technical support.


www.cellsignal.com
Orders (toll-free) 1-877-616-2355 | Technical support (toll-free) 1-877-678-8324 support@cellsignal.com | Inquiries info@cellsignal.com | Environmental Commitment eco.cellsignal.com
Articles
Cell Volume 142 Number 5, September 3, 2010

687 GPR120 Is an Omega-3 Fatty Acid Receptor D.Y. Oh, S. Talukdar, E.J. Bae, T. Imamura,
Mediating Potent Anti-inflammatory H. Morinaga, W. Fan, P. Li, W.J. Lu,
and Insulin-Sensitizing Effects S.M. Watkins, and J.M. Olefsky

699 Anti-CD47 Antibody Synergizes with M.P. Chao, A.A. Alizadeh, C. Tang, J.H. Myklebust,
Rituximab to Promote Phagocytosis and B. Varghese, S. Gill, M. Jan, A.C. Cha, C.K. Chan,
Eradicate Non-Hodgkin Lymphoma B.T. Tan, C.Y. Park, F. Zhao, H.E. Kohrt, R. Malumbres,
J. Briones, R.D. Gascoyne, I.S. Lossos, R. Levy,
I.L. Weissman, and R. Majeti

714 A C-Type Lectin Collaborates with a G. Cheng, J. Cox, P. Wang, M.N. Krishnan,
CD45 Phosphatase Homolog to Facilitate J. Dai, F. Qian, J.F. Anderson, and E. Fikrig
West Nile Virus Infection of Mosquitoes

726 The ATAC Acetyltransferase Complex T. Suganuma, A. Mushegian, S.K. Swanson,


Coordinates MAP Kinases S.M. Abmayr, L. Florens, M.P. Washburn,
to Regulate JNK Target Genes and J.L. Workman

737 A Bacterial mRNA Leader that Employs S.-Y. Park, M.J. Cromie, E.-J. Lee,
Different Mechanisms to Sense and E.A. Groisman
Disparate Intracellular Signals

749 Structural Basis of Semaphorin-Plexin H. Liu, Z.S. Juo, A.H.-R. Shim, P.J. Focia, X. Chen,
Recognition and Viral Mimicry from K.C. Garcia, and X. He
Sema7A and A39R Complexes with PlexinC1

762 Photoadaptation in Neurospora by E. Malzahn, S. Ciprianidis, K. Kaldi, T. Schafmeier,


Competitive Interaction of Activating and M. Brunner
and Inhibitory LOV Domains

773 Cell Flow Reorients the Axis B. Aigouy, R. Farhadifar, D.B. Staple, A. Sagner,
of Planar Polarity in the J.-C. Ro€per, F. Ju€licher, and S. Eaton
Wing Epithelium of Drosophila

787 Generation of Rat Pancreas in Mouse T. Kobayashi, T. Yamaguchi, S. Hamanaka,


by Interspecific Blastocyst Injection M. Kato-Itoh, Y. Yamazaki, M. Ibata, H. Sato,
of Pluripotent Stem Cells Y.-S. Lee, J.-i. Usui, A.S. Knisely, M. Hirabayashi,
and H. Nakauchi

(continued)
Let the Countess™ do the counting.
Wow! Consistent, fast, and easy.
The Countess™ Automated Cell Counter takes the subjectivity and tedium out
of one of the fundamental steps of cell culture—counting live and dead cells.
The Countess™ automated technology reduces user error and speeds up cell
counting with less sample, less hassle, and a lower cost than any automated cell
counter on the market. Start counting today at www.invitrogen.com/countcells.

THE MOST

For research use only. Not intended for any animal or human therapeutic or diagnostic use, unless otherwise stated.
© 2009 Life Technologies Corporation. All rights reserved. The trademarks mentioned herein are the property of Life Technologies Corporation or their respective owners. These products may be covered by
one or more Limited Use Label Licenses (see Invitrogen catalog or www.invitrogen.com). By use of these products you accept the terms and conditions of all applicable Limited Use Label Licenses.
800 Profiling by Image Registration R. Tomer, A.S. Denes, K. Tessmar-Raible,
Reveals Common Origin of Annelid and D. Arendt
Mushroom Bodies and Vertebrate Pallium

RESOURCE

810 The Protein Composition of Mitotic S. Ohta, J.-C. Bukowski-Wills, L. Sanchez-Pulido,


Chromosomes Determined Using F. de Lima Alves, L. Wood, Z.A. Chen, M. Platani,
Multiclassifier Combinatorial Proteomics L. Fischer, D.F. Hudson, C.P. Ponting, T. Fukagawa,
W.C. Earnshaw, and J. Rappsilber

POSITIONS AVAILABLE

On the cover: Many epithelial tissues orient external structures such as cilia and hairs, and to
do so they use the planar cell polarity (PCP) proteins, which form polarized cortical domains
at epithelial cell junctions. The mechanisms that globally orient planar polarity have been
mysterious. In this issue, Aigouy et al. (pp. 773–786) show that the pattern of PCP domain
orientation in the Drosophila wing is a consequence of the oriented cell rearrangements and
divisions that shape this tissue.
R&D Systems Tools for Cell Biology Research™

Proteins
R&D Systems Quantikine® ELISAs
Antibodies

ELISAs
The Most Referenced Immunoassays
A direct measure of product quality is the frequency of citations in the scientific literature. R&D Systems has more
Assay Services
than 20 years of experience designing, testing, and optimizing the most cited ELISA kits in the world. Find out why
MultiAnalyte Profiling scientists trust R&D Systems ELISAs more than any other brand.

Activity Assays R&D Systems is the Most Referenced ELISA Manufacturer NEW Quantikine ELISA Kits
 α1-Acid Glycoprotein
Stem Cells  Angiopoietin-like 3
 Cathepsin V
ELISpot Kits  Clusterin
17.3%  Dkk-1
R&D Systems  EGF R/ErbB1
Flow Cytometry 41.8% 7.9%  EG-VEGF/PK1
 Fetuin A
Cell Selection 3.7%  FGF-21
 Galectin-3
2.5%  Gas 6
2.1%  GDF-15
1.6%  IL-17A/F Heterodimer
1.4%
 IL-19
 Lipocalin-2/NGAL
 MBL
 Proprotein Convertase 9/PCSK9
Approximately 42% of Referenced Immunoassays are Developed and Manufactured by  Periostin/OSF-2
R&D Systems. A survey of 860 manuscripts from 44 journals was conducted to compare the  Progranulin
number of citations specifying the use of R&D Systems ELISAs to the number citing ELISAs from  ST2/IL-1 R4
other commercial sources. A total of 433 ELISA citations referencing immunoassays from 66  Thrombomodulin/CD141
different vendors were identified in the survey.  Tie-1
 TIM-1/KIM-1

For more information visit our website at www.RnDSystems.com/go/ELISA

For research use only. Not for use in diagnostic procedures.

R&D Systems, Inc. www.RnDSystems.com

R&D Systems Europe, Ltd. www.RnDSystems.co.uk

R&D Systems China Co., Ltd. www.RnDSystemsChina.com.cn


Leading Edge

In This Issue

Fish Oil Turns the Tide on Insulin Resistance


PAGE 687
Inflammation mediated by macrophages promotes insulin resistance in obesity. Oh et al. now identify the G protein-coupled
receptor 120 (GPR120) on macrophages and fat cells as a receptor for omega-3 fatty acids (u-3 FAs). The authors show that
GPR120 activation by u-3 FAs inhibits inflammation pathways in macrophages and can reverse insulin resistance in mice.
These results provide a molecular basis for the anti-inflammatory effects of u-3 FAs and suggest that anti-inflammatory
treatments may ameliorate insulin resistance in obesity.

West Nile Virus Stopped at Its Source


PAGE 714
West Nile virus, a potentially deadly virus in humans, propagates in mosquitoes.
Cheng et al. now find that infection of West Nile virus triggers mosquito cells to
produce the lectin protein mosGCTL-1. This C-type lectin enhances entry of the
virus into additional mosquito cells through its interaction with a protein tyrosine
phosphatase receptor, which is homologous to human CD45. Blocking
mosGTCL-1 with an antibody disrupts the infective cycle of West Nile virus in
mosquitoes, suggesting a new strategy for controlling viral dissemination.

Antibodies Are Double-Trouble for Cancer


PAGE 699
Monoclonal antibodies are standard therapeutics for several cancers, including the
anti-CD20 antibody rituximab for B cell non-Hodgkin lymphoma (NHL). However,
antibodies are not curative and must be combined with cytotoxic chemotherapy
for clinical benefits. Now, Chao et al. identify CD47 as an antibody target in NHL
and demonstrate that combining anti-CD47 antibody with the rituximab antibody eradicates human NHL in mice. The synergistic
mechanism used by these two antibodies may be applicable to combined antibody treatments for many types of cancers.

ATAC Wears Two HATs in MAPK Signaling


PAGE 726
Extracellular cues often trigger MAP kinase (MAPK) signaling pathways, which
then activate downstream transcription factors like c-Jun. Here, Suganuma
et al. demonstrate that the ATAC histone acetyltransferase (HAT) governs the
response to MAPK signaling by serving as both a coactivator of transcription
and a suppressor of upstream signaling in the MAPK pathway. The authors
show that ATAC acetylates histone H4 at JNK target genes, which then serves
as a positive cofactor for basal transcription. In addition, ATAC directs upstream
MAPKs to the site of c-Jun binding and restricts the levels of JNK activation.

A Multitasking Leader mRNA


PAGE 737
Bacterial mRNAs often contain leader sequences that regulate transcription
of the adjacent coding region by binding metabolites and ions. For example,
the leader of the mRNA for the Salmonella Mg2+ transporter gene mgtA responds
to Mg2+. Now, Park et al. demonstrate that this leader also contains a short open reading frame with many proline codons, trans-
lation of which places mgtA expression under the control of cytoplasmic proline concentrations as well as Mg2+. Thus, leader
mRNAs can use distinct mechanisms to sense multiple intracellular signals.

Vivid Memories of Days Gone By


PAGE 762
Light responses and photoadaptation in the fungus Neurospora depend on the circadian transcription factor White Collar
Complex (WCC) and its negative regulator Vivid (VVD). Mazhan et al. now demonstrate how WCC and VVD cooperate to
discriminate light intensities over more than five orders of magnitude during the day. At night, previously synthesized VVD
serves as a molecular memory of the sun’s brightness during the preceding day and suppresses responses to light cues
of lower intensity, such as moonlight.

Cell 142, September 3, 2010 ª2010 Elsevier Inc. 649


Cancer Research
Detect and Characterize Genomic Variations
Use powerful Roche instruments and consumables for
454-Ultra Deep Sequencing
high-throughput analysis of copy number variation using
NimbleGen CGH microarrays. Rely on 454 Sequencing
Systems, with their long read lengths (400 to 500 bp). Or
combine both technologies using a Sequence Capture workflow.

 Detect chromosomal imbalances down to approximately


5 kb, commonly found in tumor progression, using high-
qPCR Analysis using the LightCycler® System
density NimbleGen CGH microarrays.
V617F Wild type  Identify all important genomic and transcriptomic
and LOH variants by ultra-deep sequencing of PCR products using
the 454 Sequencing System.
Mutation

Visit www.cancer-research.roche.com for application notes


and more detailed information about using Roche products in
cancer research.
Verification of molecular mutations. The JAK2 V617F mutation is characteristic
in hematopoietic malignancies. Ultra-deep 454 Sequencing detected the
mutation in just 1.16% of reads (top panel). Sensitive Carousel-Based LightCycler®
melting curve analysis confirmed the mutation in approximately
1% of cells (lower panel).

Data provided by A. Kohlmann, MLL Munich Leukemia Laboratory GmbH, Germany.


View the entire Application Note No. 7 at www.cancer-research.roche.com.

For life science research only. Not for use in diagnostic procedures.
454, 454 SEQUENCING, LIGHTCYCLER, and NIMBLEGEN are trademarks of Roche. Roche Diagnostics GmbH
Other brands and product names are trademarks of their respective holders.
Roche Applied Science
© 2010 Roche Diagnostics GmbH. All rights reserved. 82372 Penzberg, Germany
Cell Polarity Goes with the Flow
PAGE 773
Planar polarization of epithelial cells allows the uniform alignment of hairs, cilia, and other cellular structures with tissue shape.
Now, Aigouy et al. combine experimental and theoretical approaches to show that polarity patterns in the Drosophila wing arise
during growth. Specifically, cell polarity is reoriented from a radial to a proximal-distal axis when mechanical stresses during
growth cause the cells to rotate or ‘‘flow’’ with respect to each other. Linking planar polarity to morphogenesis provides a simple
mechanism for coordinating the global polarity pattern with tissue shape.

Com-plexin’ with Semaphorins


PAGE 749
Semaphorins and their receptors, Plexins, are widely expressed protein families
that mediate repulsive signaling during cell guidance. Here, Liu et al. present two
X-ray crystal structures of PlexinC1, one in complex with the Semaphorin
Sema7A and another in complex with the Semaphorin mimetic A39R from the
smallpox virus. In both structures, the Semaphorin interacts with a PlexinC1
dimer in a novel edge-on, orthogonal geometry. These findings suggest that Plex-
ins are activated by ligand-induced dimerization during cell guidance.

Interspecies Organogenesis
PAGE 787
A goal of regenerative medicine is to derive organs from a patient’s pluripotent
stem cells (PSCs), but in vitro organogenesis is complex. Here, Kobayashi et al.
generate a functioning rat pancreas in mice without a pancreas by injecting rat
PSCs into mouse blastocysts. These interspecific chimeras provide proof of
principle for in vivo generation of organs derived from donor PSCs and for inter-
specific blastocyst complementation.

A Wormhole to the Origin of the Cortex


PAGE 800
The cerebral cortex or pallium controls the highest-order processing in mammals, but its evolution remains enigmatic. Now,
Tomer et al. develop an expression profiling technique to generate a gene expression map for the developing brain of the
segmented worm Platynereis dumerilii. Comparison of this map with that observed for the developing cerebral cortex
suggests a common evolutionary origin for the mammalian cortex and the worm’s mushroom body, a brain region in inver-
tebrates that processes sensory input.

Will the Real Chromosomal Proteins Please


Stand Up
PAGE 810
Proteomic analysis of large cellular structures is frequently hindered by the
presence of contaminants. In their analysis of mitotic chromosomes, Ohta
et al. overcome this problem by integrating proteomics with additional quanti-
tative and bioinformatic data—effectively adding a final purification step in silico.
This approach successfully pinpoints hitchhikers from amidst the 4,000 iden-
tified proteins and provides insight into the functional relationships among the
genuine constituents, including evidence that many more kinetochore-associ-
ated proteins exist than recognized previously.

Cell 142, September 3, 2010 ª2010 Elsevier Inc. 651


Next-gen GWAS.
NOW.
It’s a content revolution.
Up to 50% more coverage of common and rare
variants than all other arrays. Maximum power for
any population.

The Omni family of microarrays can propel


your studies into true next-gen GWAS. With
a clearly defined product path to the future.
Immediate utility. Future flexibility.

Get on the path to next-gen GWAS.

Now is the time. Get started at


www.illumina.com/GWAS
Leading Edge

Molecular Biology Select

The central importance of the tumor suppressor Retinoblastoma protein (Rb) in cell-cycle progression makes
its regulation a focal point for diverse biological processes, as evidenced by recent work described in this
issue’s Molecular Biology Select. These findings reveal new insight into Rb’s involvement in tissue regener-
ation and differentiation, as well as previously unrecognized mechanisms of Rb regulation.

Short-Term Inactivation, Lasting Benefits


Tissue regeneration in mammals is greatly restricted, whereas
many other types of vertebrates display astonishing feats of tissue
replacement, including regrowth of major structures such as
limbs. According to new findings by Pajcini et al. (2010), mamma-
lian innovations in the realm of tumor suppression are one factor
that is likely to contribute to this pronounced deficiency. The
authors show that combined inactivation of the tumor suppressors
retinoblastoma (Rb) and ARF reverses the differentiation of mouse
muscle cells, turning postmitotic cells into proliferating myoblasts,
and when these induced myoblasts are transplanted into donor
mice they successfully fuse into existing myofibers. Prior work in
newts has shown that Rb phosphorylation, which inactivates the
Dedifferentiated myocytes redifferentiate and fuse protein, promotes the reentry of myotubes into the cell cycle, a crit-
into existing muscles in vivo (visualized with green
ical initiating event in limb regeneration. The inspiration to addition-
fluorescent protein).
ally inactivate ARF was motivated by available evidence suggesting
its potential exclusivity to mammals and birds, and thus the authors
reasoned that ARF might be particularly important in mediating differences in regeneration potential between
vertebrate species. Could a similar strategy of Rb and ARF inactivation be used to promote cell-cycle entry of
endogenous cells for regeneration therapy? Although this remains to be seen, the observation that only tran-
sient inactivation of these tumor suppressors is needed for successful creation of regenerative cells may go
some way toward allaying concerns that such an approach would invariably promote cancer. Future efforts
are also likely to address whether this intervention triggers cell-cycle reentry for a range of mammalian cell
types.
K.V. Pajcini et al. (2010). Cell Stem Cell 7, 198–213.

Fat Chance for Bone Formation


Previous in vitro studies suggested that Rb plays a critical role in the
decision of meschenchymal cells to become adipocyte or bone
cells, but in vivo evidence for this hypothesis has been lacking.
Calo et al. (2010) now show that Rb gives meschenchymal cells
the extra nudge they need to commit to becoming bone-forming
osteoblasts. Without Rb, these cells are more likely to differentiate
into brown fat cells, leading to reduced levels of calcified bones
and increased levels of brown adipose tissue in mice. To sort out Deletion of Rb in the embryo proper using Meox2-
how Rb regulates the fate of the meschenchymal cells during devel- Cre reduces the level of calcified bone as detected
opment, the authors engineered mice with the RB1 gene and/or by Alizarin Red staining. Image courtesy of J. Lees.
the p53 gene deleted only in uncommitted meschenchymal cells.
As expected, animals missing the tumor suppressor p53 develop multiple types of tumors, including osteosar-
comas. Combining the p53 mutation with a deletion of one RB1 allele increases the frequency of osteosarcomas,
whereas deletion of both RB1 alleles shifts the tumor distribution away from osteosarcomas towards brown fat
tumors. Thus, Rb regulates the fate of meschenchymal cells in a dose-dependent manner. These results are
surprising given that the majority of human osteosarcomas contain RB1 mutations. The authors speculate
that osteosarcoma tumors most likely arise from cells already committed to becoming osteoblasts, and muta-
tions in RB1 promote dedifferentiation of these cells and thus tumorigenesis.
E. Calo et al. (2010). Nature. Published online August 4, 2010. 10.1038/nature09264.

Cell 142, September 3, 2010 ª2010 Elsevier Inc. 653


.%7

Reach Your Ideal Candidate!


så0OSTåONEåORåMULTIPLEåå
JOBåOPENINGSå

så0ROMOTEåANåUPCOMINGåå
CONFERENCEåORåEVENTå

så!NNOUNCEåAåGRANTAWARDå

så%LEVATEåYOURåå
ORGANIZATIONSåPROlLE

careers.cell.com
Rb Gets Caught in a Custody Battle
Rb is caught in a tug-of-war between cyclin-dependent kinases
(Cdks) that inactivate it by phosphorylation to permit cell-cycle
progression and phosphatases that remove the modification
to promote cell-cycle arrest. New findings of Hirschi et al. (2010)
demonstrate that this conflict is waged over the same binding
interface of Rb. They provide structural evidence that the phospha-
tase PP1 interacts with Rb in a region previously shown critical for
the interaction of Rb with Cdks. They further show that PP1 can
suppress the activity of Cdk2-cyclin A toward Rb to block cell-
cycle progression in a human osteosarcoma cell line, and that
the complex of PP1 and Rb appears to be stable, or at least
more prevalent, at mitotic exit. Among the interesting questions
this work raises is, what factors determine the outcome of PP1
and Cdk competition? The authors propose that concentration
and subcellular localization of the competing proteins likely play
a role, but it remains unclear how this molecular dispute is settled
X-ray crystal structure of Rb (magenta) in complex under specific biological circumstances, for example after DNA
with the PP1 catalytic domain (gray). Image cour- damage.
tesy of S. Rubin.
A. Hirschi et al. (2010). Nat. Struct. Mol. Bio. Published online August
8, 2010. 10.1038/nsmb.1868.

Methylation Moves to the Front of the Line


Although first reported more than 30 years ago, methylation
of protein N termini (a-N-methylation) remains a poorly understood
protein modification. Thus, the recent identification by Schaner-Too-
ley et al. (2010) of an enzyme that catalyzes the reaction promises to
accelerate understanding of the modification’s functions. The
authors use methylation of a known target of a-N-methylation called
RCC1 (a Ran guanine nucleotide exchange factor) as an indicator of
the presence or absence of the enzymatic activity from fractionated
HeLa cell nuclear extracts. Fractions with methyltransferase activity
were subjected to mass spectrometry, leading to the identification of
the methyltransferase responsible, which the authors name NMRT
(N-terminal RCC1 methyltransferase). The NMRT crystal structure
facilitated the modeling of substrate recognition, and further analysis
defining the consensus sequence for target recognition suggested
Rb as a potential substrate. Subsequent assays provide evidence NMRT structure with RCC peptide modeled in the
active site. Image courtesy of I. Macara.
that Rb is modified by a-N-methylation, at least in some cell types.
a-N-methylation of RCC1 promotes stable association with chro-
matin, and loss of NMRT or the absence of the RCC1 methylation results in defects in mitosis. In contrast, the
purpose of Rb a-N-methylation remains a tantalizing mystery. Regardless of whether Rb a-N-methylation is rele-
vant to its roles in cell-cycle control, the discovery of NMRT opens a door through which others will likely follow.
Are there other a-N-methyltransferases? And if so, is their substrate specificity similar to NMRT? The answer may
be an indicator of whether this modification is an exotic posttranslational event or might instead be considerably
more common than previously appreciated. Another compelling question is whether there are a-N-demethylases
that reverse the modification.
C.E. Schaner-Tooley et al. (2010). Nature. Published online July 29, 2010. 10.1038/nature09343.

Robert P. Kruger

Cell 142, September 3, 2010 ª2010 Elsevier Inc. 655


,OOKING¬FOR¬WHAT¬#ELL¬
HAS¬PUBLISHED¬IN¬¬
)MMUNOLOGY¬LATELY


PHOIDå#ELLS å å åPPån

ONåINå,YM Då-OTIF
Så!CTIVATI L å  å*AN OSINE "ASE
CTERIZEå2A SE å* 0 å# EL IC å4 YR
Så#HARA OO LA SM
å(YSTERESI KRABORTYå!
+ å2 Nå#YTOP n
NALINGåAND EISSå! å#HA å#$EPSILO  åPPå
$IGITALå3IG OV ER Nå # å9ANGå- å7 N E å" IN D INGåOFåTHE L å   å. OVå å
ANå* å' ICå-EMB RA å# EL
- å:IKHERM åBYå$YNAM RPFENNIGå+7 LOPMENT
$ASå* å(Oå !CTIVATION ** å7UCHE Cå#ELLå$EVE HUANGå9 å2
EIZISå
Rå å#6 å#HOUå RI TI
Få4 C E LLå2 E C E P TO
IE TE RS å# $ å# AR M AN
A C YT O ID å$ E N D
OL TE Rå7 å2AUCHå! å:
2EGULATIO
NåO LLå*2 å3CH W RåOFå0LASM 3 å+ANTå3'
å(
NONå% å#AL
Lå-% å3CHNE Cå2EGULATO å(OLLANDERå.
8Uå# å'AG E N TI A LåA NDå3PECIl RG å$ å: WEIERå# åDEN
åISåANå%SS LEYå2 å(OLM
BE ISEASE
RI P TI O N å& ACTORå%  å4 å3 CH EUå3 å,OCKS A M M A TO RYå"OWELå$
4RANSC AE DA UMANå)N m
EHNERå- å- å2ISKåFORå( BERGå23åå
ATONå-, å, 
RSå'ENETIC ERå,( å"LUM
#ISSEå" å# å å   åPPån N D å# O N FE å3 CH RE IB ER å3 å'LIMCH
"å#ELL å
å/CT ATIONåA å(IGGINSå$
%
ALå)NmAMM ENHUISå%%
2 å3 TR E SS åTOå)NTESTIN SS IG å3 å4 ILGå( å.IEUW
Så% KMANå*. å:
EI
8"0å,INK NKEå! å'LIC
EEå!( å&RA 
+ASERå! å, åPPån
EPå å MUNITY n
#EL L å   å3 åOFå!UTOIM  åPPå EOSTASIS
)N TR IN SI C å)NITIATION   å! UGå å MUNEå(OM
VE N TS å# E LL Vå 2 å# EL L å
IN TA IN Så )M
4REXå0RE EIDMANNå4
å-EDZH ITO THATå-A å#HENå9(åå
å+Oå*3 å( å)MMUNITYå ( å9ANGå8
3TETSONå$" N A TE åA N Då!DAPTIVE å(ILLIARDå" å(Uå3 å3HENå
Få)N Gå, å8Uå, RACTIONS
EGULATORåO å3UNå* å+ON OMICå)NTE
EGATIVEå2 ILLIARDå! å,Iå, ANGEå'EN
4)0% åAå. OD Yå 2 * å( Rå , O N G 2
Gå3 å#ARM TIONSåFO REå#åå
3UNå( å'ON åPPån
 Så)MPLICA CHå4! å-UR
  å- AYå å å( E A VY # HAINå,OCU å' RO SV ELDå&' å+NO
L å GLOBULIN **
#EL
Eå)MMUNO ANå$ONGENå
å2IBLETå2 åV JURY
U CTUREåOFåTH å#UTCHINå3 TEå,UNGå)N
4H E å $ å3 TR
å: EL M å- # å0 EA Kå - -
E Yå 0 A TH W AYåOFå!CU Gå ( å, IUå( å3UNå9
åå
ALAå3 åVAN n GåASåAå+ NGå9( å7AN NINGERå*-åå
*HUNJHUNW å PP å  å3 IG N A LIN ZE Nå 2 å, EU Rå# * å0 EN
å!PRå å
 ECEPTORå VAå- å6ELDH
UI
IANGå# å"IN
DE
#ELL å å4OLL ,IKEå2 å' å%RMOLAE ICHOLLSå* å*
D A TI VE å3 TRESSåAND ER KM AN Nå 4 åVANå,OO ST å- å( OL MDAHLå2 å.
TIONåOFå/XI Iå2 å0 VI
AN -ALHAM KIRAå3 å(ULTQ
)DENTIlCA ' å9AGHUBI EIRISå*3 å3LU
TSKYå!3 å!
å9 å+ UB Aå + å.EELYå' AV AR Iå3 å0
)MAI å-ECHå# å"
- å+OPFå-
0ASPARAKISå å   åPPå
n
 å! PR å 
#EL L å 

,OOKå!GAIN
$ISCOVERå-ORE
WWWCELLCOM
“ TRUSTED PARTNERS
LIKE MILLIPORE ARE
RARE BUT CENTRAL
TO OUR SUCCESS.”

Dr. Michael West


CEO, BioTime, Inc.
Renowned thought leader in
stem cell therapeutics

Millipore is the source for sterile filtration you trust.


Stericup® and Steritop® sterile filters combine the superior flow rates and low non-
specific binding you need to protect everything you’ve poured into your cell culture work.
With over 50 years of filtration expertise, Millipore continues to set the industry standard
for high-performance membrane technology and its application in sterile filtration.

Find out
Go to themore atat
source
www.millipore.com/trustStericup1
www.millipore.com/amicon

Millipore, Advancing Life Science Together, Stericup and Steritop are registered trademarks of Millipore Corporation.
The M mark is a trademark of Millipore Corporation. ©2010 Millipore Corporation. All rights reserved. LS-SBU-10-03205
Stay Connected
Join ASHG Today!
Benefits of Membership:
• Subscription to AJHG, now published
by Cell Press
• Reduced registration fees for the
Annual Meeting
• Discounted subscriptions to Cell
Press journals
• Access to educational materials
• Influence on public policy process

www.ashg.org
MAGPIX®—THE NEW WAVE IN MULTIPLEXING
The magnetic-bead, low-cost platform that attracts multiplexing to
every lab. There’s never been a better time to jump in.
Accelerate your research at a price you can afford, using the xMAP® technology you trust.
The new MAGPIX instrument easily detects up to 50 analytes simultaneously in a small sample.
To complete this flexible solution, choose from Millipore’s wide selection of magnetic bead
kits encompassing metabolic disease, inflammation, toxicity, neuroscience and cell signaling.

ADVANCING LIFE SCIENCE TOGETHER® Catch the wave at


Research. Development. Production. www.millipore.com/MAGPIX2

Millipore and Advancing Life Science Together are registered trademarks of Millipore Corporation. The M Mark
is a trademark of Millipore Corporation. MAGPIX and xMAP are registered trademarks of Luminex Corporation.
©2010 Millipore Corporation. All rights reserved.
¬,OOK¬!GAIN

$ISCOVER¬-ORE
s¬!CCESS¬TO¬THE¬¬#ELL¬0RESS¬PRIMARY¬RESEARCH¬JOURNALS¬AND¬
¬4RENDS¬REVIEWS¬TITLES ¬ALL¬ON¬THE¬SAME¬PLATFORM
s¬)MPROVED ¬MORE¬ROBUST¬ARTICLE¬AND¬AUTHOR¬SEARCH
s¬6IDEO ¬ANIMATIONS¬AND¬SOUND¬lLES
s¬%ASY¬TO¬NAVIGATE¬HOME¬PAGE ¬ARTICLES¬PAGES¬AND¬ARCHIVE

WWWCELLCOM
Leading Edge
Essay

Phosphotyrosine Signaling: Evolving a New


Cellular Communication System
Wendell A. Lim1,2,* and Tony Pawson3,4,*
1Howard Hughes Medical Institute
2Department of Cellular and Molecular Pharmacology
University of California, San Francisco, San Francisco, CA 94158, USA
3Samuel Lunenfeld Research Institute, Mount Sinai Hospital, Toronto, Ontario M5G 1X5, Canada
4Department of Molecular Genetics, University of Toronto, Toronto, Ontario M5S 1A8, Canada

*Correspondence: lim@cmp.ucsf.edu (W.A.L.), pawson@lunenfeld.ca (T.P.)


DOI 10.1016/j.cell.2010.08.023

Tyrosine phosphorylation controls many cellular functions. Yet the three-part toolkit that regulates
phosphotyrosine signaling—tyrosine kinases, phosphotyrosine phosphatases, and Src Homology
2 (SH2) domains—is a relatively new innovation. Genomic analyses reveal how this revolutionary
signaling system may have originated and why it rapidly became critical to metazoans.

Throughout human history, new technolo- (TyrK) phosphorylate specific target with an evolutionary process? Proteins
gies and technological platforms have tyrosine residues, phosphotyrosine phos- that bind or remove a posttranslational
constantly been invented. Only a small phatases (PTP) remove the phosphates, modification would seem useless without
fraction of these technologies go on to and Src Homology 2 (SH2) domains an enzyme to generate the modification
be widely adopted, but these can recognize the modifications (Pawson and, in principle, would not provide
ultimately have transformational conse- 1995). Together, these three modules a fitness advantage leading to its retention
quences. In the evolutionary history of form the ‘‘writer,’’ ‘‘eraser,’’ and ‘‘reader’’ and spread. The pTyr signaling platform
living organisms, we know that innovative toolkit that is common to many diverse provides a case study to look for plausible
molecular systems have appeared at key cellular information processing platforms stepwise pathways of the evolution of
points in time, and these are thought to (Figure 1A). A rich array of diverse and a multipart system.
have played a transformative role in major complex regulatory schemes can be Here, we reconstruct a possible history
evolutionary transitions in the tree of life. achieved through the dynamic interplay for the evolution of pTyr signaling. This
But how do such innovative molecular of these three modular functions (Pawson reconstruction is based on the recent
systems emerge, and how and why do et al., 1993; Pawson, 1995; Bhattachar- sequencing of the genomes of a number
some proliferate and become stably yya et al., 2006; Kholodenko, 2006). of organisms that originated both before
adopted by subsequent lineages? A combination of these modules can and after the emergence of metazoans
An example of such an innovative lead to higher-order functions (Figure 1B). from single-celled eukaryotic ancestors
molecular system is phosphotyrosine For example, there are several proteins (King et al., 2008). The genome sequence
(pTyr)-based signal transduction. This containing a combination of SH2 and of the choanoflagellate, Monosiga brevi-
molecular system for transmitting cellular kinase domains that can generate collis, has been particularly illuminating
regulatory information is estimated to positive feedback (phosphorylation of as choanoflagellates are thought to be
have appeared relatively recently in the tyrosine sites leads to SH2-mediated one of the closest single-celled relatives
history of life—600 million years ago, recruitment of the kinase, and subse- of metazoans. We present a model for
just prior to the emergence of multicellular quently, more extensive phosphorylation) how this three-part signaling system
animals (King et al., 2003; Pincus et al., (Pawson, 2004). Similarly SH2-phospha- could have plausibly evolved in a stepwise
2008; Manning et al., 2008). The pTyr sig- tase domain combinations can generate manner. We propose that once the
naling system has become an essential negative feedback (Tonks and Neel, complete three-part system was in place,
part of metazoan biology. For example, 2001). it may have rapidly taken hold in subse-
pTyr signaling plays a central role in many The three-part pTyr signaling toolkit quent lineages because it could generate
cell-to-cell communication pathways, thus raises a classic question in evolu- new regulatory behaviors without signifi-
including those that regulate proliferation, tionary biology: how do complex, interde- cant cross-interference with existing
differentiation, adhesion, hormone res- pendent systems arise? It is clear why regulatory circuits. We also discuss the
ponses, and immune defense (Hunter, a new system encompassing a writer, possible role of this new communication
2009). eraser, and reader might be extremely system in facilitating the transformative
In modern metazoans, pTyr signaling is useful. But, given their interdependence, evolutionary shift to multicellularity.
mediated by a toolkit of three distinct how could these individual components Given the incomplete record, however,
functional modules: tyrosine kinases arise in a stepwise fashion consistent such an evolutionary reconstruction is

Cell 142, September 3, 2010 ª2010 Elsevier Inc. 661


by an upstream Ser/Thr kinase on both
a Thr and Tyr residue within its activation
loop; Cdk1 is phosphorylated on Tyr14
by the inhibitory kinase Wee1). These
tyrosine modifications are clearly not
recognized by SH2 domains, but they
exert direct allosteric effects within the
proteins in which they occur. Thus, PTP
domains may have provided a fitness
benefit by negatively modulating these
rare but functionally important phosphor-
ylation events. Consistent with this model,
the proteins PTP2 and PTP3 in yeast
clearly have a functionally important role
in downregulating MAPK-mediated sig-
naling in response to pheromones or
osmolarity changes, explaining their
Figure 1. The Writer, Reader, Eraser pTyr Toolkit fitness benefit (Pincus et al., 2008). In
(A) In pTyr signaling, the tyrosine kinase (TyrK), Src Homology 2 (SH2), and phosphotyrosine phosphatase addition, PTPs may have played a general
(PTP) domains form a highly interdependent signaling platform. This platform serves as the writer, reader, role buffering against the occasional
and eraser modules, respectively, for processing pTyr marks.
(B) Components of pTyr signaling can be used to build complex circuits. For example, recruitment of an harmful stray phosphorylation of function-
SH2-TyrK protein to an initiating pTyr site can lead to amplification of tyrosine phosphorylation through ally important tyrosines.
a positive feedback loop. Where did these PTPs come from?
PTPs are likely to have arisen from
highly speculative. For example, we have tyrosine phosphatase activity. We a common ancestor of the related dual-
cannot rule out more complex paths refer to the single putative ‘‘SH2’’ domain specificity phosphatases, which are also
involving cycles of evolutionary gain and in yeast, found within the gene Spt6, as found in most single-celled eukaryotes
loss of components, nor the possibility a proto-SH2 domain because it does not (Kennelly, 2001; Alonso et al., 2004).
that similar components in distinct line- show pTyr binding (the domain has been Dual-specificity phosphatases are cata-
ages have independent origins. Nonethe- reported to show phospho-Ser/Thr lytic domains that can dephosphorylate
less, this model may provide a useful binding; Dengl et al., 2009). Thus, func- both pSer/Thr and pTyr substrates. The
framework for focusing studies of pTyr tionally, it cannot be considered a PTP and dual-specificity phosphatase
signaling origins and the origins of analo- ‘‘reader’’ domain that is part of a pTyr catalytic domains are distinct but are
gous multicomponent signaling plat- regulatory system. These observations evolutionarily related. They share a
forms. suggest a simple model: the first step in common fold and the core catalytic motif
We describe three possible stages in the evolution of the three-part pTyr HC(X)5R, in which a phosphocysteine
the emergence of the modern pTyr signaling machinery was likely to have enzyme intermediate is generated during
signaling toolkit, each represented by an been the emergence of a functional tyro- catalysis. (Sometimes, both dual-speci-
extant model organism (Figure 2). These sine phosphatase. But why would PTPs ficity phosphatases and classical PTPs
stages are representative; we do not arise in the pre-tyrosine kinase world? are referred to as PTPs; here, we use
claim to define the exact path of evolution, What functional use and fitness advan- this nomenclature only for the classical
but rather focus on identifying the domi- tage would this eraser domain provide in PTP domains that act only on pTyr). The
nant classes of stable intermediates that organisms lacking a writer domain? domains of dual-specificity phosphatases
can exist in the broader evolutionary land- The answer may lie in the fact that some have a shallower active site than classical
scape. Ser/Thr kinase domains, which are more PTPs, which may explain why they can
ancient than tyrosine kinases (dating dephosphorylate either Tyr or Thr/Ser
PTPs in a Pre-Tyrosine back close to the origins of eukaryotes), residues. In some lineages, dual-speci-
Kinase World can carry out sporadic but functionally ficity phosphatases have functionally
What came first, TyrK, PTP, or SH2 important phosphorylation of tyrosines. diverged further, giving rise to members
domains? Sequence analysis suggests Phosphoamino acid analysis of yeast that can act on lipid substrates, such as
that it was PTP domains. The genome of reveals a small but significant population the phosphoinositide phosphatases
a simple single-celled eukaryote like the of pTyr (Schieven et al., 1986). Moreover, PTEN and the myotubularins (Alonso
budding yeast Saccharomyces cerevisiae certain events, such as the activation et al., 2004). Thus, the PTPs appear to
shows no TyrK proteins and one proto- of mitogen-activated protein kinases have arisen from a somewhat promis-
SH2 domain, but a handful of PTP (MAPKs) and inhibition of the cell-cycle cuous class of multifunctional phospha-
proteins (Pincus et al., 2008) (Figure 2, kinase Cdk1, are known to involve phos- tases.
Stage 1). Most fungi have no more than phorylation of tyrosine residues (for acti- Despite the presence of PTP proteins in
five PTP proteins, and several of these vation, a MAPK must be phosphorylated fungi, there are striking differences

662 Cell 142, September 3, 2010 ª2010 Elsevier Inc.


Figure 2. Evolution of pTyr Signaling
Shown is a possible path for the emergence of phosphotyrosine (pTyr) signaling. We postulate three successive stages, each represented by what is observed in
a modern organism. The thickness of the tree reflects the approximate degree of usage of pTyr signaling (thicker lines mean more usage). Stage 1 (exemplified by
the budding yeast Saccharomyces cerevisiae) reflects the situation in early eukaryotes, in which PTPs emerged but were limited in number and complexity. They
were most likely used to reverse or process sporadic cross-phosphorylation of tyrosine residues by Ser/Thr kinases. S. cerevisiae has fewer than five PTP
proteins and no functional SH2 or TyrK domains. Stage 2 reflects systems in which functional SH2 domains emerged that were able to bind to pTyr motifs.
Together with Ser/Thr kinases with increased cross-reactivity for Tyr (such as tyrosine kinase-like or dual specificity Ser/Thr kinases), these systems may reflect
the most primitive of pTyr writer/reader/eraser systems. However, the lack of a dedicated Tyr kinase may have limited the utility and expansion of this toolkit. This
stage is potentially represented by the slime mold, Dictyostelium discoideum. Stage 3 reflects systems that evolved after the emergence of the modern TyrK
domain. We postulate that the full writer/reader/eraser system was of so much greater utility that its use expanded dramatically. This likely resulted in many
more proteins in these families, as well as much more complex, multidomain architectures than those seen in the earlier stages. This stage is represented by
both the multicellular metazoan and unicellular choanoflagellate lineages.

between these proteins and those proteins in which the PTP module has transfer enzymes; Bordo and Bork,
found in metazoans (Figure 2). For been functionally recombined with multi- 2002). Thus, fungal PTP proteins are
example, there are far fewer PTPs in ple other signaling modules. In contrast, very simple (one to two domains) and
fungi (5/genome versus 40/genome in fungi, the PTP domains are all either lack the combinatorial complexity of
in metazoans) and they are considerably in simple single-domain proteins or in metazoan PTP proteins. The simplicity
less complex in domain architecture combination with a single rhodanase-like and low number of PTPs in yeast sug-
(Pincus et al., 2008). Metazoan PTP domain (a putative regulatory domain gests that in early single-celled eukary-
proteins tend to be large multidomain that is homologous to a class of sulfur otes, PTP domains had fairly limited

Cell 142, September 3, 2010 ª2010 Elsevier Inc. 663


functional utility, especially when com- pTyr-binding SH2 domains and may kinase domain followed by an SH2
pared to their broad and complex usage therefore provide a living representative domain, a domain combination that is
in metazoans. of this second evolutionary stage (Fig- somewhat similar to metazoan cyto-
Unlike PTPs, there are no known pTyr- ure 2, Stage 2). Dictyostelium is a soil- plasmic tyrosine kinases like Src (Monia-
binding SH2 domains in fungi, although living amoeba that has a unicellular kis et al., 2001). The Shk catalytic domain,
there is one clearly homologous domain lifestyle in the presence of bacterial however, lacks motifs characteristic of
found in the yeast protein SPT6. This food. However, when food is depleted, bona fide tyrosine kinases and biochemi-
protein, which has a domain with an individual cells aggregate in response to cally displays dual specificity toward
SH2-like sequence and fold, is involved the chemoattractant cAMP to form serine/threonine and tyrosine residues.
in the regulation of transcription elonga- a multicellular structure, which then Indeed, Dictyostelium differs from
tion, and the SH2 domain binds to the develops into a fruiting body through the metazoans and choanoflagellates in that
Ser/Thr phosphorylated C-terminal tail of differentiation of stalk and spore cells. its genome does not encode any modern
RNA polymerase II. The domain does The rudimentary pTyr-SH2 system in tyrosine-specific protein kinases. For
not bind to pTyr (Dengl et al., 2009). Inter- Dictyostelium is important for aspects of example, metazoan STAT proteins are
estingly, a single SPT6 ortholog, with the this differentiation process, including usually phosphorylated by Janus tyrosine
same overall domain architecture, is intracellular responses to both cAMP kinases (JAKs), but there are no JAKs in
found in all eukaryotes, including all fungi and the morphogen differentiation in- Dictyostelium (Kay, 1997). This suggests
and metazoans (but not prokaryotes). This ducing factor or DIF (which induces the the possibility that signaling proteins con-
finding suggests that in early eukaryotes, differentiation of prestalk cells), as well taining SH2 domains such as STATs
a proto-SH2 domain emerged to perform as for transcriptional regulation in res- evolved before the modern tyrosine
a highly specialized function—one that ponse to hyperosmotic stress. These kinases with which they are associated
was unrelated to the flexible modular observations are consistent with early in metazoans. The identity of the kinase
pTyr recognition function of the modern pTyr-SH2 signaling playing a role in responsible for STAT tyrosine phosphory-
SH2 domain. This proto-SH2 domain cellular responses to changing environ- lation, and the consequent formation of
most likely did not ‘‘read’’ pTyr modifica- mental conditions. SH2-binding sites, remains mysterious.
tions, but instead recognized a special- The Dictyostelium genome specifies 13 How, then, is tyrosine phosphorylation
ized related modification. Thus, although proteins with SH2 domains (as well as of Dictyostelium proteins such as the
SPT6 is likely to represent an early a single Spt6 homolog). These 13 proteins STATs controlled? Thus far, genetic anal-
ancestor or relative that eventually gave cluster into five basic domain architec- ysis has not identified a specific relevant
rise to modern SH2 domains, it cannot tures, two of which are homologous to kinase, and it has been proposed that, in
be considered a functional part of a pTyr metazoan SH2 proteins. Notably, Dic- contrast to mammalian STATs, there
regulatory toolkit. We therefore postulate tyostelium has four STAT (signal trans- may be basal constitutive phosphoryla-
that early eukaryotes had only a pTyr ducers and activators of transcription) tion of Dictyostelium STAT tyrosine sites,
eraser function (mediated by PTPs) with proteins that are very similar to metazoan which is regulated by changes in PTP
no specialized complementary reader or STAT transcription factors (Kay, 1997; activity in response to extracellular
writer functions. Kawata et al., 1997). For example, they signals (Langenick et al., 2008). One of
In summary, the PTP domain and all have an SH2 domain juxtaposed to the PTPs in Dictyostelium, PTP3, binds
a structural ancestor of the SH2 domain a DNA-binding region; they are inducibly and dephosphorylates STATc, thereby
appear to have arisen in early single- phosphorylated on tyrosine residues in blocking SH2-mediated dimerization and
celled eukaryotes, but are likely to have response to stress or the extracellular STATc accumulation in the nucleus.
functional origins that are not directly signaling molecule DIF; they undergo Signaling induced by the DIF morphogen
related to their later function in modern pTyr/SH2-mediated dimerization and appears to transmit signals by inhibiting
pTyr regulatory systems. These compo- then translocate to the nucleus to regulate PTP3 activity and consequently boosting
nents may have provided a limited but the expression of specific genes. Dictyos- STATc tyrosine phosphorylation and
incremental fitness advantage, even in telium also has an ortholog of the STATc-dependent gene expression.
the absence of a specialized tyrosine mammalian E3 ubiquitin ligase Cbl, which Although Dictyostelium lacks true tyro-
kinase domain. uses SH2 and Ring domains to couple sine kinases, it is noteworthy that its
pTyr signals to the ubiquitination ma- genome has a significant expansion in
Toward a Write/Read/Erase System chinery (Langenick et al., 2008). The re- the number of putative dual-specificity
In the early days of a more sophisticated maining three domain architectures of protein kinases (there are 70, also known
pTyr-signaling system, we suggest that Dictyostelium SH2 proteins are distinct as tyrosine kinase-like or TKL kinases)
a proto-SH2 domain (mostly likely a from those found in other sequenced (Manning et al., 2008). This set includes
homolog of the yeast Spt6 protein) in organisms. The LrrB protein has an SH2 the Shk catalytic domain, described
a single-celled organism acquired the domain linked to a leucine-rich repeat above. It is unlikely that any of these
new and functionally beneficial ability to domain (Sugden et al., 2010), whereas kinases are precursors of modern tyrosine
bind to pTyr-containing peptide motifs. the FbxB protein has an F-box followed kinases. However, it is plausible that these
The slime mold Dictyostelium discoideum by an SH2 domain and ankyrin repeats. represent the first evolutionary form of
has the simplest repertoire of bona fide In addition, the Shk proteins have a protein the ‘‘writer’’ function in a prototype pTyr

664 Cell 142, September 3, 2010 ª2010 Elsevier Inc.


three-part regulatory system. The union of sine kinases (Lee and Jia, 2009). It is functions, thus leading to the subsequent
an SH2 domain and a dual specificity therefore probable that BY kinases expansion of the complete set. Although
kinase domain, as found in the Shk evolved separately from metazoan tyro- the PTP and SH2 domains had utility in
proteins, may be an early example of link- sine kinases and operate in a different simpler organisms, their much larger
ing ‘‘reader’’ and ‘‘writer’’ modules to fashion. functional potential was not unleashed
achieve more complex functions such as The new eukaryotic tyrosine kinase until the emergence of the TyrK domain.
positive feedback. Nonetheless, the domain appears to have been a game The rapid expansion of the pTyr
limited functionality of the dual specificity changing innovation (Figure 2, Stage 3). signaling machinery in the ancestors of
kinases in carrying out tyrosine phosphor- The total number of tyrosine kinase choanoflagellates and animals is reminis-
ylation may have limited the capabilities of proteins in both choanoflagellate and cent of how technology expands in
this early system. This may explain the metazoan species is in the range of 30– quantum jumps, especially in situations
very modest expansion of pTyr signaling 150 per genome (Pincus et al., 2008; involving codependent technologies. For
in organisms such as Dictyostelium. Manning et al., 2008). Among sequenced example, the value of the laser expanded
These observations paint the following genomes, there is a striking absence of dramatically after the later invention of
picture of Dictyostelium pTyr signaling species with only a small number of TyrK the complementary technology of fiber
and, by extension, of an early phase in proteins. This all-or-none sudden jump in optics. This codependent technology al-
the evolution of pTyr communication. the number of TyrK proteins suggests lowed lasers to be repurposed to rapidly
SH2 domains have acquired pTyr-binding their importance as they appear to have displace electrical transmission via
activity and are found in several distinct undergone rapid expansion and subse- copper wires as the backbone of global
combinations with other types of signaling quent retention. communication (Alwayn, 2004). Thus,
domains. Among these, the STAT and Cbl What is perhaps more striking is the although lasers had standalone utility,
proteins are shared with metazoans, observation that the emergence of the their major application had to await the
whereas the LrrB, FbxB, and Shk proteins TyrK domain and its rapid expansion introduction of complementary tech-
are unique to Dictyostelium. But no dedi- correlates with an equally rapid expansion nology. The expansion of molecular com-
cated modern tyrosine kinases have of PTP and SH2 domains within the same ponents in biology is likely to be similar.
been found, and the dynamic control of genomes (Pincus et al., 2008). Although A toolkit of writer, reader, and eraser
tyrosine phosphorylation may be primarily fungi and Dictyostelium have 5 PTP functions may be of full use only when all
regulated by PTPs. Although functionally proteins, metazoans, and choanoflagel- components are present. Thus, it may
important for aggregation and differentia- lates have 30–40 per genome. Similarly, be common for any system of this type
tion, the pTyr signaling system has not Dictyostelium has approximately ten to show a quantum ‘‘all-or-none’’ expan-
acquired the pervasive influence evident SH2 domain-containing proteins (fungi sion only when the final piece of the toolkit
in M. brevicollis and metazoans, perhaps have none), whereas metazoans and emerges.
because of the lack of an efficient tyrosine choanoflagellates have 100 each.
kinase. Put another way, Dicytostelium Thus, both PTP and SH2 proteins Applying the New pTyr Toolkit
has effective pTyr readers and erasers, undergo a roughly 10-fold increase in to Different Functions
but the writer is poorly developed. number per genome after the emergence Although both choanoflagellate and
of the TyrK domain. Moreover, the metazoan lineages show a large expan-
Invention of TyrK and Expansion proteins containing SH2 and PTP sion of the three-part pTyr regulatory
of the pTyr Toolkit domains become far more complex and machinery, the way in which these
Current analysis suggests that the varied (Jin et al., 2009). For example, in components are used appears to be quite
modern tyrosine kinases arose just prior yeast and Dictyostelium, SH2 and PTP different. When one examines the domain
to the evolution of the metazoans. Aside proteins normally are very simple one or types that co-occur with TyrK, SH2, or
from metazoans, canonical tyrosine two domain proteins. However, in line- PTP domains, one finds many distinct
kinases have thus far only been observed ages that have modern TyrK proteins, combinations that are unique to each
in the choanoflagellates, which appear to SH2 and PTP proteins almost always lineage (Pincus et al., 2008; Manning
be the closest known single-celled rela- comprise three to ten domains. et al., 2008). These differences in domain
tives of metazoans (King et al., 2008). These observations are consistent with combinations imply distinct functions for
The absence of significant numbers of the following model. When a far more proteins containing these domains in the
such tyrosine kinases in any other branch efficient TyrK domain—or ‘‘writer’’ func- choanoflagellate and metazoan lineages
of life suggests that this new catalytic tion—emerged, this dramatically increased (Li et al., 2009). Assuming that the evolu-
domain evolved in a recent common the functional utility of the pre-existing PTP tion of new TyrK, SH2, and PTP proteins
ancestor of choanoflagellates and meta- (eraser) and SH2 (reader) domains. As occurred by recombination with new
zoans, most likely as a branch of the older a three-part toolkit—a catalytic domain to accessory domains (Jin et al., 2009; Pei-
Ser/Thr kinases. Some bacteria do have generate pTyr, an interaction domain to sajovich et al., 2010), this observation
specialized tyrosine kinases (BY kinases), bind to these pTyr sites, and an enzyme also implies that the complete signaling
but these resemble P loop NTPases to dephosphorylate them—this domain toolkit emerged only shortly before the
(nucleotide triphosphatases) and are set could be used to encode and execute divergence of metazoans and choanofla-
structurally unrelated to eukaryotic tyro- a far wider and diverse range of regulatory gellates (i.e., shortly before the evolution

Cell 142, September 3, 2010 ª2010 Elsevier Inc. 665


of metazoan multicellularity) and that communication. Because of this valuable gous new molecular information curren-
much of the divergent expansion of these high encoding potential, there is extreme cies could have, in principle, been able
domain families occurred after the lineage pressure to quickly fill this region of the to serve as the substrate for dramatic
split. spectrum. Moreover, the exact type of phenotypic innovation. In this context,
Thus, earlier assumptions that pTyr information carried by each region of the plants make extensive use of protein
signaling is only used in metazoan cell- spectrum is flexible—for example, the phosphorylation and have numerous
cell communication are clearly incorrect. same region of the spectrum can be as- transmembrane receptor Ser/Thr kinases,
Choanoflagellates do not form the com- signed to different functions in different but they lack conventional tyrosine
plex and permanent cell-cell organization countries. We hypothesize that the new kinases, indicating that pTyr-based
that metazoans do, yet surprisingly they pTyr signaling system that emerged prior signaling is not the only mechanism of
have a comparable (if not greater) number to metazoans presented similar new information transfer through which organ-
of pTyr signaling proteins (Manning et al., opportunities to transmit more informa- isms can achieve multicellularity.
2008). Sequencing of other organisms tion. This virgin system was rapidly
that arose near the origins of metazoans exploited, though the way it has been Is pTyr Signaling Saturated?
is ongoing. Preliminary data also suggest used appears to be different in the two How close is the pTyr signaling system to
a large number of pTyr signaling proteins branches (metazoans and choanoflagel- being saturated? Is there still available en-
in other single-celled relatives of meta- lates) that emerged after the complete coding potential that could be tapped for
zoans. Thus, it may be more reasonable toolkit was established. the evolution of new pathways and
to view the pTyr signaling system as an It is tempting to speculate that the behaviors? It is difficult to answer these
innovative but generic information pro- emergence of a new signaling system questions. However, the fact that new
cessing system that could potentially be with high encoding potential may have pTyr signaling proteins appear to be asso-
used for transmitting many different types played a key role in the emergence of ciated with advanced processes like
of information. a new, complex biological function such adaptive immunity suggests that there
as metazoan multicellularity. Such large- was still some remaining encoding poten-
Orthogonal Signaling: A Platform scale phenotypic evolutionary innova- tial in the system as late as the evolution of
for Biological Innovation tions may require and coincide with mammals. The evolutionary history re-
When the three-part pTyr system first innovations in basic molecular compo- constructed here begs many questions.
emerged, it presented a new platform nents (King, 2004; Rokas, 2008). Are there new regulatory toolkits evolving
with which to transmit information that Indeed, we speculate that pTyr now or in the future? Will these new tool-
was orthogonal to pre-existing signaling signaling may provide a more general kits be the substrate required for the
systems. Because it was based on a model for the generation of multicompo- next big evolutionary innovation?
distinct covalent modification, new regu- nent biological systems, involving first a The importance of new molecular tool-
latory circuits could be assembled with limited stepwise development of elements kits is conversely also very relevant to
these components without significant that together have a rudimentary biolog- the emerging field of synthetic biology, in
cross-interference with pre-existing net- ical utility, followed by an explosive which the goal is to engineer cellular
works. Thus, this brand new signaling spread, once all of the components of systems with new functions. A major
apparatus probably had a high encoding the mature system are in place. Explora- potential limitation is how to build such
potential for evolving dramatically new tion of this concept, and further analysis new functions in a reliable fashion that
functions, such as those involved in multi- of the evolution of pTyr signaling, will be does not cross-interfere in unanticipated
cellularity. One possible problem that assisted by the increasing sequence infor- ways with existing systems (Lim, 2010).
could be caused by the expansion of the mation being gathered for both unicellular Can we develop new synthetic molecular
new pTyr signaling enzymes might be and multicellular eukaryotes (Srivastava signaling currencies that are orthogonal
excessive general phosphorylation of et al., 2010), which will no doubt yield to existing natural ones, and would these
tyrosine residues throughout the pro- surprises akin to the discovery of exten- systems dramatically facilitate our ability
teome. Interestingly, however, organisms sive pTyr signaling in M. brevicollis. More- to reliably and predictably endow cells
using pTyr signaling may have developed over, as genomic information bracketing with innovative new functions?
a simple solution to deal with this other major evolutionary transitions
problem—a decrease in the tyrosine becomes available, it will be interesting Conclusions
content of proteins across the proteome to see whether these innovations are Current data suggest that PTP and SH2
is observed to correlate with tyrosine also associated with the explosive expan- domains evolved before modern TyrK
kinase expansion (Tan et al., 2009). sion of new molecular toolkits. domains, most likely to process pTyr
A new orthogonal signaling system like A key point here is that the specific modifications sporadically catalyzed by
the pTyr signaling platform can be viewed emergence of the pTyr toolkit may not Ser/Thr kinases. However, the PTP and
as analogous to a newly opened region in have been essential for the evolution of SH2 domain protein families did not
the telecommunications spectrum. New multicellularity, but rather, any number of expand dramatically until the emergence
frequencies provide the opportunity for new orthogonal signaling toolkits with of an efficient TyrK. We postulate that
transmitting large amounts of information the same high encoding potential could only with the complete toolkit of writer
as there is little interference from existing have served a similar role. Other analo- (TyrK), reader (SH2), and eraser (PTP)

666 Cell 142, September 3, 2010 ª2010 Elsevier Inc.


domains, was the full encoding potential three-part system. One cannot help but Kennelly, P.J. (2001). Chem. Rev. 101, 2291–2312.
of this system unleashed, leading to rapid wonder what other simple pieces of Kholodenko, B.N. (2006). Nat. Rev. Mol. Cell Biol.
expansion and elaboration of these molecular machinery may be lying around 7, 165–176.
domain families. This type of explosive in today’s biological systems, of limited King, N. (2004). Dev. Cell 7, 313–325.
increase in component usage may prove utility now, but awaiting the emergence King, N., Hittinger, C.T., and Carroll, S.B. (2003).
to be common to all multipart molecular of some as yet unknown complementary Science 301, 361–363.
systems. The emergence of the modern component that will generate a complete King, N., Westbrook, M.J., Young, S.L., Kuo, A.,
TyrK maps just prior to the split between toolkit that will help to drive future evolu- Abedin, M., Chapman, J., Fairclough, S., Hellsten,
metazoans and choanoflagellates. These tionary innovation. U., Isogai, Y., Letunic, I., et al. (2008). Nature 451,
two lineages appear to have used this 783–788.
new molecular communication system in ACKNOWLEDGMENTS Langenick, J., Araki, T., Yamada, Y., and Williams,
distinct ways—multicellular metazoans J.G. (2008). J. Cell Sci. 121, 3524–3530.
We thank D. Pincus, B. Mayer, P. Beltrao, O. Hoeller,
used it for cell-cell coordination, whereas Lee, D.C., and Jia, Z. (2009). Trends Biochem. Sci.
R.Linding, G. Superti-Furga, N.King, N.Helman, L.
unicellular choanoflagellates used it for 34, 351–357.
Holt, A. Horwitz, T. Miller, G.Manning, T. Hunter,
distinct but as yet uncharacterized D.Morgan, J. Williams, H. Bourne, and I. Ernberg Li, W., Scarlata, S., and Miller, W.T. (2009).
functions. for helpful comments. This work was supported by Biochemistry 48, 5180–5186.
Thus, we are able to reconstruct a plau- the Howard Hughes Medical Institute (W.L.), the Lim, W.A. (2010). Nat. Rev. Mol. Cell Biol. 11,
sible model by which the pTyr signaling National Institutes of health (GM55040, GM62583, 393–403.
GM081879, and EY016546—W.L.), the Packard Manning, G., Young, S.L., Miller, W.T., and Zhai, Y.
machinery could have evolved in a rela-
Foundation (W.L.), the National Science Foundation (2008). Proc. Natl. Acad. Sci. USA 105, 9674–9679.
tively simple stepwise manner into what Synthetic Biology Engineering Research Center
today is a complex and highly interdepen- Moniakis, J., Funamoto, S., Fukuzawa, M.,
(W.L.), the Canadian Institutes for Health Research
dent system. In this model, evolution is Meisenhelder, J., Araki, T., Abe, T., Meili, R.,
(T.P.—MOP-6849), Genome Canada (T.P.), and
Hunter, T., Williams, J., and Firtel, R.A. (2001).
opportunistic and forward looking, the Canadian Cancer Society Research Institute
Genes Dev. 15, 687–698.
borrowing, and repurposing machinery (T.P.).
Pawson, T. (1995). Nature 373, 573–580.
that pre-exists. The first simple PTP
proteins likely arose from the more REFERENCES Pawson, T. (2004). Cell 116, 191–203.

ancient Ser/Thr phosphatase family and Pawson, T., Olivier, P., Rozakis-Adcock, M.,
Alonso, A., Sasin, J., Bottini, N., Friedberg, I., McGlade, J., and Henkemeyer, M. (1993). Philos.
may have been maintained initially as Friedberg, I., Osterman, A., Godzik, A., Hunter, Trans. R. Soc. Lond. B Biol. Sci. 340, 279–285.
a way to reverse the unavoidable occa- T., Dixon, J., and Mustelin, T. (2004). Cell 117,
sional tyrosine phosphorylation event Peisajovich, S.G., Garbarino, J.E., Wei, P., and
699–711.
Lim, W.A. (2010). Science 328, 368–372.
catalyzed by a Ser/Thr kinase. In some Alwayn, V. (2004). Optical Network Design and Im-
cases, like the MAPKs, which are present plementation (Indianapolis, IN: Cisco Press). Pincus, D., Letunic, I., Bork, P., and Lim, W.A.
(2008). Proc. Natl. Acad. Sci. USA 105, 9680–9684.
in all eukaryotes, these tyrosine phos- Bhattacharyya, R.P., Reményi, A., Yeh, B.J., and
phorylation events appear to have Lim, W.A. (2006). Annu. Rev. Biochem. 75, Rokas, A. (2008). Annu. Rev. Genet. 42, 235–251.

become exploited and fixed as actual 655–680. Schieven, G., Thorner, J., and Martin, G.S. (1986).
parts of signal transmission, alongside Bordo, D., and Bork, P. (2002). EMBO Rep. 3, 741– Science 231, 390–393.
746. Srivastava, M., Simakov, O., Chapman, J., Fahey,
Ser/Thr phosphorylation events. SH2
domains also likely arose from a pre-ex- Dengl, S., Mayer, A., Sun, M., and Cramer, P. B., Gauthier, M.E., Mitros, T., Richards, G.S., Con-
(2009). J. Mol. Biol. 389, 211–225. aco, C., Dacre, M., Hellsten, U., et al. (2010).
isting fold in the SPT6 protein, which is
Hunter, T. (2009). Curr. Opin. Cell Biol. 21, Nature 466, 720–726.
found in all eukaryotes but has no pTyr
140–146. Sugden, C., Ross, S., Bloomfield, G., Ivens, A.,
binding activity. But this fold, once co-
Jin, J., Xie, X., Chen, C., Park, J.G., Stark, C., Skelton, J., Mueller-Taubenberger, A., and Wil-
opted for this function, began to expand, liams, J.G. (2010). J. Biol. Chem. 285, 22927–
James, D.A., Olhovsky, M., Linding, R., Mao, Y.,
most likely because of its ability to and Pawson, T. (2009). Sci. Signal. 2, ra76. 22935.
contribute to a wider range of modular Kawata, T., Shevchenko, A., Fukuzawa, M., Tan, C.S., Pasculescu, A., Lim, W.A., Pawson, T.,
signaling events. But the full utility of these Jermyn, K.A., Totty, N.F., Zhukovskaya, N.V., Ster- Bader, G.D., and Linding, R. (2009). Science 325,
components was only unleashed upon ling, A.E., Mann, M., and Williams, J.G. (1997). Cell 1686–1688.
the emergence of the modern TyrK 89, 909–916. Tonks, N.K., and Neel, B.G. (2001). Curr. Opin. Cell
domain, which led to the highly expanded Kay, R.R. (1997). Curr. Biol. 7, R723–R725. Biol. 13, 182–195.

Cell 142, September 3, 2010 ª2010 Elsevier Inc. 667


Leading Edge
Correspondence

the inhibitor (Figure S1B, lanes 3 and 4).


What Controls T Cell Receptor Time course experiments indicated that
rather than enhancing it, the Emut1+2
Phosphorylation? mutations somewhat slowed the kinetics
of CD33CD phosphorylation (data not
shown). In vivo, therefore, the wild-type
In a study published in Cell, Xu et al. (2008) To show that CD33CD is not phosphor- form of CD33CD seemed to be at least
raised the intriguing possibility that tyro- ylated in resting Jurkat T cells, Xu et al. as accessible to tyrosine kinases as
sine residues present in the immunore- used a chimeric protein consisting of the mutated, membrane nonassociating
ceptor tyrosine-based activation motifs murine CD33CD fused to the extracellular form of the protein when kinase activity
(ITAMs) of the cytoplasmic domain of the and transmembrane domains of the could be observed by inhibiting phospha-
CD33 subunit of the T cell receptor (TCR) human natural killer cell inhibitory recep- tase activity. Finally, CD33 was among the
are sequestered in the membrane. Here, tor KIR2DL3. We generated FLAG-tagged more heavily tyrosine phosphorylated
they are inaccessible to tyrosine kinases forms of this protein and a second proteins present in whole-cell lysates
that would otherwise drive signaling. chimera consisting of the extracellular from pervanadate-treated wild-type
As a consequence, the authors proposed and transmembrane domains of KIR2DL3 Jurkat T cells (Figure S1C), emphasizing
that ‘‘triggering’’ of the TCR—that is, the fused with CD33CD mutated at the set of its accessibility to kinases relative to other
events leading to phosphorylation of the clustered positively charged residues kinase targets.
receptor—must be dependent upon proposed to mediate membrane asso- How can our observations be recon-
some, as yet undefined, process that ciation, that is, the mutant Emut1+2 ciled with those of Xu et al.? Our experi-
liberates the ITAMs from the membrane. described by Xu et al. (see Experimental ments suggest that membrane associa-
In an accompanying Preview, Kuhns and Procedures). We expressed the two tion, if it occurs, has less impact on
Davis (2008) invoked the idea that mem- chimeras at similar levels in Jurkat CD33CD phosphorylation than might
brane association constitutes a ‘‘safety T cells using a lentivirus expression have been expected on the basis of the
catch’’ that prevents untoward triggering system (Figure S1A) and probed for Xu et al. work. The strongest evidence for
of the TCR. Such effects add an additional phosphorylation of CD33CD by western lipid association is, of course, the NMR
layer of complexity to the TCR triggering blotting (Figure S1B). Neither KIR2DL3/ structure of CD33CD stably bound to
problem. CD33CD nor KIR2DL3/CD33CDEmut1+2 acidic lipids. It should be borne in mind,
Using equilibrium dialysis, Xu et al. immunoprecipitated with anti-FLAG anti- however, that we know little about the
showed that the cytoplasmic domain of body showed detectable phosphorylation actual distribution of inner leaflet
CD33 (CD33CD) partitioned into acidic in resting T cells (Figure S1B, lanes 1 lipids around receptors embedded in the
rather than zwitterionic lipids. They then and 2). Even after very long exposures of membrane. Recent work by Zech et al.
went on to derive a structural model for the blots, we were unable to detect phos- (2009) shows that the acidic phospholipid
ITAMs bound by acidic lipids using phorylation over background levels of phosphatidylserine is enriched in mem-
nuclear magnetic resonance (NMR)- cross-reactivity of the antibody and we brane sheets isolated from activated
based methods. A new in vivo FRET- saw no differences in signal for the wild- T cells using beads coated with anti-CD3
based assay suggested that the associa- type versus the mutant proteins (Fig- antibodies, but the extent to which this
tion of the ITAM with the membrane was ure S1B). This was also the case for reflects the distribution of lipids in the
dependent on a cluster of positively wild-type and Emut1+2-mutated forms immediate vicinity of the TCR in resting
charged residues in the membrane-proxi- of full-length CD33 incorporated into the or activated cells is unclear. The density
mal amino-terminal region of CD33CD; TCR complex (data not shown). or properties of the acidic lipids adjacent
mutation of these residues reduced the These findings suggest either that to CD33 in resting cells might be insuffi-
association of CD33CD with the mem- the Emut1+2 mutations do not release cient to sustain an interaction of the type
brane as detected by the FRET assay. the ITAMs from the membrane or that, demonstrated in vitro by Xu et al. Whatever
The obvious question was, does mem- in vivo, the association with the mem- interaction ITAMs have with membranes
brane association prevent kinases from brane is weaker than expected and in vivo might be too weak and dynamic to
accessing ITAMs in vivo? Xu et al. showed some other process prevents CD33CD prevent access of the kinases.
that the wild-type CD33 ITAM is not phos- phosphorylation in resting cells. We sus- The reasons for the low levels of CD33CD
phorylated in resting T cells, implying that pected that tyrosine phosphatases might phosphorylation in resting T cells remain to
it may indeed be completely inaccessible be responsible for the lack of phosphory- be fully worked out. In any discussion of the
to kinases. Somewhat surprisingly, how- lation of the wild-type and mutant forms of possible constraints on tyrosine phosphor-
ever, they did not go on to clinch the argu- CD33CD. To test this, we incubated cells ylation of the TCR, some consideration
ment by showing that there was increased expressing the chimeric proteins with must be given to the protein tyrosine phos-
phosphorylation of the membrane nonas- the tyrosine phosphatase inhibitor, perva- phatases (PTPs) present at the cell surface,
sociating CD33CD mutant. The results nadate (Swarup et al., 1982). Both the such as CD45 (Hermiston et al., 2003).
of this experiment turn out to be inconsis- wild-type and mutated forms of CD33CD Leukocytes express as many as 105 CD45
tent with the predictions of Xu and were heavily phosphorylated following molecules, i.e., 2–3 for every TCR (Wil-
colleagues. incubation of the cells for 20 min with liams and Barclay, 1986), each with very

668 Cell 142, September 3, 2010 ª2010 Elsevier Inc.


broad substrate specificity (Barr et al., ACKNOWLEDGMENTS REFERENCES
2009); the catalytic activities of PTPs have
This work was funded by the Wellcome Trust, the Barr, A.J., Ugochukwu, E., Lee, W.H., King, O.N.,
been estimated to be 10- to 1000-fold
United Kingdom Medical Research Council, and Filippakopoulos, P., Alfano, I., Savitsky, P.,
higher than that of tyrosine kinases the Portuguese Fundação para a Ciência e a Tec- Burgess-Brown, N.A., Müller, S., and Knapp, S.
(discussed by Fischer et al., 1991). Another nologia. (2009). Cell 136, 352–363.
contributing factor is the activity of inhibi- Fischer, E.H., Charbonneau, H., and Tonks, N.K.
tory kinases such as Csk, which also Ricardo A. Fernandes,1,4 Chao Yu,1,4 (1991). Science 253, 401–406.
constrain T cell activation (Schoenborn Alexandre M. Carmo,2 Hermiston, M.L., Xu, Z., and Weiss, A. (2003).
et al., 2009). Progress in understanding Edward J. Evans,1 Annu. Rev. Immunol. 21, 107–137.
TCR triggering will require the teasing P. Anton van der Merwe,3 Kuhns, M.S., and Davis, M.M. (2008). Cell 135,
apart of these and other competing and Simon J. Davis1,* 594–596.
1Nuffield Department of Clinical Medicine
factors and the identification of those O’Shea, J.J., McVicar, D.W., Bailey, T.L., Burns,
factors with the largest impact on net and Medical Research Council Human
C., and Smyth, M.J. (1992). Proc. Natl. Acad. Sci.
Immunology Unit, The Weatherall Institute of
phosphorylation of the TCR. The effects USA 89, 10306–10310.
Molecular Medicine, University of Oxford,
of pervanadate on phosphorylation levels Schoenborn, J.R., Zhang, C., Shokat, K.M., and
Oxford OX3 9DU, UK
in resting T cells observed by ourselves 2Group of Cell Activation and Gene Weiss, A. (2009). J. Immunol. 182, Abstract 35.4.
(Figures S1B and S1C) and by others Expression, Institute for Molecular and Swarup, G., Cohen, S., and Garbers, D.L. (1982). Bi-
many years ago (e.g., O’Shea et al., 1992) Cellular Biology, 4150-180 Porto, Portugal ochem. Biophys. Res. Commun. 107, 1104–1109.
suggest that PTPs could be key. For us, and Abel Salazar Institute for Biomedical Williams, A.F., and Barclay, A.N. (1986). Glycopro-
the sheer weight of the numbers also Sciences, University of Porto, tein antigens of the lymphocyte surface and their
warrants their serious consideration. Porto 4099-003, Portugal purification by antibody affinity chromatography.
3Sir William Dunn School of Pathology, In Handbook of Experimental Immunology (Oxford:
The University of Oxford, Oxford OX1 3RE, Blackwell Scientific Publications), pp. 22.1–22.24.
UK Xu, C., Gagnon, E., Call, M.E., Schnell, J.R., Schwi-
4These authors contributed equally to this eters, C.D., Carman, C.V., Chou, J.J., and
SUPPLEMENTAL INFORMATION
work Wucherpfennig, K.W. (2008). Cell 135, 702–713.
Supplemental Information includes Experimental *Correspondence: simon.davis@ndm.ox. Zech, T., Ejsing, C.S., Gaus, K., de Wet, B., Shev-
Procedures and one figure and can be found with ac.uk chenko, A., Simons, K., and Harder, T. (2009).
this article online at doi:10.1016/j.cell.2010.08.018. DOI 10.1016/j.cell.2010.08.018 EMBO J. 28, 466–476.

membrane bound in these transfected


Response nonstimulated Jurkat cells. However, we
did not claim that all CD33 cytoplasmic
Multilayered Control of T Cell domains in a given T cell are completely
bound to the plasma membrane. This
would be impossible in a biological sys-
Receptor Phosphorylation tem because binding events always
follow equilibrium conditions, with the
size of the bound versus free fractions
We would like to respond to the Corre- munoreceptor tyrosine-based activation being determined by the ratio of on and
spondence by Fernandes et al., but first motif (ITAM) partitioned into the hydro- off rates. It follows that changes in equilib-
we will summarize the data in our Cell phobic core of the bilayer in a dynamic rium can result in dissociation of CD33CD
paper (Xu et al., 2008). Our study used manner, with substantial movement of from the membrane. Our data showed
a biophysical approach to examine the the two tyrosines and other elements of a structure with substantial mobility in
binding of the CD33 cytoplasmic domain the cytoplasmic domain (Figure 6, Xu which changes in equilibrium, such as
(CD33CD) to the plasma membrane. We et al., 2008). Microscopy data yielded recruitment of the tyrosine kinase Lck,
reported a new FRET assay to examine similar FRET values for CD33CD tagged could enable phosphorylation.
CD33CD binding to the inner leaflet of with C-terminal teal fluorescent protein We wish to point out that we did not
the plasma membrane in live Jurkat cells, (TFP) and for a positive control in which claim that binding of CD33CD to the
a transformed T cell line. Furthermore, we TFP was in close proximity to the plasma membrane was the only mechanism that
developed an approach to determine membrane (there was a three amino acid prevents spontaneous T cell signaling.
the NMR structure of CD33CD bound in linker between the transmembrane and It is well established that there is
a lipid bilayer environment. The structure TFP domains). These data indicated that multilayered control of T cell receptor
showed that the two tyrosines of the im- most of the cytoplasmic domain was (TCR) signaling because complete

Cell 142, September 3, 2010 ª2010 Elsevier Inc. 669


phosphorylation of even one or a few TCRs after 2 min, whereas phosphorylation of assessed whether the negative charge of
is sufficient to result in T cell activation the mutant was delayed, with a 3.5- to the inner leaflet of the plasma membrane
(Bergman et al., 1992; Davis and van der 5-fold reduction at 10 min (Figure S1B). could be affected. Phosphatidylserine is
Merwe, 2006). Without highly effective However, both proteins were phosphory- the most abundant negatively charged
mechanisms to prevent spontaneous lated to a similar degree at a later time lipid in the inner leaflet. We used a highly
signaling, rampant chronic inflammation point (30 min), as Fernandes et al. also specific calcium-independent phosphati-
and autoimmunity would result. Sponta- showed. The greatly reduced interaction dylserine probe, the Lactadherin C2
neous signaling is inhibited by the Csk of the mutant protein with Lck as well as domain (Lact-C2), to study the dynamics
kinase, which phosphorylates Lck kinase the multiple control mechanisms regu- of phosphatidylserine distribution (Yeung
at inhibitory tyrosine 505, as well as lating T cell activation could account for et al., 2008). Jurkat cells were transduced
multiple phosphatases (Bergman et al., the absence of observable phosphoryla- with a lentivirus containing the Lact-C2
1992; Davis and van der Merwe, 2006). In tion of this particular CD33 mutant in non- probe, and the probe’s localization in live
agreement with Fernandes et al., we did stimulated Jurkat cells. It is not known cells was analyzed by confocal micros-
not detect phosphorylation of the why mutation of six basic residues in the copy (Figure S1E). Under resting condi-
CD33CD Emut1+2 mutant protein in Jurkat N-terminal part of CD33CD reduces the tions, the Lact-C2 probe predominantly
cells (Figure S1B, CD33-Mut time 0), which interaction with Lck, but conformational associated with the plasma membrane in
was a negative control for the FRET exper- changes in the cytoplasmic domain or 75% of cells. In contrast, pervanadate
iments. However, if binding of CD33 loss of long-range charge interactions treatment reduced the Lact-C2 signal
to the membrane together with other inhib- with Lck could be involved. at the plasma membrane and increased
itory mechanisms guards against sponta- Pervanadate oxidizes the active site the amount of this phosphatidylserine
neous phosphorylation, this result is not cysteines of many different phosphatases probe in internal membranes in 92%
surprising. Phosphorylation may only be and is generated by mixing hydro- of cells, suggesting that the plasma
favored with appropriate TCR localization gen peroxide with sodium orthovanadate membrane inner leaflet had a reduced
into microclusters that exclude the CD45 (Huyer et al., 1997). Given the potential negative charge (Figures S1E and S1F).
phosphatase (Varma et al., 2006), as well for unanticipated effects, we used our Phosphatidylserine at the cell surface
as simultaneous colocalization of active FRET assay to examine the conse- was also slightly increased, as measured
Lck bound to CD4/CD8 coreceptors close quences of pervanadate treatment on by annexin-V labeling (Figure S1G). We
to the clustered TCR-CD3 complexes. CD33CD membrane binding (Xu et al., conclude that pervanadate treatment
Fernandes et al. assume that the 2008). The assay measures the interac- reduces the negative charge of the inner
CD33CD Emut1+2 mutant protein would tion of a TFP domain attached to the leaflet and induces release of CD33CD
normally interact with Lck. However, the C terminus of CD33CD with a lipophilic from the plasma membrane. These find-
loss of six basic residues substantially dye R18 (octadecyl rhodamine B) incor- ings complicate the interpretation of
changes its charge properties. We porated in the plasma membrane; label- experiments using pervanadate to assess
therefore directly compared the phos- ing the cells with R18 caused a decrease whether CD33CD is bound to the plasma
phorylation of the mutant and wild-type in the TFP signal (quenching) due to membrane.
CD33CD proteins in an in vitro phosphor- energy transfer from TFP to R18 (Fig- A recent study confirmed that CD33CD
ylation assay with the purified cyto- ure S1C). In the absence of pervanadate, binds to the plasma membrane (Deford-
plasmic domains of the wild-type and the FRET efficiency was high (average Watts et al., 2009), and prior work has
Emut1+2 proteins and purified Lck in the 50%; Figure S1D), which is similar to shown that CD3z is membrane bound
absence of liposomes (Figure S1A). We the FRET signal obtained with the positive (Aivazian and Stern, 2000). Furthermore,
observed greatly reduced phosphoryla- control in which TFP is positioned close other cytoplasmic peptides have been
tion of the mutant protein, even though to the plasma membrane (a three amino shown to interact with the inner leaflet
equal amounts of wild-type and mutant acid linker between the transmembrane by similar mechanisms. McLaughlin and
proteins were used. The mutant protein domain and TFP) (Xu et al., 2008). After colleagues showed that the MARCKS
was not detectably phosphorylated with 20 min of pervanadate treatment, the peptide binds to the inner leaflet using
100 ng of Lck kinase and was only phos- FRET signal was reduced on average clusters of basic amino acids and five
phorylated at low levels with 400 ng Lck; to 15% (Figure S1D), similar to the phenylalanine residues (Zhang et al.,
the wild-type protein was robustly phos- negative control with a 50 amino acid flex- 2003). NMR measurements demon-
phorylated under both conditions. Fer- ible linker between the transmembrane strated directly that these phenylalanine
nandes et al. also mention delayed phos- domain and TFP (Xu et al., 2008). Perva- residues are located in the hydrophobic
phorylation kinetics for this mutant nadate treatment thus results in dissocia- core of the lipid bilayer, similar to the tyro-
protein in cells treated with pervanadate, tion of the CD33 cytoplasmic domain from sines of CD33CD (Zhang et al., 2003).
which inhibits a broad range of phospha- the membrane. Also, the cytoplasmic small GTPase Rit
tases (data not shown). We confirmed We next examined the potential mech- localizes to the plasma membrane using
this finding and observed substantially anisms that could contribute to this effect. three clusters of basic amino acids
delayed phosphorylation of the mutant We had previously shown that basic resi- and interspersed hydrophobic residues;
protein after pervanadate treatment: the dues of CD33CD are critical for its binding mutation of a tryptophan in this segment
wild-type protein was phosphorylated to the plasma membrane. Therefore, we results in loss of membrane binding

670 Cell 142, September 3, 2010 ª2010 Elsevier Inc.


(Heo et al., 2006). Therefore, usage of Etienne Gagnon,1 Chenqi Xu,2 Davis, S.J., and van der Merwe, P.A. (2006). Nat.
both basic and hydrophobic residues Wei Yang,2 H. Hamlet Chu,1 Immunol. 7, 803–809.
for plasma membrane binding is a more Matthew E. Call,3 James J. Chou,3
general theme that extends beyond the and Kai W. Wucherpfennig1,4,* Deford-Watts, L.M., Tassin, T.C., Becker, A.M.,
1Department of Cancer Immunology & AIDS, Medeiros, J.J., Albanesi, J.P., Love, P.E., Wulfing,
CD33 and z cytoplasmic domains of the
Dana-Farber Cancer Institute, Boston, C., and van Oers, N.S. (2009). J. Immunol. 183,
TCR-CD3 complex. 1055–1064.
MA 02115, USA
In their Correspondence, Fernandes 2Institute of Biochemistry and Cell Biology,
et al. raise questions about the functional Shanghai Institutes for Biological Sciences, Heo, W.D., Inoue, T., Park, W.S., Kim, M.L., Park,
relevance of the binding of CD33CD to the Chinese Academy of Sciences, B.O., Wandless, T.J., and Meyer, T. (2006).
plasma membrane during TCR activation. Shanghai 200031, China Science 314, 1458–1461.
It will be important to further study the 3Department of Biological Chemistry and

functional significance of the binding of Molecular Pharmacology Huyer, G., Liu, S., Kelly, J., Moffat, J., Payette, P.,
4Department of Neurology and Program in Kennedy, B., Tsaprailis, G., Gresser, M.J., and
CD33CD to the plasma membrane in
a physiological setting using primary Immunology Ramachandran, C. (1997). J. Biol. Chem. 272,
Harvard Medical School, Boston, MA 02115, 843–851.
T cells because the early events in TCR
USA
triggering are very complex, and subtle
*Correspondence: kai_wucherpfennig@dfci. Varma, R., Campi, G., Yokosuka, T., Saito, T., and
changes may result in receptor activation
harvard.edu Dustin, M.L. (2006). Immunity 25, 117–127.
after ligand binding. Analysis of multiple DOI 10.1016/j.cell.2010.08.019
CD33CD mutant proteins will be useful, Xu, C., Gagnon, E., Call, M.E., Schnell, J.R., Schwi-
including mutants with reduced rather eters, C.D., Carman, C.V., Chou, J.J., and
than complete loss of membrane binding. REFERENCES Wucherpfennig, K.W. (2008). Cell 135, 702–713.

Aivazian, D., and Stern, L.J. (2000). Nat. Struct. Yeung, T., Gilbert, G.E., Shi, J., Silvius, J., Kapus,
SUPPLEMENTAL INFORMATION
Biol. 7, 1023–1026. A., and Grinstein, S. (2008). Science 319, 210–213.
Supplemental Information includes Experimental Bergman, M., Mustelin, T., Oetken, C., Partanen,
Procedures and one figure and can be found with J., Flint, N.A., Amrein, K.E., Autero, M., Burn, P., Zhang, W., Crocker, E., McLaughlin, S., and Smith,
this article online at doi:10.1016/j.cell.2010.08.019. and Alitalo, K. (1992). EMBO J. 11, 2919–2924. S.O. (2003). J. Biol. Chem. 278, 21459–21466.

Cell 142, September 3, 2010 ª2010 Elsevier Inc. 671


Leading Edge
Previews

Fishing Out a Sensor for Anti-inflammatory Oils


Alan R. Saltiel1,*
1Life Sciences Institute, Departments of Internal Medicine and Molecular and Integrative Physiology, University of Michigan Medical School,

Ann Arbor, MI 48109, USA


*Correspondence: saltiel@lsi.umich.edu
DOI 10.1016/j.cell.2010.08.022

The u-3 fatty acids have anti-inflammatory and antidiabetic effects in humans. Now, Oh et al. (2010)
demonstrate that the G protein-coupled receptor GPR120 is a receptor for u-3 fatty acids on
macrophages and fat cells. Activation of GPR120 by u-3 fatty acids inhibits multiple inflammation
cascades in macrophages and reverses insulin resistance in obese mice.

Evidence providing an inflammatory link evidence indicates that u-3 fatty acids stream signaling molecules (Rajagopal
between obesity and type 2 diabetes is derived from fish oils, such as docosahex- et al., 2010). In a series of gene silencing
accumulating. In numerous animal and anoic acid (DHA) and eicosapentanoic experiments, Oh and colleagues demon-
clinical studies, obesity is associated acid (EPA), have an anti-inflammatory strate that b-arrestin2 is essential for
with a state of low-grade, chronic inflam- effect (Serhan et al., 2008). the anti-inflammatory effects of u-3 fatty
mation in liver and adipose tissue, To sort out the metabolic impact of acids in macrophage cells, but Gq is
which includes activation of the innate various types of fatty acids, Oh et al. surprisingly dispensable for this process.
immune system and the appearance of characterized the tissue expression pat- Moreover, b-arrestin2 inhibits both the
proinflammatory immune cells (Hotamisli- terns of five G protein-coupled receptors JNK and NF-kB pathways by seques-
gil, 2006; Shoelson and Goldfine, 2009). (GPCRs) known to bind and respond tering the TAK1 binding protein TAB1.
Most notably, macrophages conspire to fatty acids. Among these receptors, The inhibition of TAB1 prevents phos-
with increased levels of inflammatory GPR120 was the only one with an expres- phorylation and thus activation of IkB
cytokines to attenuate insulin action and sion profile that correlated well with a kinase upstream of NFkB and MKK4
increase lipid accumulation. potential role in regulating metabolism. (mitogen-activated protein kinase kinase 4)
Recent studies indicate that the NF-kB They found that GPR120 is highly ex- upstream of JNK.
and JNK (JUN N-terminal kinase) path- pressed in adipose tissue macrophages, These new insights into the sensing and
ways play important roles in the com- fat cells, and specialized macrophages signaling of GPR120 offered Oh and
munication among macrophages, adipo- in the liver called Kupffer cells. Moreover, colleagues a unique opportunity to study
cytes, and liver cells (Arkan et al., 2005; a high-fat diet increases the expression of the molecular mechanisms underlying
Chiang et al., 2009; Solinas et al., 2007). this receptor on macrophages, suggest- the metabolic benefits of u-3 fatty acids
However, key questions still remain about ing that GPR120 might be controlled by in vivo. First, Oh and colleagues establish
the initial establishment of this inflamma- inflammatory signals. that the high-fat diet given to mice in the
tory state. Do fat and liver cells first sense GPR120 is an orphan receptor for laboratory is low in u-3 fatty acids. They
an overload of energy and respond by which no endogenous ligands are known. then show that supplementing this diet
secreting chemokines, which then recruit Using a heterologous reporter system, Oh with DHA and EPA reverses the delete-
macrophages to the liver and fat? Or do et al. now find that the u-3 fatty acids rious effects that the high-fat diet has on
fatty acids in the diet directly initiate the DHA, EPA, and palmitoleate are agonists glucose homeostasis and lipid storage
inflammatory cascade, and, if so, which of GPR120. Furthermore, activation of (i.e., steatosis). Although the authors do
ones? In this issue of Cell, Oh et al. GPR120 by DHA antagonizes the proin- not address whether u-3 fatty acids can
(2010) address the latter question by iden- flammatory effects of TNFa and lipopoly- prevent insulin resistance or glucose intol-
tifying a sensor for u-3 fatty acids that is saccharide in a macrophage cell line. erance in mice, they do demonstrate that
upregulated in obese mice. Furthermore, DHA not only blocks the NFkB and JNK DHA and EPA reverse insulin resistance
activation of this receptor exerts potent pathways but also prevents expression caused by the high-fat diet. Moreover,
anti-inflammatory effects that improve of cytokines (Figure 1, bottom right). disruption of the GPR120 gene abolishes
insulin resistance and other symptoms of GPR120 is known to couple with the the benefits of u-3 fatty acids on glucose
metabolic syndrome in mice. Gq/11 family of G proteins. After ligand binds homeostasis and insulin sensitivity in
Previous studies suggest that saturated and Gq/11 is released, G protein receptor mice. These results demonstrate the
fatty acids promote inflammation by acti- kinases phosphorylate the receptor. This crucial role GPR120 plays in the meta-
vating Toll-like receptor 4 (TLR4) on fat generates binding sites for b-arrestins, bolic benefits of DHA and EPA. Interest-
cells and macrophages (Shi et al., 2006). which mediate internalization and down- ingly, mice receiving bone marrow
In contrast, most unsaturated fats are regulation of the receptors. However, transplants from the GPR120-deficient
metabolically neutral. However, recent b-arrestins can also interact with down- mice are also resistant to the beneficial

672 Cell 142, September 3, 2010 ª2010 Elsevier Inc.


GPR120 expression, and what is its phys-
iological role in adipose and liver tissue?
Also, does GPR120 respond to endoge-
nous ligands to control macrophage
activity? Although the new study by Oh
and colleagues explains how activation
of GPR120 inhibits inflammatory path-
ways, it is still unknown how this receptor
increases the presence of anti-inflamma-
tory M2 macrophages in adipose tissue
and from where these cells arise. Do M2
macrophages (or their precursors)
express GPR120 in order to develop or
maintain the anti-inflammatory pheno-
type, or does activation of GPR120
induce the transdifferentiation of inflam-
matory M1 macrophages to anti-inflam-
matory M2 macrophages in situ?
Finally, previous studies demonstrate
that fish oils have diverse benefits on
multiple tissues. For example, u-3 fatty
acids can inhibit the production of proin-
flammatory eicosanoids and serve as
precursors for resolvins, (i.e., protective
Figure 1. A Model for How u-3 Fatty Acids Preserve Insulin Sensitivity through GPR120 lipids that help reduce inflammation)
A high-fat diet with a disproportionate ratio of saturated fatty acids to u-3 fatty acids triggers activation of
Toll-like receptor 4 (TLR4) in adipocytes and circulating immune cells. This launches an inflammatory
(Serhan et al., 2008). In addition, fish oils
cascade that results in the recruitment of proinflammatory M1 macrophages, increased secretion of help prevent cardiovascular disease and
TNFa, and insulin resistance in adipocytes. The addition of u-3 fatty acids to the diet activates the have positive effects on many inflamma-
G protein-coupled receptor GPR120 on proinflammatory M1 macrophages (Oh et al., 2010), which in
tory disorders, such as arthritis, asthma,
turn attenuates the inflammatory response and recruits anti-inflammatory M2 macrophages to adipose
tissue. Eventually, these M2 macrophages restore secretion of interleukin-10 and improve insulin and ulcerative colitis. Is GPR120 the only
sensitivity. receptor responsible for these various
benefits? Future studies are also needed
to determine whether dietary supple-
properties of DHA and EPA. Thus, the u-3 These cells produce cytokines, such as ments and ingestion of fatty fish can
fatty acids appear to act primarily through TNFa, which further activate the macro- provide high enough concentrations of
macrophages. phages and attenuate insulin action in circulating u-3 fatty acids to promote
Taken together with previous data, adipocytes. Eventually, this leads to GPR120 activation. Nevertheless, the
these findings from Oh and colleagues local and then systemic insulin resistance. new insights presented by Oh and
support a model in which dietary fatty However, these activated M1 macro- colleagues into the anti-inflammatory
acids control the inflammatory properties phages also express elevated levels of mechanisms of u-3 fatty acids provide
of macrophages in adipose tissue by GPR120. Thus, addition of u-3 fatty acids a platform for investigating these impor-
regulating the activity and expression to the diet stimulates GPR120 and tant questions. Plus, the identification of
of opposing receptors (Figure 1). With initiates a signaling pathway through GPR120 pinpoints a new therapeutic
a normal diet containing a balanced b-arrestin2, which blocks the effects of target for treating the inflammatory state
ratio of saturated and u-3 unsaturated TLR4 and inflammatory cytokine recep- associated with obesity and type 2 dia-
fatty acids, anti-inflammatory M2 macro- tors. This reduces the inflammatory state betes. This alone is cause for a bit of
phages protect adipose cells by damp- of these cells and simultaneously pro- excitement.
ening excess inflammation and maintain- motes the return of anti-inflammatory
ing insulin sensitivity of fat cells (Lumeng M2 macrophages to adipose tissue, REFERENCES
et al., 2007). When mice are given which leads to the restoration of insulin
a high-fat diet with excess calories sensitivity. Arkan, M.C., Hevener, A.L., Greten, F.R., Maeda,
S., Li, Z.W., Long, J.M., Wynshaw-Boris, A., Poli,
and little u-3 fatty acids, TLR4 is left This model raises many new questions
G., Olefsky, J., and Karin, M. (2005). Nat. Med.
unchecked in fat cells. The activated about the crosstalk between adipose 11, 191–198.
receptor induces expression and release tissue and immune cells in obesity. First, Chiang, S.H., Bazuine, M., Lumeng, C.N., Geletka,
of chemokines, such as MCP-1 (mono- could GRP120 activation serve as L.M., Mowers, J., White, N.M., Ma, J.T., Zhou, J.,
cyte chemotactic protein-1), which then a homeostatic mechanism in macro- Qi, N., Westcott, D., et al. (2009). Cell 138,
recruit proinflammatory M1 macrophages phages to resolve inflammation in obese 961–975.
into adipose tissue (Lumeng et al., 2007). animals? If so, then what controls Hotamisligil, G.S. (2006). Nature 444, 860–867.

Cell 142, September 3, 2010 ª2010 Elsevier Inc. 673


Lumeng, C.N., Bodzin, J.L., and Saltiel, A.R. R.J. (2010). Proc. Natl. Acad. Sci. USA 107, Shoelson, S.E., and Goldfine, A.B. (2009). Nat.
(2007). J. Clin. Invest. 117, 175–184. 628–632. Med. 15, 373–374.
Oh, D.Y., Talukdar, S., Bae, E.J., Imamura, T.,
Serhan, C.N., Chiang, N., and Van Dyke, T.E.
Morinaga, H., Fan, W., Li, P., Lu, W.J., Watkins, Solinas, G., Vilcu, C., Neels, J.G., Bandyopad-
(2008). Nat. Rev. Immunol. 8, 349–361.
S.M., and Olefsky, J.W. (2010). Cell 142, this issue, hyay, G.K., Luo, J.L., Naugler, W., Grivennikov,
687–698. Shi, H., Kokoeva, M.V., Inouye, K., Tzameli, I., Yin, S., Wynshaw-Boris, A., Scadeng, M., Olefsky,
Rajagopal, S., Kim, J., Ahn, S., Craig, S., Lam, H., and Flier, J.S. (2006). J. Clin. Invest. 116, 3015– J.M., and Karin, M. (2007). Cell Metab. 6,
C.M., Gerard, N.P., Gerard, C., and Lefkowitz, 3025. 386–397.

A New Spin on Planar Cell Polarity


Patricio Olguin1 and Marek Mlodzik1,*
1Department of Developmental and Regenerative Biology, Mount Sinai School of Medicine, New York, NY 10029, USA

*Correspondence: marek.mlodzik@mssm.edu
DOI 10.1016/j.cell.2010.08.025

The generation of planar cell polarity (PCP) and tissue shape during morphogenesis is tightly linked,
but it is not clear how. Aigouy et al. (2010) now show in the developing Drosophila wing that PCP
initially has a radial orientation that becomes realigned to the proximal-distal axis of organ shape
by mechanical forces and cell rearrangements mediated by Dachsous.

Most tissues and organs composed of and tion). The core Frizzled/PCP factors form Vang/Stbm signaling as the nonautono-
organized as epithelial cell layers display, two distinct complexes that become mous behavior of frizzled mutant cell
in addition to the common apical-basal localized asymmetrically to either the patches (clones) affecting the polarity of
epithelial polarity, a polarity within the proximal (Vang/Stbm and associated wild-type cells flanking the frizzled mutant
epithelial plane. This is commonly referred proteins) or the distal side of pupal wing cells (Vinson and Adler, 1987) is already
to as planar cell polarity (PCP). Genetic cells (Frizzled and associated proteins). observed at this stage. Strikingly, in
studies in the fruit fly Drosophila mela- These complexes are stabilized by feed- contrast to the nonautonomous effects
nogaster have established that there are back loop interactions among themselves observed at the distal side of frizzled
two molecular systems coordinating the (Seifert and Mlodzik, 2007; Strutt, 2003). In mutant cell patches in late pupal and adult
cellular asymmetries in the plane of addition, Frizzled and Vang/Stbm protein wings (Vinson and Adler, 1987), early friz-
tissues. These include the Frizzled/PCP complexes may be required earlier to zled clones influence the polarity of wild-
signaling pathway containing the Van- coordinate global tissue polarity/PCP type cells residing between the wing
Gogh (Vang, also known as Strabismus/ within the wing epithelium (Classen et al., margin and the clone within the radial
Stbm) protein and other factors (Strutt, 2005; Wu and Mlodzik, 2009). polarity axis. This confirms a ‘‘signaling
2003; Seifert and Mlodzik, 2007) and Supporting this idea, the new study by axis’’ toward the wing margin at early
a pathway mediated by the protocadher- Aigouy et al. (2010) in this issue of Cell stages. Taken together, the observations
ins Fat and Dachsous (Lawrence et al., helps to establish that subcellular asym- of Aigouy et al. (2010) indicate that (1)
2007). Although the molecular relation- metries among the Frizzled/PCP core PCP, mediated by Frizzled-Vang/Stbm
ships between these two systems are group proteins are already present at signaling, is established during late-
unclear, there is strong evidence that early pupal stages during wing develop- larval/early-pupal stages in a radial axis
they act in parallel, probably affect ment (14–15 hr after puparium formation perpendicular to the margin and (2) the
different effectors, and may compensate or earlier). Strikingly, the Frizzled/PCP polarity/PCP seen in the adult wing is
for each other in some tissues (Lawrence complexes display radial polarity that is a result of cellular rearrangements during
et al., 2007; Wu and Mlodzik, 2009). perpendicular to the wing margin wing morphogenesis within the prox-
In the developing Drosophila wing, (Figure 1A), confirming that coordination imal-distal axis that are dependent on
cellular asymmetries in the plane of the of global Frizzled/PCP signaling is estab- Dachsous.
epithelium are first detected at later pupal lished early in pupal fly wings. The authors How is polarity realigned along the
stages along the proximal-distal axis further demonstrate that these early proximal-distal axis as morphogenesis
(at around 24–30 hr after puparium forma- asymmetries indeed depend on Frizzled- proceeds? As PCP is already established

674 Cell 142, September 3, 2010 ª2010 Elsevier Inc.


at early stages, its final align- cues’’ that orient the initial
ment from the radial orienta- polarity of Frizzled/PCP
tion to the proximal-distal complexes, but this hypoth-
axis must be redirected esis has been challenged by
through active relocalization strong genetic evidence
of Frizzled and Vang/Stbm showing that the Fat/Dachs-
complexes and/or through ous and Frizzled/PCP
the shifting or moving of the systems act in parallel (Law-
cells as a whole. For rence et al., 2007). During fly
example, polarity could be larval stages, the Fat/Dachs-
achieved by rotation of the ous system is required to
cells toward the distal axis regulate the growth and
as happens with the rotation shape of the wing, the latter
of photoreceptor cell clusters (at least in part) by orienting
(ommatidia) in the Drosophila the axis of cell division
eye toward the anterior- perpendicular to the margin
posterior axis (Seifert and (Baena-López et al., 2005).
Mlodzik, 2007). But how Aigouy et al. (2010) show
would such a rotation be that high expression of
regulated? At early stages of Dachsous in the hinge region
pupal development, the is not required for its contrac-
proximal half of the wing tion, but correct Dachsous
epithelium (hinge) and the levels in the wing blade are
wing blade are similar in size Figure 1. Planar Cell Polarity in the Fly Wing required for the wing blade
(A) During fly pupal development, the initial axis of planar cell polarity (PCP) is
(Figure 1A). Subsequently, radial, that is, oriented toward the wing margin (black arrows).
to respond to the anisotropic
preceding and coinciding (B) As development proceeds, the hinge region (blue) contracts creating an aniso- mechanical stress that
with PCP realignment, the tropic mechanical stress on the wing blade, resulting in movements of wing cells orients cell elongation along
and realignment of PCP to the proximal-distal axis. Green arrows indicate the
hinge contracts and gener- the proximal-distal axis.
direction of cell movement, and red arrows show the direction of cellular rotations.
ates an anisotropic mechan- These processes take place between 14 and 24 hr after puparium formation. Moreover, cell polarity
ical stress on the blade, (C) The final orientation of PCP is in the proximal-distal axis in late pupal/adult fly defects correlate with the
which leads to its elongation wings (black arrows). inversion of local tissue rota-
in the proximal-distal axis tion in wild-type wings, with
(Figure 1B). Strikingly, epithelial cell elon- a proximal-distal axis within the wing (Fig- altered levels of Dachsous in the posterior
gation could drive the realignment of ure 1C). Interestingly, during this remodel- compartment, suggesting that Dachsous
cortical microtubules with the proximal- ing process, the global coordination/long- imbues cells with the ability to respond
distal axis, which appears essential for range coherence of PCP is diminished, coordinately to mechanical stress. Inter-
the delivery of Frizzled to the distal side which may be the reason why previous estingly, Harumoto et al. (2010) show
of cells (Shimada et al., 2006). New work studies of PCP during development have that Dachsous and Fat are required to
appearing in Developmental Cell by missed the early asymmetry/polarity of align cortical microtubules along the prox-
Uemura and colleagues (Harumoto et al., PCP core proteins. As pupal wing cells re- imal-distal axis and that Dachsous biases
2010) shows that, prior to hinge contrac- pack as they acquire a hexagonal shape, the direction of microtubule growth from
tion, cortical microtubules align perpen- the global coordination/long-range coher- high to low Dachsous levels, similar to its
dicular to the margin in the proximal ence of PCP increases again. This role in cell orientation. Whether the Fat/
region of the wing blade. This supports phenomenon is a consequence of the Dachsous system regulates cell remodel-
a general role for the orientation of cortical persistence of the Vang/Stbm and Frizzled ing by controlling the polarity of cortical
microtubules in PCP and in the realign- complexes at boundaries formed in the microtubules or vice versa remains to be
ment of PCP later in development. early stages of development and of the resolved.
Quantitative analyses of time-lapse proximal-distal alignment of new bound- In their elegant new study, Aigouy et al.
imaging of the pupal wing by Aigouy et al. aries. On the other hand, the Frizzled/ (2010) analyzed the timeline of events for
(2010) show that, in response to the aniso- PCP core factors are required for hexag- the establishment of PCP in the devel-
tropic stress, cells move with respect to onal cell packing, probably by polarizing oping Drosophila wing. They have
each other in a proximal direction and membrane trafficking along the proximal- provided evidence that the early Friz-
inwards with different velocities (Figure 1B). distal axis (Classen et al., 2005). Thus, zled/PCP core polarization toward the
This behavior causes shear and the local both early polarity and cellular packing wing margin (in a radial orientation) is real-
rotation of cells, mainly clockwise in the would feed in to one another to shape igned along the proximal-distal axis by
anterior and anticlockwise in the posterior and repolarize the epithelia. anisotropic mechanical stress and
half of the blade (Figure 1B). As a conse- It has been proposed that the Fat/ Dachsous-mediated tissue remodeling.
quence, PCP is reoriented from a radial to Dachsous system would provide ‘‘global These conclusions are consistent with,

Cell 142, September 3, 2010 ª2010 Elsevier Inc. 675


and supported by, the phenotypic PCP relation to new mechanisms that sculpt Lawrence, P.A., Struhl, G., and Casal, J. (2007).
features of Frizzled/PCP core group the shape of organs in general. Nat. Rev. Genet. 8, 555–563.
genes on one side and that of the Fat/
Dachsous system on the other. Flies REFERENCES Seifert, J.R., and Mlodzik, M. (2007). Nat. Rev.
Genet. 8, 126–138.
carrying mutations in Frizzled/PCP core
proteins exhibit defects in PCP Aigouy, B., Farhadifar, R., Staple, D.B., Sagner, A.,
Röper, J., Jülicher, F., and Eaton, S. (2010). Cell Shimada, Y., Yonemura, S., Ohkura, H., Strutt, D.,
throughout the wing. In contrast, the
142, this issue, 773–786. and Uemura, T. (2006). Dev. Cell 10, 209–222.
Fat/Dachsous system mainly affects
Baena-López, L.A., Baonza, A., and Garcı́a-Bel-
polarity in the proximal half of the wing, Strutt, D. (2003). Development 130, 4501–4513.
lido, A. (2005). Curr. Biol. 15, 1640–1644.
as this area strongly depends on cellular
Classen, A.K., Anderson, K.I., Marois, E., and Ea-
realignment and rotation during the ton, S. (2005). Dev. Cell 9, 805–817. Vinson, C.R., and Adler, P.N. (1987). Nature 329,
switch to the proximal-distal PCP axis. 549–551.
Harumoto, T., Ito, M., Shimada, Y., Kobayashi,
Together, these observations provide an T.J., Ueda, H.R., Lu, B., and Uemura, T. (2010).
exciting new framework for under- Dev. Cell 19. Published online September 2, Wu, J., and Mlodzik, M. (2009). Trends Cell Biol. 19,
standing the generation of PCP and its 2010. 10.1016/j.devcel.2010.08.004. 295–305.

Viable Rat-Mouse Chimeras:


Where Do We Go from Here?
Davor Solter1,*
1Institute
of Medical Biology, A*STAR, 138648 Singapore, Republic of Singapore
*Correspondence: davor.solter@imb.a-star.edu.sg
DOI 10.1016/j.cell.2010.08.021

In a tour-de-force study, Kobayashi et al. (2010) describe the first viable rat-mouse chimeras and
demonstrate that rat induced pluripotent stem (iPS) cells can rescue organ deficiency in mice.
Rat iPS cells formed a fully functional pancreas when injected into mouse blastocysts lacking the
Pdx1 gene required for pancreas formation.

Experimentally produced chimeras for the mouse and capitalizing on the the geep between a sheep (Ovis aries)
between different mouse strains (Tarkow- recent isolation of rat ES and iPS cells and a goat (Capra hircus) (Fehilly et al.,
ski, 1961) have been an exceedingly (Buehr et al., 2008; Li et al., 2008). Both 1984).
useful tool for developmental biologists, mouse and rat cells were tagged with To test the possibility that viable rat-
contributing to our understanding of the different fluorescent dyes, allowing the mouse chimeras could be formed,
establishment of cell lineages, cell deter- authors to follow their distribution in the Kobayashi et al. (2010) injected fluores-
mination, and the development of the developing chimeras. The authors wanted cently labeled mouse or rat iPS cells into
immune system and other organs. In this to prove, at least in principle, that xenoge- rat or mouse blastocysts, respectively,
issue of Cell, Kobayashi et al. (2010) neic organ complementation could be and returned them to blastocyst-compat-
dramatically extend the potential of mam- achieved, that is, that donor cells of one ible pseudopregnant females (that is,
malian chimeras with their report of viable species could rescue a defect in organ foster mothers of the same species as
rat-mouse chimeras that can develop to development in a recipient of a different the blastocysts). The authors then exam-
term and become fully functional adults. species. So, as a first step, they set out ined the resulting fetuses, newborns, and
In their study, Kobayashi and col- to produce viable chimeras between rats adults and found evidence of a substantial
leagues relied on previous knowledge and mice, even though many previous contribution of donor stem cells to tis-
but also added a few new wrinkles. They efforts to make such chimeras had failed. sues and organs of the host (Figure 1A).
first derived mouse and rat embryonic The only viable intergeneric chimera— Despite a big contribution of donor cells,
stem (ES) cells and induced pluripotent that is, a hybrid between animals from the size of newborn and adult chimeras
stem (iPS) cells using standard methods different genera—reported so far is (with one exception) was determined by

676 Cell 142, September 3, 2010 ª2010 Elsevier Inc.


Figure 1. Generating Rat-Mouse Chimeras
(A) Induced pluripotent stem (iPS) cells were derived from adult mouse and rat cells and were labeled with different fluorescent proteins. Rat (blue) and mouse
(red) iPS cells were injected into reciprocal blastocysts (mouse into rat and vice versa) to produce intergeneric chimeras. From these blastocysts, several
chimeras were born and some survived to adulthood. The contribution of injected donor stem cells was observed throughout the body of the host. The size
and morphology of the newborn and adult chimeras was determined by the host blastocyst.
(B) Fluorescently labeled rat iPS cells (blue) were injected into normal mouse blastocysts (left) or blastocysts lacking the Pdx1 gene (right), which encodes the
transcription factor pancreatic and duodenal homeobox 1 that is required for pancreas development. Chimeras derived from normal or Pdx1-deficient mouse
blastocysts showed an extensive contribution of rat cells to all tissues. However, in the Pdx1-deficient chimeras, the entire pancreas was derived from donor
rat cells (inset, blue) and was fully functional, including production of insulin by b islet cells.

the species of the host blastocyst. It is not produced by combining rat stem cells are injected into tetraploid blastocysts)
clear whether it is the embryo itself or the and mouse blastocysts, the resulting to produce rat-mouse chimeras (Nagy
uterine environment that determines the ‘‘mouse-like’’ chimeras had a gall bladder et al., 1993). Tetraploid blastocyst cells
extent of chimera growth. To distinguish despite the significant contribution of rat cannot participate in formation of the
between these possibilities, one would cells to abdominal organs. Reciprocal embryo proper; thus, the resulting fetus
have to transfer chimeric embryos into chimeras were ‘‘rat-like’’ and, again, (and adult) is derived entirely from the
the uterus of pseudopregnant females of despite a significant contribution from injected cells, whereas the placenta and
the same species as the donor stem cells mouse cells to abdominal organs, the extraembryonic membranes are derived
(not the blastocysts). Previous studies gall bladder was absent. These results from the tetraploid blastocyst. It remains
suggest that such experiments would fail suggest that cells of the blastocyst inner to be seen whether this approach could
because of the need for compatibility cell mass possess a ‘‘morphogenetic’’ produce a fetus derived entirely from
between the fetal part of the placenta and capacity that controls the behavior of mouse ES cells after their injection into
the uterus (Rossant et al., 1982). injected stem cells at all developmental a rat tetraploid blastocyst that then
Besides controlling the size and growth stages. develops in the uterus of a pseudopreg-
of the chimera, the host blastocyst seems This may explain why Kobayashi et al. nant rat female.
to impose additional morphogenetic reg- were able to successfully inject rat stem A major goal of the Kobayashi et al.
ulation. The postimplantation develop- cells into mouse blastocysts, whereas study was to determine whether stem cells
ment of normal rat and mouse embryos insertion of the rat inner cell mass into from a xenogeneic donor mammal could
is very similar, but there are differences the mouse blastocyst cavity did not result correct a genetic defect in a recipient
in organ morphogenesis. One of the in viable rat-mouse chimeras (Gardner mammal of a different species. So, in their
most noticeable differences is the pres- and Johnson, 1973). This notion could next set of experiments, the authors in-
ence of a gall bladder in mice and its be tested further using tetraploid comple- jected rat iPS cells into recipient mouse
absence in rats. In all adult chimeras mentation (that is, donor ES or iPS cells blastocysts that lacked the Pdx1 gene,

Cell 142, September 3, 2010 ª2010 Elsevier Inc. 677


which encodes a transcription factor technical challenges. For example, the potential of human stem cells. Yet such
(pancreatic and duodenal homeobox 1) mouse and rat are developmentally very experiments will be complicated, time
that is essential for development of the similar (apart from size), but it is not clear consuming, difficult to interpret, and, I
pancreas and formation of insulin- that chimeras between animals belonging suspect, will never become part of the
producing b islet cells. Although they to different phylogenetic families or orders standard protocols regulating the medical
observed a substantial contribution of rat would be viable. Indeed, the only attempts use of human stem cells. Although xeno-
cells to different organs and tissues, most to make such chimeras (between a mouse geneic organ complementation is unlikely
importantly, the pancreas of the rat-mouse and a bank vole) have failed (Mystkowska, to be a viable strategy for regenerative
chimeras was composed exclusively of rat 1975). In this experiment, the mouse-bank medicine, the elegant work of Kobayashi
cells (Figure 1B). Thus, cells derived from vole chimeras were made by aggregation et al. is a boon for researchers seeking
rat iPS cells were able to completely of embryos; it is possible that injection of to better understand the biology of stem
rescue the genetic deficiency of the host bank vole stem cells into mouse blasto- cells and mammalian development.
mouse blastocyst. These rat-mouse cysts, followed by their development
chimeras developed into adult animals in the uterus of mouse foster mothers
with a normal functional pancreas, demon- might yield positive results. Successful REFERENCES
strating that xenogeneic organ comple- chimerism between members of different
mentation is achievable. This is a remark- orders (the pig and human, for example) Buehr, M., Meek, S., Blair, K., Yang, J., Ure, J.,
Silva, J., McLay, R., Hall, J., Ying, Q.-L., and Smith,
able accomplishment. seems very unlikely, and attempts to
A. (2008). Cell 135, 1287–1298.
So where do we go from here? Although produce early postimplantation human-
human ES and iPS cells offer hope for mouse chimeras have not been encour- Fehilly, C.B., Willadsen, S.M., and Tucker, E.M.
(1984). Nature 307, 634–636.
tissue and cell replacement therapies in aging (James et al., 2006). Even if we
the not too distant future, the replacement succeed in developing organ-deficient Gardner, R.L., and Johnson, M.H. (1973). Nat. New
of complex organs—lung, kidney, liver, pigs by genetic manipulation and pro- Biol. 246, 86–89.

gut, and, of course, pancreas—is likely to ducing pig-human chimeras in which the James, D., Noggle, S.A., Swigut, T., and Brivanlou,
be much more difficult. Several strategies parenchymal cells of the specific organ A.H. (2006). Dev. Biol. 295, 90–102.
for organ replacement are being tested. are entirely derived from human cells, Kobayashi, T., Yamaguchi, T., Hamanaka, S.,
These include the growth of organs immune rejection will still be a problem Kato-Itoh, M., Yamazaki, Y., Ibata, M., Sato, H.,
in vitro with mixtures of different stem cells because the human organ carried by the Lee, Y.-S.,, Usui, J.-i., Knisely, A.S., et al. (2010).
Cell 142, this issue, 787–799.
and biocompatible scaffolds or the gener- pig will contain pig-derived stromal cells
ation of ‘‘humanized’’ pigs engineered to and blood vessels. Li, P., Tong, C., Mehrian-Shai, R., Jia, L., Wu, N.,
lack certain antigens so that their organs Finally, there are huge legal and ethi- Yan, Y., Maxson, R.E., Schulze, E.N., Song, H.,
Hsieh, C.-L., et al. (2008). Cell 135, 1299–1310.
can be used for transplantation in human cal barriers to creating human-animal
patients with a reduced chance of immune chimeras and, indeed, their production is Mystkowska, E.T. (1975). J. Embryol. Exp. Mor-
rejection. Could production of human forbidden in most countries. However, it phol. 33, 731–744.

organs in, for example, human-pig is possible that injecting human ES or Nagy, A., Rossant, J., Nagy, R., Abramow-
chimeras be an alternative approach? iPS cells into a mouse blastocyst and al- Newerly, W., and Roder, J.C. (1993). Proc. Natl.
Acad. Sci. USA 90, 8424–8428.
Although production of viable rat-mouse lowing limited (early postimplantation)
chimeras could be viewed as a first step development of human-mouse chimeras Rossant, J., Mauro, V.M., and Croy, B.A. (1982).
in this direction, as Kobayashi et al. would be approved for the specific J. Embryol. Exp. Morphol. 69, 141–149.
propose, there are huge biological and purpose of testing the differentiation Tarkowski, A.K. (1961). Nature 190, 857–860.

678 Cell 142, September 3, 2010 ª2010 Elsevier Inc.


Leading Edge
Previews

‘Fore Brain:
A Hint of the Ancestral Cortex
Lora B. Sweeney1,2,3 and Liqun Luo1,2,3,*
1Neurosciences Program
2Department of Biology
3Howard Hughes Medical Institute

Stanford University, Stanford, CA 94305, USA


*Correspondence: lluo@stanford.edu
DOI 10.1016/j.cell.2010.08.024

By combining gene expression profiling with image registration, Tomer et al. (2010) find that the
mushroom body of the segmented worm Platynereis dumerilii shares many features with the
mammalian cerebral cortex. The authors propose that the mushroom body and cortex evolved
from the same structure in the common ancestor of vertebrates and invertebrates.

The mammalian cerebral cortex underlies identify structures in the developing brain tion factors such as Bf1 (brain factor 1,
many higher-order processes, such as of Platynereis that are possibly related also known as Foxg1) and Pax 6 (paired
perception, memory, language, and ad- to the vertebrate pallium, Tomer et al. box gene 6) (Hébert and Fishell, 2008).
vanced motor skills. With its intricate characterized the expression patterns of With this technique, Tomer et al. iden-
furrows and ridges (i.e., the sulci and many genes at different stages of neural tify a structure in Platynereis with gene
gyri), the complexity of the cerebral cortex development in the worm. The standard patterns that mirror those observed in
is evident even at its surface. Beneath the methods for characterizing gene expres- the vertebrate pallium. The authors then
surface, the cerebral cortex is separated sion are in situ hybridizations and immuno- follow this structure during the develop-
into layers of densely packed neurons staining with antibodies. However, these ment of Platynereis and find that its
with axons reaching deep into the white techniques are generally restricted to neurons develop into the mushroom
matter. These layers are divided further one or two genes at a time and thus are body, a brain region in insects and worms
into functional regions that correspond to unable to provide direct comparisons of involved in sensory processing and
the body plan. How does such a complex expression patterns for a large number of memory (Figure 1). Moreover, this embry-
structure develop such precise organiza- genes. onic structure in the worm gives rise
tion? Many researchers have approached To overcome this technical hurdle, to similar types of neurons as found
this question from a developmental Tomer et al. use advanced image registra- in the pallium (e.g., glutamatergic and
perspective by identifying and perturbing tion methods (including linear transforma- GABAergic neurons).
molecular components that contribute tion and nonlinear warping), in which Similarities between the mammalian
to the structure of the cerebral cortex multiple microscopy images are aligned cortex and the invertebrate mushroom
(Hébert and Fishell, 2008). Alternatively, to one coordinate system, standardized body have been noted previously, but
one can take an evolutionary approach to one size, and smoothed to correct arti- such clear parallels in their gene pattern-
and search for an ancestral precursor to facts due to stretches during sample ing have not been observed (Strausfeld
the cortex. In this issue of Cell, Tomer preparation (Rueckert et al., 1999; Rohlf- et al., 1998; Farris, 2008). Why not? Mush-
et al. (2010) follow the latter approach ing et al., 2001; Kurylas et al., 2008). room bodies are well characterized in in-
and identify a brain region of the seg- Specifically, Tomer et al. use the axon sects, especially in the fruit fly Drosophila,
mented worm Platynereis dumerilii (an scaffold (i.e., bundles of axons in the and choosing to study an annelid was
annelid) called the mushroom body that developing brain) as a landmark to align critical to making this new connection.
shares the same ‘‘molecular fingerprint’’ individual gene expression patterns from Based on the arrangement of the axon
as the developing mammalian cortex. multiple worms onto a standard brain fibers and the function of its neurons, the
The cerebral cortex derives from the template. This allows the simultaneous mushroom body of insects has been
pallium. From the Latin for ‘‘cloak,’’ mapping of expression profiles for an loosely compared to the mammalian
pallium refers to the outer layer of the unlimited number of genes during neural cerebral cortex, the hippocampus, or the
brain. The mammalian palium consists of development. In this manner, the authors cerebellum (Strausfeld et al., 1998).
the cerebral cortex, olfactory cortex, and define the spatial relationship for the However, insects are fast-evolving inver-
the hippocampus. The evolutionary origin expression of more than fifteen genes in tebrates and thus may deviate quickly
of the vertebrate pallium has fascinated Platynereis that were previously shown from their ancestors. By contrast, anne-
biologists for centuries because our high- to regulate the patterning of the cortex in lids evolve slowly and therefore make
est mental functions originate from it. To mammals. These include many transcrip- excellent organisms for comparing the

Cell 142, September 3, 2010 ª2010 Elsevier Inc. 679


the vertebrate pallium and invertebrate
mushroom body evolved from the same
structure in the last common ancestor
of these organisms (Figure 1). Moreover,
this structural precursor may also have
processed sensory information, as the
cortex and mushroom body do.
That said, however, the vertebrate
pallium and invertebrate mushroom body
are considerably different. First, they have
different shapes (Figure 1). This may result
in part from considerable differences
in neural developmental programs in
vertebrates and invertebrates. Second,
it is unclear whether the mushroom body
exhibits the same type of layering and
functional specialization as the cortex.
Third, gene expression patterns appear
to remain constant over time during
development of the annelid mushroom
body, but the patterns are quite dynamic
during the development of the mamma-
lian cortex. This temporal variation com-
plicates the interpretation of the homolo-
Figure 1. A Common Origin for the Mushroom Body and Cerebral Cortex? gous gene expression patterns detected
In mammals, the pallium develops into the cerebral cortex, olfactory cortex, and hippocampus. Gene for the two structures. Finally, the expres-
expression profiling combined with image registration in the developing brain of the segmented worm
Platynereis dumerilii identified a brain region with gene expression patterns similar to that of the mouse sion profiles are not identical for the
cortex (Tomer et al., 2010). This region in the worm develops into the mushroom body, which is involved two structures. For example, expression
in sensory processing and memory formation in insects and likely worms. These results suggest that the of Wnt3a was present in the vertebrate
mushroom body and the cerebral cortex evolved from the same structure in a common ancestor of verte-
brates and annelids 600 million years ago (mya). The estimates of divergence follow Peterson et al.
pallium but not in the developing
(2004). mushroom body. Such differences are
probably not surprising, given that verte-
evolution of invertebrates with that of ing olfactory inputs and for memory brates and annelids diverged from a
vertebrates. For example, an analysis formation (Heisenberg, 2003). Indeed, common precursor 600 million years
of intron abundance across the entire Tomer and colleagues found that a subset ago (Figure 1).
genomes of vertebrates, annelids, and of genes that share similar expression In summary, the data presented by
insects found that two-thirds of the patterns in the annelid mushroom body Tomer and colleagues provide a tanta-
human introns were also present in and the mammalian cortex are also lizing hint that one of the highest-order
annelids but were largely absent from expressed in the Drosophila mushroom processing centers of the human brain
insects, confirming that insects have body. shares an evolutionary origin with the
evolved faster than annelids (Raible Two distinct mechanisms can explain mushroom body of worms and insects.
et al., 2005). Moreover, annelids are ex- the molecular similarities of the inverte- In principle, the techniques presented
tremely amenable to experimental anal- brate mushroom body and the vertebrate by the authors for profiling expression
ysis because they are easy to breed and cerebral cortex. They could arise inde- patterns of numerous genes simulta-
observe in vivo. Further, they develop pendently through convergent evolution, neously can be used in other organisms
with highly stereotyped structures that or they could evolve from the same to garner further support for this hypoth-
vary little between individuals. structure in a common ancestor. Tomer esis. Furthermore, comparative struc-
Given the similarity in the gene expres- and colleagues present a statistical anal- ture-function analyses may also provide
sion patterns of the developing annelid ysis indicating that the similarities in additional insights into the function of
mushroom body and the mammalian gene expression that they observed are this ancestral structure, including how its
cerebral cortex, do these structures unlikely to occur by convergent evolution. form has adapted over the last 600 million
share similar functions? The cerebral Although these arguments suffer from years.
cortex transforms sensory information the caveat that expression patterns of
into action. The mushroom body of anne- different genes may not have evolved REFERENCES
lids also processes sensory information, independently, the number of genes
receiving input from chemosensory cells studied by Tomer and colleagues do Farris, S.M. (2008). Brain Behav. Evol. 72, 106–122.
on their mouthparts. In insects, the analo- indeed favor the possibility of a common Hébert, J.M., and Fishell, G. (2008). Nat. Rev.
gous structure is best known for process- origin. Together, their data suggest that Neurosci. 9, 678–685.

680 Cell 142, September 3, 2010 ª2010 Elsevier Inc.


Heisenberg, M. (2003). Nat. Rev. Neurosci. 4, Raible, F., Tessmar-Raible, K., Osoegawa, K., Rueckert, D., Sonoda, L.I., Hayes, C., Hill, D.L.,
266–275. Wincker, P., Jubin, C., Balavoine, G., Ferrier, D., Leach, M.O., and Hawkes, D.J. (1999). IEEE Trans.
Benes, V., de Jong, P., Weissenbach, J., et al. Med. Imaging 18, 712–721.
Kurylas, A.E., Rohlfing, T., Krofczik, S., Jenett, A., (2005). Science 310, 1325–1326.
and Homberg, U. (2008). Cell Tissue Res. 333,
Rohlfing, T., Brandt, R., Maurer, C.R., Jr., and Strausfeld, N.J., Hansen, L., Li, Y., Gomez, R.S.,
125–145.
Menzel, R. (2001). IEEE Workshop on Mathemat- and Ito, K. (1998). Learn. Mem. 5, 11–37.
Peterson, K.J., Lyons, J.B., Nowak, K.S., Takacs, ical Methods in Biomedical Image Analysis,
C.M., Wargo, M.J., and McPeek, M.A. (2004). L. Staib, ed. Kauai, HI, IEEE Computer Society, Tomer, R., Denes, A.S., Tessmar-Raible, K., and
Proc. Natl. Acad. Sci. USA 101, 6536–6541. Los Alamitos, CA, pp. 187–194. Arendt, D. (2010). Cell 142, this issue, 800–809.

Cell 142, September 3, 2010 ª2010 Elsevier Inc. 681


Leading Edge
Minireview

The Language of Histone Crosstalk


Jung-Shin Lee,1 Edwin Smith,1 and Ali Shilatifard1,*
1Stowers Institute for Medical Research, 1000 East 50th Street, Kansas City, MO 64110, USA

*Correspondence: ash@stowers.org
DOI 10.1016/j.cell.2010.08.011

It has been suggested that a specific pattern of histone posttranslational modifications and their
crosstalk may constitute a code that determines transcriptional outcomes. However, recent studies
indicate that histone modifications have context-dependent effects, making their interplay more
like a language within the chromatin signaling pathway than a code.

For almost two decades, a primary focus in the field of transcrip- phosphorylation of serine 10 on histone H3 stimulates the ability
tional regulation was to identify DNA elements that control the of Gcn5 to acetylate histone H3 at lysine 14 (H3K14) (Cheung
expression of genes. These efforts were in part motivated by the et al., 2000; Lo et al., 2000) (Figure 1A). Another well-character-
expectation that it would one day be possible to look at a gene ized example is the requirement of histone H2B monoubiquitina-
and its regulatory sequences and predict when and where tion for proper H3K4 methylation by the H3K4 methylase
a gene was going to be transcribed. We then learned that along complex COMPASS (Figure 1B) (Shilatifard, 2006). This process,
with the sequence-specific binding of activators and repressors, initially discovered in yeast (Shilatifard, 2006), is now known to
there is an additional world of factors that modify, interact, and be highly conserved among eukaryotes (Kim et al., 2009). Addi-
remodel chromatin to regulate gene expression. The identification tionally, a recent comprehensive mutation analysis of all histone
of a multitude of histone modifications—some correlated with residues reveals that a single point mutation in histone H3K14,
activation, some with repression—led to the proposal that the a site of acetylation, results in the specific loss of H3K4 trimethy-
modifications constitute a code that could be recognized by tran- lation, but not mono- or dimethylation (Nakanishi et al., 2008).
scription factors to determine the transcriptional state of a gene. This screen also demonstrated that H3K4 trimethylation is regu-
However, additional research has since added layers of lated by both monoubiquitination-dependent and monoubiquiti-
complexity, revealing a nuanced and intriguing language, not nation-independent processes (Nakanishi et al., 2008).
a strict code, as the basis for transcriptional regulation through Given that histone-modifying enzymes are often found in mul-
the chromatin signaling pathway. Here, we review the complex tisubunit complexes, modification of nearby residues can create
crosstalk among histone modifications, including recent studies binding sites for the components of the complex helping to
that illustrate how the context and timing of these modifications anchor an enzyme to a nucleosome. For example, the PHD finger
are critical for a particular transcriptional readout. of Yng1, a subunit of the NuA3 histone acetyltransferase
complex, recognizes methylated H3K4 and helps recruit this
Histone Crosstalk and Gene Activity histone acetyltransferase complex for acetylation of H3K14
In eukaryotic cells, gene expression can be regulated at the level (Martin et al., 2006; Taverna et al., 2006). The Yng1-related
of chromatin structure. Numerous residues within the histone ING2 can also bind methylated H3K4; however, it is present in
tails and several residues within the histone globular domains a histone deacetylase complex (Shi et al., 2006). Therefore,
can be modified in a variety of ways, including acetylation, H3K4 methylation can serve as a landing platform for a variety
phosphorylation, ubiquitination, and methylation. A well-charac- of histone-modifying enzymes with opposing activities.
terized posttranslational modification regulating chromatin Modifications of nearby residues can also prevent the recog-
structure is the acetylation of histone N-terminal tails, which is nition of a substrate by an enzyme, as recently reported to occur
thought to facilitate transcriptional activation either by charge when methylation of histone H3 arginine 2 (H3R2) interferes with
neutralization of the tails’ interaction with DNA or by forming H3K4 methylation by Set1/COMPASS in yeast and COMPASS-
a binding site for bromodomain-containing transcription factors, like complexes in mammalian cells (Guccione et al., 2007;
some of which can remodel nucleosomes. Another well-studied Kirmizis et al., 2007) (Figure 1B). Histone modifications can
histone modification is the methylation of lysine 4 of histone H3 also prevent the recruitment of factors other than enzymes.
(H3K4), a modification generally associated with transcriptionally For example, heterochromatin protein 1 (HP1), which binds
active genes and a binding site for a variety of factors that include methylated H3K9, cannot do so when the adjacent serine 10
histone-modifying and -remodeling activities (Shilatifard, 2006). (H3S10) is phosphorylated during mitosis or during gene activa-
More complex scenarios arise when histone modifications act tion (Fischle et al., 2005; Mateescu et al., 2008).
combinatorially in a context-dependent manner to facilitate or Multiple types of histone crosstalk, involving numerous histone-
repress chromatin-mediated transcription. In some cases the modifying complexes, can occur at any one gene. A major chal-
modification of one residue can alter the ability of a second lenge is to understand the events that regulate changes in gene
residue to be implemented by its modifying enzyme(s) (Figure 1). expression through these modifications/crosstalk. One strategy
The first example of histone crosstalk falls into this category: the has been to profile histone modifications genomewide, with the

682 Cell 142, September 3, 2010 ª2010 Elsevier Inc.


a gene activated in response to serum (Zippo et al., 2009) (Fig-
ure 2A). They present evidence for a transcription activation
pathway in which the phosphorylation of H3 tails leads to the
acetylation of H4 tails. In turn, acetylation of H4 tails is required
for the recruitment of the RNA Pol II positive transcription elonga-
tion factor, P-TEFb (Figure 2A). Previously, the authors found
that activation of the FOSL1 gene requires PIM1, a proto-onco-
gene whose kinase activity is activated through MAP kinase
signaling. Numerous cellular substrates of PIM1 have been iden-
tified, including H3S10. Other H3S10 kinases, such as MSK1 and
MSK2 (MSK1/2), are also implicated in the phosphorylation of
histones at serum-responsive genes, including FOSL1. Zippo
and colleagues find that the spatiotemporal pattern of H3S10
Figure 1. Examples of Histone Crosstalk phosphorylation differs for PIM1 and MSK1/2. MSK1/2 mediates
(A) The first characterized example of histone crosstalk is the stimulation of
the phosphorylation of H3S10 at the promoter of FOSL1 at early
acetyltransferase activity of GCN5 toward the histone H3 tail by prior phos-
phorylation (P) of serine 10. Acetylation, Ac. time points of gene expression, whereas PIM1 is required for
(B) Crosstalk among histone modifications can span more than one histone. H3S10 phosphorylation at a FOSL1 enhancer after the MSK1/
Monoubiquitination of histone H2B on lysine 120 of the C-terminal helix can 2-mediated phosphorylation of H3S10 (Figure 2A).
lead to the trimethylation of lysine 4 in the histone 3 tail (H3K4) by Set1/
COMPASS. However, H3K4 methylation by COMPASS and COMPASS-like Screening for other histone modifications specifically associ-
complexes can be blocked if the nearby arginine of H3 is already methylated. ated with the FOSL1 enhancer shows that the acetylation of
H4K16 coincides with H3S10 phosphorylation. RNA interfer-
expectation that a given pattern will indicate a transcriptional ence-mediated knockdown of PIM1 results in loss of H4K16
outcome due to the recruitment of specific proteins by these modi- acetylation, suggesting a trans-tail crosstalk from H3S10 phos-
fications. However, some recent examples of trans-histone cross- phorylation to H4K16 acetylation. Zippo and colleagues asked
talk illustrate that transcriptional readout depends on context and whether 14-3-3 g, 3, and z proteins, previously shown to bind
timing by which these modifications are introduced. Simply put, phosphorylated H3S10, are recruited to the promoter and the
just looking at the pattern of chromatin modifications at a locus enhancer of FOSL1 in response to serum. They find that 14-3-
is not sufficient to determine its gene expression status. These 33 and 14-3-3z are recruited to both the promoter and enhancer
studies provide new insight into the language of histone crosstalk. of FOSL1 after serum stimulation. However, 14-3-3 is required
for recruiting the H4K16 histone acetyltransferase MOF only to
From Histone Phosphorylation to Transcription the enhancer and not to the promoter of FOSL1. Recruitment
Elongation of MOF to the enhancer results in H4K16 acetylation, which
A novel form of crosstalk was recently discovered by Zippo and can be bound by the bromodomain-containing protein, Brd4.
colleagues, who studied the transcriptional control of FOSL1, Brd4 associates with P-TEFb, a kinase that phosphorylates Pol

Figure 2. Context-Dependent Outcomes of


Histone Crosstalk
(A) Zippo and colleagues (Zippo et al., 2009)
uncover a new form of histone crosstalk by
studying the transcriptional control of FOSL1,
a gene activated in response to serum. Activation
requires the binding of PIM1 to the FOSL1
enhancer. PIM1 is a kinase responsible for phos-
phorylation (P) of serine 10 on the histone H3 tail
(H3S10). Phosphorylated H3S10 creates a binding
site for 14-3-3, a phosphoserine binding protein.
Acetylation (Ac) of lysine 16 on the H4 (H4K16)
tail occurs through interaction of 14-3-3 with the
histone acetyltransferase MOF. Acetylated
H4K16 can in turn form a binding site for the bro-
modomain-containing protein Brd4, a component
of P-TEFb, a kinase that phosphorylates the
C-terminal domain of RNA Pol II to facilitate tran-
scription elongation. However, at an earlier stage
of serum stimulation, an MSK1/2 kinase is re-
cruited to the promoter where it phosphorylates
H3S10. 14-3-3 is then recruited to the promoter,
but unlike the situation at the enhancer, MOF is
not recruited to the promoter. Thus, the timing,
location, and perhaps identity of the H3 kinase, and not the H3S10 modification alone, determines downstream events.
(B) Another example of how the order of implementation of histone modifications can affect transcription comes from work from Wang et al. (2009). They report
that despite correlations between histone acetylation and H3K4 methylation, artificially increasing acetylation through treatment of cells with deacetylase inhib-
itors (HDACs) does not lead to productive transcription, despite the presence of H3K4 methylation and Pol II recruitment. Therefore, patterns of histone modi-
fications cannot simply be ‘‘read’’ but instead have distinct effects depending on the cellular context and upstream signaling events.

Cell 142, September 3, 2010 ª2010 Elsevier Inc. 683


II to facilitate transcription elongation (Figure 2A). Thus, Zippo with H3K4 methylation rarely show this increase in acetylation
and colleagues propose that crosstalk between modifications in response to deacetylase inhibitors.
on two different histone tails regulate productive transcription In order to determine whether H3K4 methylation is functionally
elongation through the sequential recruitment of proteins that linked to H4K16 acetylation, Wang and colleagues use RNA
bind these modifications. interference-mediated knockdown of WDR5, a common compo-
One question raised by this study is why H3S10 phosphoryla- nent of the Set1 and MLL (mixed-lineage leukemia) COMPASS-
tion produces different results at the enhancer than at the like H3K4 methyltransferase complexes. Upon knockdown of
promoter of FOSL1 even though 14-3-3 is recruited to both sites. WDR5, they observe reduced levels of histone acetylation at
At the enhancer, 14-3-3 recruits the histone acetyltransferase the subset of transcriptionally quiescent genes marked by
MOF. At the promoter, it does not. What is the difference H3K4 methylation. Based on this information, Wang and
between 14-3-3 at the promoter and at the enhancer? Interest- colleagues suggest that H3K4 methylation primes certain genes
ingly, 14-3-33 and 14-3-3z are thought to be regulated via lysine for an increase in H3K9 and H4K16 acetylation. Interestingly,
acetylation (Choudhary et al., 2009) and an acetyltransferase, what was not considered by Wang and colleagues is the fact
Tip60, is preferentially recruited to the promoter of FOSL1. One that WDR5 is also a component of complexes that can acetylate
possibility is that Tip60 acetylates 14-3-3 and prevents its inter- H4K16 or H3K9. WDR5 associates with the H4K16 acetyltrans-
action with MOF. ferase MOF in the NSL/MSL1v complex (Cai et al., 2010; Li
Another intriguing aspect of the study by Zippo and colleagues et al., 2009) as well as with the H3K9/K14 acetyltransferase
is the link between H3S10 phosphorylation and H4K16 acetyla- Gcn5 in the ATAC complex (Suganuma et al., 2008; Wang
tion. These two modifications were previously linked in studies of et al., 2008). As such, the effect of WDR5 knockdown could be
dosage compensation in the fruit fly Drosophila. In Drosophila a consequence of WDR5’s role as a subunit of the H3K4 methyl-
dosage compensation, MOF is recruited to the coding region ases, or WDR5’s role as a subunit of the H3 and H4 acetyltrans-
of X-linked genes in males where it mediates H4K16 acetylation ferase complexes, or a combination of the two. Given that WDR5
in a process thought to facilitate transcription elongation. Coloc- is part of the H3 and H4 acetyltransferase complexes, the exis-
alizing with MOF on the male X chromosome is the JIL-1 kinase, tence of crosstalk between H3K4 and H4K16 needs to be further
an MSK1/2-related kinase that mediates the phosphorylation of characterized.
H3S10 on this chromosome. In the case of Drosophila dosage One surprising finding of the study by Wang and colleagues is
compensation, recruitment of the JIL-1 kinase to the male X that transcription is rarely induced at the genes tested, although
chromosome occurs later than H4K16 acetylation (Wang et al., Pol II is recruited following treatment with a deacetylase inhibitor
2001), a reversal of the order of the addition of these marks at (Figure 2B). Thus, Pol II recruitment does not lead to the antici-
FOSL1 in response to serum. Concordantly, the MOF complex pated increase in transcription, despite the fact that H3K9 and
that mediates acetylation in coding regions is likely to be distinct H4K16 acetylation are increased. These modifications coincide
from the MOF complex that mediates promoter/enhancer acet- with transcriptional activation at FOSL1 upon serum treatment
ylation of genes (Li et al., 2009). Thus, by all appearances, these (Zippo et al., 2009). Together with the studies of H3S10 phos-
two examples of the coexistence of both H3S10 phosphorylation phorylation and H4K16 acetylation at the FOSL1 enhancer, it is
and H4K16 acetylation are unrelated in their order of implemen- clear that knowing the mechanism and timing of these modifica-
tation and in their biological meaning. This suggests that tions is necessary for determining the transcriptional outcome.
descriptions of histone modification patterns, without under-
standing the mechanisms leading to the implementation of these The Emerging Grammar of Histone Crosstalk
marks, should be interpreted with caution. Importantly, the study The existence of a histone modification code was proposed
by Zippo and colleagues begins to determine the role of histone 10 years ago as a way to approach the study of the quickly
modifications in the activation of FOSL1, with a spatial and growing number of histone modifications involved in the regula-
temporal dissection of how a cascade of histone modifications tion of gene expression and other DNA-templated processes,
can lead to a particular transcriptional outcome. such as replication, repair, and recombination. New ‘‘words’’ of
histone modifications are being discovered, and they continue
to appear in interesting combinations. However, discovering
Trimethylation Converses with Acetylation the exact roles these modifications play in gene expression
Another example of trans-tail crosstalk was proposed by Wang has been complicated by finding a counterexample for almost
et al. (2009). In this case the communication takes place between every example of crosstalk, such as the case of H3K4 and
the H3 and H4 tails and, like the example provided by Zippo et al. H3K36 methylation recruiting both histone acetyltransferases
(2009), involves H4K16 acetylation and FOSL1 transcription. By and deacetylases.
analyzing genomewide profiles of several histone acetyltrans- A common theme of recent research on histone crosstalk is
ferases, deacetylases, and modifications, these investigators that the order and mechanism of the addition and removal of
find a link between H3K4 methylation and H4K16 acetylation at histone modifications are important for the transcriptional
some inducible genes, including FOSL1. The authors show readout of a gene. The recent examples of histone crosstalk
that a subset of transcriptionally quiescent genes, marked by that we have addressed here illustrate this point. In one study,
the presence of H3K4 methylation, display a marked increase the implementation of H3S10 phosphorylation at two different
in histone acetylation at H3K9 and H4K16 after addition of de- locations, by two different enzymes, and at two different times
acetylase inhibitors. In contrast, quiescent genes not marked after serum stimulation had disparate effects on subsequent

684 Cell 142, September 3, 2010 ª2010 Elsevier Inc.


histone acetylation at the respective locations (Zippo et al., 2009) Choudhary, C., Kumar, C., Gnad, F., Nielsen, M.L., Rehman, M., Walther, T.C.,
(Figure 2A). Zippo and colleagues were able to propose a mech- Olsen, J.V., and Mann, M. (2009). Science 325, 834–840.
anism of gene activation by identifying the histone-modifying Fischle, W., Tseng, B.S., Dormann, H.L., Ueberheide, B.M., Garcia, B.A., Sha-
enzymes, the histone modifications, and a set of proteins that banowitz, J., Hunt, D.F., Funabiki, H., and Allis, C.D. (2005). Nature 438, 1116–
1122.
recognized these modifications on the FOSL1 gene after serum
stimulation. In another study, Wang and colleagues found Guccione, E., Bassi, C., Casadio, F., Martinato, F., Cesaroni, M., Schuchlautz,
H., Luscher, B., and Amati, B. (2007). Nature 449, 933–937.
that artificially recreating histone modifications that correlate
with gene expression could result in the recruitment of RNA Kim, J., Guermah, M., McGinty, R.K., Lee, J.S., Tang, Z., Milne, T.A., Shilati-
fard, A., Muir, T.W., and Roeder, R.G. (2009). Cell 137, 459–471.
Pol II, but this was not sufficient for transcription (Figure 2B).
Thus, simply mapping histone modification patterns without Kirmizis, A., Santos-Rosa, H., Penkett, C.J., Singer, M.A., Vermeulen, M.,
Mann, M., Bahler, J., Green, R.D., and Kouzarides, T. (2007). Nature 449,
understanding the recruitment, regulation, and interactions of
928–932.
the complexes implementing these marks is not sufficient
Li, X., Wu, L., Corsa, C.A., Kunkel, S., and Dou, Y. (2009). Mol. Cell 36,
to understand the mechanisms regulating gene expression.
290–301.
Genomewide profiling techniques have now become widely
Lo, W.S., Trievel, R.C., Rojas, J.R., Duggan, L., Hsu, J.Y., Allis, C.D., Marmor-
adopted, providing the ability to map histone modifications,
stein, R., and Berger, S.L. (2000). Mol. Cell 5, 917–926.
the enzymes implementing these marks, and the factors that
Martin, D.G., Baetz, K., Shi, X., Walter, K.L., MacDonald, V.E., Wlodarski, M.J.,
recruit them under different experimental conditions.
Gozani, O., Hieter, P., and Howe, L. (2006). Mol. Cell. Biol. 26, 7871–7879.
The study of the regulation of gene expression has grown from
Mateescu, B., Bourachot, B., Rachez, C., Ogryzko, V., and Muchardt, C.
identifying transcription factors and their binding sites to include
(2008). EMBO Rep. 9, 267–272.
a wide variety of other binding events associated with the modi-
Nakanishi, S., Sanderson, B.W., Delventhal, K.M., Bradford, W.D., Staehling-
fications of histones that package the DNA. Future progress will
Hampton, K., and Shilatifard, A. (2008). Nat. Struct. Mol. Biol. 15, 881–888.
require us to learn much more about how the words comprising
Shi, X., Hong, T., Walter, K.L., Ewalt, M., Michishita, E., Hung, T., Carney, D.,
the dictionary of histone crosstalk are used in a particular order Pena, P., Lan, F., Kaadige, M.R., et al. (2006). Nature 442, 96–99.
to provide the grammar of this complex language.
Shilatifard, A. (2006). Annu. Rev. Biochem. 75, 243–269.
ACKNOWLEDGMENTS Suganuma, T., Gutierrez, J.L., Li, B., Florens, L., Swanson, S.K., Washburn,
M.P., Abmayr, S.M., and Workman, J.L. (2008). Nat. Struct. Mol. Biol. 15,
We thank Dr. E. Park for the critical reading of the manuscript. We also thank 364–372.
L. Shilatifard for editorial assistance. Studies in the Shilatifard laboratory are Taverna, S.D., Ilin, S., Rogers, R.S., Tanny, J.C., Lavender, H., Li, H., Baker, L.,
supported in part by grants from the NIH: R01CA89455, R01GM069905, and Boyle, J., Blair, L.P., Chait, B.T., et al. (2006). Mol. Cell 24, 785–796.
R01CA150265.
Wang, Y., Zhang, W., Jin, Y., Johansen, J., and Johansen, K.M. (2001). Cell
105, 433–443.
REFERENCES
Wang, Y.L., Faiola, F., Xu, M., Pan, S., and Martinez, E. (2008). J. Biol. Chem.
283, 33808–33815.
Cai, Y., Jin, J., Swanson, S.K., Cole, M.D., Choi, S.H., Florens, L., Washburn,
M.P., Conaway, J.W., and Conaway, R.C. (2010). J. Biol. Chem. 285, 4268– Wang, Z., Zang, C., Cui, K., Schones, D.E., Barski, A., Peng, W., and Zhao, K.
4272. (2009). Cell 138, 1019–1031.
Cheung, P., Tanner, K.G., Cheung, W.L., Sassone-Corsi, P., Denu, J.M., and Zippo, A., Serafini, R., Rocchigiani, M., Pennacchini, S., Krepelova, A., and
Allis, C.D. (2000). Mol. Cell 5, 905–915. Oliviero, S. (2009). Cell 138, 1122–1136.

Cell 142, September 3, 2010 ª2010 Elsevier Inc. 685


GPR120 Is an Omega-3 Fatty Acid Receptor
Mediating Potent Anti-inflammatory
and Insulin-Sensitizing Effects
Da Young Oh,1,4 Saswata Talukdar,1,4 Eun Ju Bae,1 Takeshi Imamura,2 Hidetaka Morinaga,1 WuQiang Fan,1 Pingping Li,1
Wendell J. Lu,1 Steven M. Watkins,3 and Jerrold M. Olefsky1,*
1Department of Medicine, Division of Endocrinology and Metabolism, University of California, San Diego, La Jolla, CA 92093, USA
2Divisionof Pharmacology, Shiga University of Medical Science, Tsukinowa, Seta, Otsu-city, Shiga, 520-2192 Japan
3Tethys Bioscience, 3410 Industrial Boulevard, Suite 103, West Sacramento, CA 95691, USA
4These authors contributed equally to this work

*Correspondence: jolefsky@ucsd.edu
DOI 10.1016/j.cell.2010.07.041

SUMMARY and anti-inflammatory molecules within the specialized popula-


tion of proinflammatory tissue macrophages.
Omega-3 fatty acids (u-3 FAs), DHA and EPA, exert G protein-coupled receptors (GPCRs) are important signaling
anti-inflammatory effects, but the mechanisms are molecules for many aspects of cellular function. They are
poorly understood. Here, we show that the G members of a large family that share common structural motifs
protein-coupled receptor 120 (GPR120) functions as such as seven transmembrane helices and the ability to acti-
an u-3 FA receptor/sensor. Stimulation of GPR120 vate heterotrimeric G proteins, such as Gas, Gai, and Gaq.
Ligands bind specifically to GPCRs to stimulate and induce
with u-3 FAs or a chemical agonist causes broad
a variety of cellular responses via several second messenger
anti-inflammatory effects in monocytic RAW 264.7 pathways; e.g., modulation of cAMP production, the phospho-
cells and in primary intraperitoneal macrophages. lipase C pathway, ion channels, and mitogen-activated protein
All of these effects are abrogated by GPR120 knock- kinases (Ulloa-Aguirre et al., 1999; Gether, 2000; Schulte and
down. Since chronic macrophage-mediated tissue Fredholm, 2003. Recently, several groups reported that five
inflammation is a key mechanism for insulin resis- orphan receptors, GPR40, GPR41, GPR43, GPR84, and
tance in obesity, we fed obese WT and GPR120 GPR120, can be activated by free fatty acids (FFAs). Short-
knockout mice a high-fat diet with or without u-3 FA chain fatty acids (FAs) are specific agonists for GPR41 and
supplementation. The u-3 FA treatment inhibited GPR43 (Tazoe et al., 2008) and medium-chain FAs for GPR84
inflammation and enhanced systemic insulin sensi- (Wang et al., 2006a). Long-chain FAs can activate GPR40
tivity in WT mice, but was without effect in GPR120 (Itoh et al., 2003) and GPR120 (Hirasawa et al., 2005). Hirasawa
et al. showed that the stimulation of GPR120 by FFAs resulted
knockout mice. In conclusion, GPR120 is a functional
in elevation of [Ca2+]i and activation of the ERK cascade which
u-3 FA receptor/sensor and mediates potent insulin suggests interactions with the Gaq family of G proteins.
sensitizing and antidiabetic effects in vivo by repres- However, other physiological functions of GPR120 remain to
sing macrophage-induced tissue inflammation. be explored. In the current study, we found that GPR120 was
highly expressed in adipose tissue, and proinflammatory
macrophages. The high expression level of GPR120 in mature
INTRODUCTION adipocytes and macrophages indicates that GPR120 might
play an important role in these cell types. We stimulated
Chronic activation of inflammatory pathways plays an important GPR120 with a synthetic agonist (GW9508) and omega-3 fatty
role in the pathogenesis of insulin resistance and the macro- acids (u-3 FAs) and examined whether activation of GPR120
phage/adipocyte nexus provides a key mechanism underlying affected LPS- and TNF-a-induced inflammatory signaling
the common disease states of decreased insulin sensitivity responses. While SFAs are proinflammatory and unsaturated
(Schenk et al., 2008). This involves migration of monocytes/ FAs are generally neutral, we found that u-3 FAs (docosahex-
macrophages to adipose tissue (including intramuscular fat aenoic acid (C22:6n3, DHA) and eicosapentaenoic acid
depots) and liver with subsequent activation of macrophage (C20:5n3, EPA)), the major ingredients in fish oil, exert potent
proinflammatory pathways and cytokine secretion. Through anti-inflammatory effects through GPR120.
paracrine effects, these events promote inflammation and b-arrestins can serve as scaffold or adaptor proteins for a wide
decreased insulin sensitivity in nearby insulin target cells range of GPCRs, as well as a selected group of other receptor
(Shoelson et al., 2007; Schenk et al., 2008). In these studies, subtypes (Miller and Lefkowitz, 2001). After ligand binding,
we explore the interlocking biology between proinflammatory b-arrestins can associate with the cytoplasmic domains of

Cell 142, 687–698, September 3, 2010 ª2010 Elsevier Inc. 687


Figure 1. Expression Level of GPR120 and
GPR120-Mediated Anti-inflammatory Res-
ponse in RAW 264.7 Cells
(A and B) (A) The mRNA expression pattern of
various lipid sensing GPCRs is shown in adipose
tissue, (B) CD11c+ bone marrow-derived dendritic
cells (BMDCs), bone marrow-derived macro-
phages (BMDMs), IPMacs, 3T3-L1 preadipocytes,
differentiated 3T3-L1 adipocytes, RAW 264.7
cells, and L6 myocytes. Ribosomal protein S3
(RPS3) was used as internal control.
(C) Expression of GPR120 in SVF, adipocytes and
hepatic Kupffer cells from chow (NC)- or HFD-fed
mice was examined by q-PCR. Data are ex-
pressed as the mean ± SEM of at least three
independent experiments in triplicate. *p < 0.05
versus NC.
(D) RAW 264.7 cells, transfected with scrambled
(Scr) or GPR120 #2 siRNA (GPR120 KD), were
treated with 100 mM of GW9508 for 1 hr prior to
LPS (100 ng/ml) treatment for 10 min and then
subjected to western blotting. Left panel is a repre-
sentative image from three independent experi-
ments, and the scanned bar graph (right panel)
shows fold induction over basal conditions.
Knockdown efficiency of GPR120 siRNA is shown
in Figure S1.
(E) Cytokine secretion level was measured in RAW
264.7 cells by ELISA. Data are expressed as the
mean ± SEM of three independent experiments.
*p < 0.05 versus LPS treatment in scrambled
siRNA transfected cells. See also Figures S1
and S2.

GPCRs and couple the receptor to specific downstream sensing GPCR which is highly expressed in adipose tissue,
signaling pathways, as well as mediate receptor endocytosis proinflammatory CD11c+ macrophages (BMDCs), mature adipo-
(Luttrell and Lefkowitz, 2002). Here we find that b-arrestin2 asso- cytes, and monocytic RAW 264.7 cells (Figure 1A and 1B).
ciates with ligand-stimulated GPR120 and participates in down- GPR120 is induced in the stromal vascular fraction (SVF) of
stream signaling mechanisms. adipose tissue (which contains the macrophages), as well as in
Since chronic inflammation is a mechanistic feature of hepatic Kupffer cells, during high-fat diet (HFD) feeding in mice
obesity-related insulin resistance, we postulated that the anti- (Figure 1C). GPR120 is also expressed in enteroendocrine L cells
inflammatory effect of GPR120 stimulation could promote insulin with negligible expression in muscle (Figure S4C available
sensitization. In the present study, we elucidate the role of online), hepatocytes or other cell types (Hirasawa et al., 2005;
GPR120 activation in integrating anti-inflammatory and insulin Gotoh et al., 2007).
sensitizing effects in vitro and in vivo.
Ligand-Stimulated GPR120 Exerts Anti-inflammatory
RESULTS Effects
It has been previously reported that GPR120 signals via
GPR120 Expression a Gaq/11-coupled pathway and can respond to long chain FAs
Fatty acids (FAs) can function as endogenous ligands modu- (Hirasawa et al., 2005). To pursue the biology of GPR120,
lating inflammatory responses, but not all FAs work in the a tool compound was needed, and, some years ago, Glaxo pub-
same way. In general, saturated FAs (SFAs) are proinflammatory, lished GW9508 as a GPR40 selective agonist. However, this
unsaturated FAs are weakly proinflammatory or neutral, and u3- compound was not specific and also stimulated GPR120 (Bris-
FAs can be anti-inflammatory (Lee et al., 2003; Calder, 2005; coe et al., 2006). Since macrophages and adipocytes do not
Solinas et al., 2007). Because of the importance of inflammation express GPR40 (this was confirmed by repeated q-PCR and
in a number of chronic human diseases including insulin resis- RT-PCR measures, Figure S1A), GW9508 is a functional
tance, obesity, and type 2 diabetes mellitus, we surveyed the GPR120 specific compound in these cell types. Using this
family of FA sensing GPCRs (GPR40, 41, 43, 84, and 120). Based approach, we found that GW9508 treatment broadly and mark-
on its tissue expression pattern, GPR120 emerged as a receptor edly repressed the ability of the TLR4 ligand LPS to stimulate
of particular interest. As seen in Figure 1, GPR120 is the only lipid inflammatory responses in RAW 264.7 cells (Figure 1D and E).

688 Cell 142, 687–698, September 3, 2010 ª2010 Elsevier Inc.


Figure 2. Omega-3 FA Stimulates GPR120
and Mediates Anti-inflammatory Effects
(A–D) GPR120-mediated SRE-luc activity after
treatment with various FAs. Results are fold
activities over basal. Each data point represents
mean ± SEM of three independent experiments
performed in triplicate. Black lines indicate SRE-
luc activities without GPR120 transfection or with
non-stimulating FAs. DHA inhibits LPS-induced
inflammatory signaling (B), cytokine secretion (C),
and inflammatory gene mRNA expression level
(D) in RAW 264.7 cells, but not in GPR120 knock-
down cells.
(E and F) GPR120 stimulation inhibits LPS-induced
inflammatory response in WT primary macro-
phage. Data are expressed as the mean ± SEM
of three independent experiments. *p < 0.05
versus LPS treatment in scrambled siRNA trans-
fected cells or WT IPMacs. See also Figure S2.

lation in RAW 264.7 cells, but only DHA-


and a-linolenic acid-mediated ERK phos-
phorylation were abolished by GPR120
knockdown (Figure S2A). These results
indicate that u-3 FAs, but not SFAs,
specifically activated ERK via GPR120.
The activation of GPR120 by u-3 FAs,
as well as its expression in adipocytes
and macrophages, led us to study
whether DHA, a representative u-3 FA,
can modulate inflammation through
GPR120 in these cells. To examine this,
we pretreated RAW 264.7 cells and
3T3-L1 adipocytes with GW9508 or DHA
for 1 hr, followed by LPS (TLR4), TNF-a,
TLR2, or TLR3 stimulation, respectively.
We found that GW9508 and, more impor-
tantly, DHA, strongly inhibited LPS-
Thus, GW9508 inhibited LPS stimulated phosphorylation of IKKb induced phosphorylation of JNK and IKKb, IkB degradation,
and JNK, prevented IkB degradation, and inhibited TNF-a and cytokine secretion and inflammatory gene expression level in
IL-6 secretion. All of these effects of GW9508 were completely RAW 264.7 cells (Figures 2B–2D) as well as TNF-a, TLR2 and
abrogated by siRNA mediated knockdown of GPR120 (Figures TLR3-induced JNK and IKKb phosphorylation in 3T3-L1 adipo-
1D and 1E and Figure S1F). cytes (Figure S2B) or RAW 264.7 cells (Figure S2C). All of the
Based on these remarkable anti-inflammatory effects of effects of GW9508 and DHA were completely prevented by
GPR120 stimulation, we established a cell based reporter GPR120 knockdown, demonstrating that these anti-inflamma-
system by transfecting HEK293 cells with constructs for tory effects were specifically exerted through GPR120 (Figure 1,
GPR120 along with a serum response element-luciferase Figure 2, Figure S1, and Figure S2). Similar results were seen in
promoter/reporter (SRE-luc). Since GPR120 is a Gaq/11- primary wild-type (WT) intraperitoneal macrophages (IPMacs)
coupled receptor, it stimulates both PKC and MAP kinase, and and GPR120 knockout (KO) IPMacs (Figures 2E and 2F). These
both of these biologic effects are detected by the SRE-driven data argue that GPR120 is an u-3 FA receptor or sensor, and
reporter system (Oh et al., 2005). The reporter cells were treated provide a molecular mechanism for the anti-inflammatory effects
with various FAs and the synthetic GW9508 ligand. We found of this class of FAs.
that GW9508, the u-3 FAs (DHA and EPA) and palmitoleate
(C16:1n7), all activated the SRE-luc reporter with an EC50 of Role of b-arrestin2 in GPR120 Signaling
1-10 mM (Figure 2A), while SFAs were without effect. GW9508 Given these potent cell selective anti-inflammatory effects,
and DHA were used at 100 mM in all subsequent studies to it was of interest to understand the specific mechanisms
achieve maximal action. The u-3 FAs (DHA and a-linolenic whereby signals from GPR120 inhibit inflammatory pathways.
acid), and SFA (palmitic acid (C16:0)) activated ERK phosphory- To further assess this, we used RNA interference to examine

Cell 142, 687–698, September 3, 2010 ª2010 Elsevier Inc. 689


Figure 3. GPR120 Internalization with
b-arrestin2 Mediates Anti-inflammatory
Effects
(A) RAW 264.7 cells were transfected with siRNA as
indicated and stimulated with or without 100 mM of
DHA 1 hr prior to LPS (100 ng/ml) treatment for
10 min and then subjected to western blotting.
(B) TNF-a secretion was measured in RAW 264.7
cell cultured media with or without RNA interfer-
ence as indicated.
(C) Phosphorylation of TAK1 and MKK4 in RAW
264.7 cells with or without siRNA transfection as
indicated.
(D) HEK293 cells were cotransfected with HA-
GPR120 and b-arrestin2$GFP to analyze
GPR120 internalization after DHA stimulation for
the indicated times. GPR120 (red) and b-arrestin2
(green) were localized by confocal microscopy.
(E–H) (E) Coimmunoprecipitation between GPR120
and b-arrestin2 with DHA stimulation for 30 min
in RAW 264.7 cells and, (F) HEK293 cells (HA-
GPR120 and b-arrestin2$GFP), respectively. Lysate
indicates 1/10 input in each experiment. Interaction
between TAB1 and b-arrestin2 (G) and interaction
between TAB1 and TAK1 (H) were detected by
coimmunoprecipitation and the scanned bar graph
quantitates the association in RAW 264.7 cells.
(I) Schematic diagram of the b-arrestin2 and
GPR120-mediated anti-inflammatory mechanism.
Red colored letters and arrows indicate the DHA-
mediated anti-inflammatory effect, and black
colored letters and arrows indicate the LPS- and
TNF-a-induced inflammatory pathway. See also
Figure S2.

phorylation in a GPR120 and b-arrestin2-


dependent manner. Since TLR2/3/4 and
TNF-a signaling were inhibited by
GPR120 activation, these results indicate
that DHA signaling intersects at TAK1 and
molecules involved in generation of GPR120 signals. As seen in inhibits all upstream input activating signals via a GPR120/b-
Figures 3A and 3B, LPS signaling was not affected by b-ar- arrestin2 interaction (Figure 3I).
restin1, -2, or Gaq/11 knockdown. However, with b-arrestin2 After ligand stimulation, b-arrestin2 can translocate to
knockdown, DHA-mediated anti-inflammatory signaling was ex- a number of GPCRs where it mediates receptor internalization
tinguished, while b-arrestin1 and Gaq/11 knockdown were and signaling (Barak et al., 1997). We transfected HEK293 cells
without effect (Figure 3A). with b-arrestin2$GFP to visualize intracellular trafficking of b-ar-
Figure 3A and Figure S2 show that GPR120 stimulation inhibits restin2 following activation of GPR120 (Figure 3D). In the basal
both TLR4- and TNF-a mediated inflammatory responses. Since state, GPR120 was localized to the plasma membrane as
the TNF-a and TLR signaling cascades converge downstream of assessed by immunostaining (red fluorescence, Figure 3D),
GPR120 activation, these results indicate that the site of while b-arrestin2 exhibited a diffuse, largely cytoplasmic stain-
GPR120-induced inhibition is either at, or upstream, of JNK/ ing pattern (green, Figure 3D). Following DHA treatment for
IKKb. LPS activates inflammation through the TLR4 pathway 5 min, b-arrestin2$GFP translocated from the cytosol to the
by engaging the serine kinase IRAK, leading to phosphorylation plasma membrane and can be seen colocalized with GPR120
of transforming growth factor-b activated kinase 1 (TAK1) which (merged, right fields). After 30 min of DHA treatment, much of
is upstream of MKK4/JNK and IKKb (Kawai and Akira, 2006, the GPR120 is internalized, as visualized by punctate intracel-
Figure 3I). TNF-a and TLR2/3 also leads to stimulation of lular staining (lower left panel), and b-arrestin2$GFP is now
TAK1, resulting in activation of IKKb and JNK (Takaesu et al., colocalized with the intracellular GPR120 (lower right panel,
2003). Consequently, we determined whether DHA stimulation Figure 3D). DHA-stimulated binding of b-arrestin2 to activated
of GPR120 inhibited TAK1 and MKK4. As seen in Figure 3C, GPR120 was also detected by coimmunoprecipitation
DHA treatment abrogated LPS-induced TAK1 and MKK4 phos- (Figure 3E and F).

690 Cell 142, 687–698, September 3, 2010 ª2010 Elsevier Inc.


Figure 4. GPR120 Activation Enhances
GLUT4 Translocation and Glucose Uptake
(A) 3T3-L1 adipocytes were transfected with
a dually tagged HA-GLUT4-GFP construct. Total
GLUT4 expression was determined by GFP fluo-
rescence, and GLUT4 translocation to the cell
surface after 100 ng/ml insulin or 100 mM DHA
stimulation for 30 min was determined by indirect
immunofluorescence of the HA-conjugated with
Alexa 594 in fixed cells. Translocation following
insulin stimulation was expressed as a percentage
of the maximum response. The bar graph
represents the mean ± SEM data from four inde-
pendent experiments. *p < 0.05 versus vehicle
treatment. (B) Glucose uptake was measured in
WT and GPR120 KO mouse primary adipose
tissue and in (C–H) 3T3-L1 adipocytes ± siRNA
with the indicated treatment. Data are expressed
as mean ± SEM of three independent experiments
in triplicate. *p < 0.05 versus basal activity. The
indicated siRNA knockdown efficiency was vali-
dated by western blotting. See also Figure S3.

GPR120 Activation Enhances


Glucose Uptake in 3T3-L1
Adipocytes
Since our data show that GPR120 is
expressed in mature adipocytes and sig-
nals through Gaq/11 in these cells, we
assessed the effects of GPR120 stimula-
tion on insulin sensitivity in primary adi-
pose tissue cultures and in 3T3-L1 adipo-
cytes. Primary adipose tissue explants
and 3T3-L1 adipocytes were pretreated
for 30 min with GW9508 or DHA, followed
by measurement of basal and insulin
stimulated GLUT4 translocation (Fig-
LPS or TNF-a signaling activate TAK1 by causing the associ- ure 4A) and 2-deoxyglucose (2-DOG) transport (Figures 4B–
ation of TAK1 binding protein 1 (TAB1) with TAK1. Figure 3H 4H). Ligand-stimulation of GPR120 led to an increase in glucose
shows that LPS stimulation of RAW 264.7 cells causes TAB1/ transport and translocation of GLUT4 to the plasma membrane
TAK1 association. DHA treatment leads to the association of in adipocytes, but was without effect in muscle cells (Figure S4D)
b-arrestin2 with TAB1 (Figure 3G) and largely blocks TAK1/ which don’t express GPR120 (Figure 1B and Figure S4C). This
TAB1 association (Figure 3H). To further examine the interaction stimulatory effect of DHA and GW9508 was blocked when
site of b-arrestin2 and GPR120 or TAB1, we pursued coimmuno- GPR120 or Gaq/11 was depleted by siRNA knockdown
precipitation with a series of b-arrestin2 truncation/deletion (Figure 4D and Figure 4F). GLUT4 knockdown also blocked the
mutants (Figure S2D). Full-length b-arrestin2 was able to bind effects of DHA and GW9508, while the effects of insulin were
to GPR120 and TAB1 only in the presence of DHA, clearly decreased by 90% (Figure 4E). This, along with the GLUT4
showing the DHA dependency of this interaction. Interestingly, translocation data provided in Figure 4A, indicates that the
only the full-length b-arrestin2 coprecipitated with GPR120 and stimulatory effects of GPR120 are indeed working through
TAB1, while a series of deletion/truncation b-arrestin2 mutants GLUT4. Further assessment of this pathway showed that DHA
did not, indicating that the interactions are dependent on the had a modest effect to stimulate phosphorylation of Akt, but
complete tertiary structure of b-arrestin2 (Figure S2D; Luttrell that this was abrogated with GPR120 knockdown (Figure S3A).
et al., 1999). Taken together, these results suggest that The effects of DHA to stimulate Akt were blocked by inhibiting
GPR120 activation leads to association of b-arrestin2 with the PI3 kinase with LY294002 (Figure S3B). Finally, DHA did not
receptor and that this complex subsequently internalizes, where- stimulate IRS-1 phosphorylation (Figure S3C), indicating that
upon b-arrestin2 can bind to TAB1. The data further suggest that its glucose transport stimulatory effects were downstream of
association of b-arrestin2 with TAB1 blocks TAB1/TAK1 binding, IRS-1. Knockdown of Gaq/11 also completely blocked the
resulting in inhibition of TAK1 phosphorylation and activation effects of DHA to stimulate glucose transport (Figure 4F), while
(Figure 3I). b-arrestin1 or -2 knockdown was without effect (Figures 4G

Cell 142, 687–698, September 3, 2010 ª2010 Elsevier Inc. 691


Figure 5. In Vivo Metabolic Studies in
GPR120 KO Mice
(A) GTT in NC-fed WT and GPR120 KO mice. n = 7
per group.
(B and C) Insulin concentration was measured at
the indicated time points and (C) area-under-curve
analysis of the insulin data shows a significant differ-
ence between WT and GPR120 KO mice on NC.
(D) Hyperinsulinemic/euglycemic clamp studies in
chow-fed WT and GPR120 KO mice.
(E) Clamp studies in HFD, u-3 FA supplemented
(+u3), and Rosiglitazone treated HFD mice
(+Rosi). n = 8 per group, *p < 0.05 compared to
HFD-fed WT group.
(F) Mean ± SEM plasma concentration (mole (%))
of DHA and EPA for each diet in WT and GPR120
KO mice. n = 7 per each group. *p < 0.05,
compared to NC, and **p < 0.05 compared to
HFD. Data are represented as mean ± SEM. See
also Figure S4, Figure S5, Figure S6, and Table S2.

(GIR) required to maintain euglycemia in


the KO mice. Since 70%–80% of total
body insulin stimulated glucose disposal
is accounted for by skeletal muscle
glucose uptake (Baron et al., 1988), the
decreased insulin stimulated (IS)-glucose
disposal rate (GDR) provides direct
evidence for skeletal muscle insulin resis-
tance in the KO mice. Likewise, the
GPR120 KO mice exhibited a marked
decrease in the ability of insulin to sup-
press hepatic glucose production (HGP),
demonstrating the presence of hepatic
insulin resistance. Thus, the decreased
and 4H). Interestingly, Gaq/11 knockdown not only inhibited GIR was 50% related to muscle and 50% due to liver insulin
DHA and GW9508 stimulated glucose transport, but it also atten- resistance, respectively. Since the chow diet contains exoge-
uated insulin stimulatory effects, and the latter is fully consistent nous u-3 FAs, we conclude that blunted u-3 FA signaling in the
with previous publication (Imamura et al., 1999) showing the role KO mice, accounts for the decreased insulin sensitivity.
of Gaq/11 in insulin signaling to glucose transport in adipocytes. Since u-3 FA administration can improve insulin sensitivity in
This scheme is shown in Figure S3D. rats (Buettner et al., 2006), we reasoned that u-3 FA supplemen-
tation could alleviate HFD/obesity-induced insulin resistance in
In Vivo Metabolic Studies in GPR120 KO Mice WT mice, but would be ineffective in GPR120 KOs. Accordingly,
Since chronic tissue inflammation can cause insulin resistance, WT and GPR120 KO mice were placed on 60% HFD for 15 weeks.
we hypothesized that deletion of GPR120 would enhance the At this point, separate groups of 15 mice each, were treated for
proinflammatory tone, promoting glucose intolerance and five additional weeks with 60% HFD or an isocaloric HFD diet con-
decreased insulin sensitivity. To test this idea, GPR120 taining 27% fish oil supplementation enriched in u-3 FAs. This diet
KO mice and WT littermates were evaluated on normal chow provided 50 and 100 mg of DHA and EPA, respectively, per
diet (NC). Body weights were similar in both groups, and as mouse, per day. Figure 5E shows that administration of the u-3
summarized in Figure 5, glucose tolerance tests (GTT) showed FA diet led to improved insulin sensitivity with increased glucose
a mild degree of impairment in GPR120 KO animals compared infusion rates, enhanced muscle insulin sensitivity (increased IS-
to WTs (Figure 5A). More impressively, insulin secretion was GDR), greater hepatic insulin sensitivity (increased HGP suppres-
more than 2-fold greater in the KO animals, and the combination sion), and decreased hepatic steatosis (Figures S6A and S6B).
of hyperinsulinemia and mild glucose intolerance indicates the Importantly, the u-3 FA diet was completely without effect in the
presence of insulin resistance (Figures 5B and 5C). This was GPR120 KO mice. A separate group of WT mice were treated
confirmed by performing hyperinsulinemic/euglycemic clamp with the insulin sensitizing thiazolidinedione Rosiglitazone, and
studies in the chow fed WT and KO mice (Figure 5D). These the effects of u-3 FAs were equal to or greater (HGP suppression)
studies revealed a 31% decrease in the glucose infusion rate than the effects of this clinically used insulin sensitizing drug.

692 Cell 142, 687–698, September 3, 2010 ª2010 Elsevier Inc.


In addition to improving hepatic insulin sensitivity, u-3 FA was minimal in both groups on HFD (Figure 6A). On the u-3 FA
treatment had a beneficial effect on hepatic lipid metabolism, diet, we observed a decrease in F4/80 staining, along with
causing decreased liver triglycerides, DAGs, along with reduced a marked increase in MGL1 positive cells in WT mice. Impor-
SFA and u-6 FA content in the various lipid classes (Figures tantly, no change in F4/80 or MGL1 staining was noted in the
S6A–S6C and Table S2). The u-3 FA supplementation was GPR120 KO mice on the u-3 FA diet. SVFs were prepared
entirely without effect, or much less effective, at reducing hepatic from adipose tissue and analyzed by flow cytometry to quanti-
lipid levels in the GPR120 KOs. tate the total number of ATMs, as well as the content of
Interestingly, in the absence of u-3 FA supplementation, CD11b+ and CD11c+ and negative macrophage subpopulations
GPR120 KO mice were just as susceptible to HFD-induced (Figure 6B). HFD led to a large but comparable increase in
insulin resistance as were the WT mice. We hypothesize that CD11b+ and CD11c+ ATM content in WT and GPR120 KO
this was because the 60% HFD is relatively deficient of exoge- mice (Figure 6B, middle panel). Treatment with the u-3 FA-en-
nous u-3 FAs, so that ligands for GPR120 were relatively absent riched HFD caused a striking decrease in CD11b+ and CD11c+
in these animals. To assess this, we performed a lipomics anal- ATMs in WT mice, but was without effect in the GPR120 KO
ysis of the various fatty acid classes in the chow and HFD-fed WT group (Figure 6B, right panel). Thus, the FACS analysis was fully
and KO mice. As predicted, circulating concentrations of u-3 consistent with the histological results. Interestingly, CD11c+
FAs were much lower on HFD compared to chow diets, and ATM content was also greater in the GPR120 KOs on the chow
the administration of the u-3 FA supplement to the HFD led to diet relative to WT consistent with the insulin resistance in the
a large increase in plasma u-3 FA content in both genotypes KO animals.
(Figure 5F). This would account for the relative lack of effect of It seemed possible that the reduction in ATM content in WT
GPR120 KO on HFD alone, since u-3 FA ligand stimulation is animals on the u-3 FA diet reflected decreased chemotaxis of
negligible, while the KO animals displayed an insulin resistant macrophages. To test this hypothesis, we measured the migra-
phenotype on chow diets when a moderate level of u-3 FAs tory capacity of IPMacs from WT and GPR120 KO mice using
was provided. Importantly, the GPR120 KO mice are completely an in vitro transwell chemotaxis assay. As seen in Figure 6C,
refractory to the insulin sensitizing effects of u-3 FA administra- macrophages from both groups readily migrated toward condi-
tion on HFD. tioned media (CM) harvested from 3T3-L1 adipocytes. Pretreat-
To address the contribution of macrophages to the overall ment of macrophages with DHA for 3 hr before exposure to CM
in vivo phenotype, we performed bone marrow transplantation led to an 80% inhibition of chemotactic capacity in WT macro-
(BMT) from GPR120 KOs into irradiated WT mice (adoptive phages, but had no significant effect on IPMacs obtained from
transfer) to generate hematopoietic cell deletion of GPR120. the GPR120 KO mice. Similar experiments were performed
The studies in the BMT WT and BMT GPR120 KO mice on using the specific chemokine, monocyte chemotactic protein-1
chow diet revealed a highly significant 20%–30% decrease in (MCP-1) as a chemoattractant, rather than CM, and these exper-
GIR in the KOs, with a more dramatic impairment in the ability iments yielded identical results (Figure 6D). These data indicate
of insulin to suppress hepatic glucose production (Figure S4A). that u-3 FAs cause decreased macrophage chemotaxis by
Thus, the studies in the BMT animals on the chow diet are acting through the GPR120 receptor, contributing to the differ-
comparable to the results (Figure 5D) observed in WT versus ences in ATM content seen in Figures 6A and 6B.
whole body GPR120 KOs on chow diet. When studied on the
HFD ± u-3 FA supplementation (Figure S4B), the u-3 FA supple- Omega-3 FAs Decrease M1 Proinflammatory Gene
mented BMT GPR120 KO animals exhibited a 30% decrease in and Increase M2 Anti-inflammatory Gene Expression
GIR compared to the u-3 FA supplemented BMT WTs. This was in Adipose Tissue
explained by skeletal muscle insulin resistance (decreased IS- As shown in Figure 7A, expression of M1 inflammatory genes
GDR) and hepatic insulin resistance (decreased HGP suppres- such as IL-6, TNF-a, MCP-1, IL-1b, iNOS, and CD11c was
sion) in the GPR120 KOs compared to the WT BMT mice on increased by HFD compared to chow diet in both genotypes,
the u-3 FA supplemented HFD. These data are fully consistent and was reduced in the u-3 FA treated WT mice, but not in
with the results in the global KOs (Figure 5E) and reinforce the the GPR120 KO mice. Even on chow diet, expression of several
concept that the in vivo phenotype we observed can be largely inflammatory genes was higher in GPR120 KOs compared to
traced to hematopoietic cells/macrophages. WT, consistent with the insulin resistance observed in the
chow-fed KO mice. Expression of the M2 anti-inflammatory
Omega-3 FAs Reduce Inflammatory Macrophages genes, arginase 1, IL-10, MGL1, Ym-1, Clec7a, and MMR
in Adipose Tissue was increased by u-3 FAs in WT, but not in the GPR120 KO
We conducted histologic examination of adipose tissue adipose tissue (Figure 7B). These results are consistent with
macrophages (ATMs) from WT and GPR120 KO mice on HFD Figure 6 and demonstrate that the dietary change from HFD
or the u-3 FA enriched HFD by immunostaining for the M1 to u-3 FA supplemented HFD resulted in an overall decreased
macrophage marker F4/80 and the M2 macrophage marker proinflammatory profile in adipose tissue from WT, but not in
MGL1 (Lumeng et al., 2008) (Figure 6A). Consistent with previous GPR120 KO mice. These changes in gene expression were
studies (Weisberg et al., 2003; Xu et al., 2003; Nguyen et al., predominantly manifested in the SVF, except for MCP-1 and
2007), HFD induced a large increase in F4/80 positive ATMs, IL-6, which are known to be readily expressed in adipocytes
which form crown-like structures (CLS) around adipocytes in (Figure S7). Qualitatively similar results were seen in the liver
both WT and GPR120 KO mice. In contrast, MGL1 staining (Figures S6D and S6E).

Cell 142, 687–698, September 3, 2010 ª2010 Elsevier Inc. 693


Figure 6. Omega-3 FA Enriched Diet Decreases
Inflammatory Macrophage Infiltration in Adipose
Tissue
(A) Confocal merged images from epididymal fat pads
from HFD and u-3 FA enriched HFD (HFD+u3)-fed WT
and GPR120 KO mice, costained with anti-F4/80 (green)
and anti-Caveolin1 (blue) antibodies, left 4 panels, or
anti-MGL1 (green) and anti-Caveolin1 (red) antibodies,
right 4 panels. The image is representative of similar
results from three to four independent experiments. Scale
bar represents 100 mm.
(B) Dot plot representation of CD11b versus CD11c
expression for FACS data obtained from adipose tissue
SVF of NC, HFD or HFD+u3-fed WT and GPR120 KO.
Scattergram is representative from three independent
mice from each group.
(C and D) Migratory capacity of IPMacs from WT and
GPR120 KO mice as measured using an in vitro transwell
chemotaxis assay as described under supplemental
experimental procedures. Data are expressed as mean ±
SEM of three independent experiments in triplicate. *,
p<0.05 versus CM treatment.

of FAs to inhibit both the TLR2/3/4 and the


TNF-a response pathways and cause systemic
insulin sensitization.
GPR120 is a Gaq/11-coupled receptor, and
since it is expressed in enteroendocrine L cells,
past interest in this receptor has focused on its
potential ability to stimulate L cell GLP-1 secre-
tion. In the current study, we show that, in addi-
tion to L cells, GPR120 is highly expressed in
proinflammatory, M1-like macrophages and
mature adipocytes, with negligible expression
in muscle, pancreatic b-cells, and hepatocytes
(Gotoh et al., 2007). In the HFD/obese mouse
model, GPR120 expression is highly induced in
ATMs as well as resident liver macrophages
(Kupffer cells). To explore the biology around
GPR120, we established an artificial reporter
DISCUSSION cell assay and found that the u-3 FAs, DHA, and EPA, are ligands
for GPR120, and comparable to the effects of a non-selective
In this report we show that GPR120 functions as an u-3 FA GPR120 tool compound (GW9508), the u-3 FAs exert potent
receptor/sensor in proinflammatory macrophages and mature anti-inflammatory effects in macrophages. Our results also re-
adipocytes. By signaling through GPR120, DHA and EPA (the vealed the molecular mechanisms underlying these anti-inflam-
major natural u-3 FA constituents of fish oil), mediate potent matory effects. Thus, DHA stimulation of GPR120 inhibits both
anti-inflammatory effects to inhibit both TLR and TNF-a inflam- the TLR2/3/4 and TNF-a proinflammatory cascade. Since acti-
matory signaling pathways. The mechanism of GPR120-medi- vation of IKKb and JNK are common to TLR and TNF-a signaling,
ated anti-inflammation involves inhibition of TAK1 through a this indicates that the locus of GPR120 inhibition is at or proximal
b-arrestin2/TAB1 dependent effect. Since chronic tissue inflam- to these kinases. TAK1 activation stimulates both the IKKb/NFkB
mation is an important mechanism causing insulin resistance and JNK/AP1 pathways, and the TLR and TNF-a signaling path-
(Xu et al., 2003; Shoelson et al., 2007; Schenk et al., 2008), the ways converge at this step. Our data show that stimulation of
anti-inflammatory actions of u-3 FAs exert potent insulin sensi- GPR120 specifically inhibits TAK1 phosphorylation and activa-
tizing effects. The in vivo anti-inflammatory and insulin sensi- tion providing a common mechanism for the inhibition of both
tizing effects of u-3 FAs are dependent on expression of TLR and TNF-a signaling.
GPR120, as demonstrated in studies of obese GPR120 KO Beta-arrestins can serve as important adaptor and scaffold
animals and WT littermates. Thus, GPR120 is highly expressed molecules mediating the functions of a number of different
in proinflammatory macrophages and functions as an u-3 FA GPCRs, as well as other receptor subtypes (Miller and Lefkowitz,
receptor, mediating the anti-inflammatory effects of this class 2001). The C-terminal region of GPR120 contains several

694 Cell 142, 687–698, September 3, 2010 ª2010 Elsevier Inc.


Figure 7. M1 and M2 Inflammatory Gene Expres-
sion Levels in Adipose Tissue from WT versus
GPR120 KO Mice
Relative mRNA levels for M1 proinflammatory genes (A)
and M2 anti-inflammatory genes (B) in NC, HFD, or
HFD+u3 (+u3)-fed WT and GPR120 KO mice, as measured
by q-PCR. Data are expressed as mean ± SEM of three
independent experiments in triplicate. n=7 per group, *,
p<0.05 compared to the HFD-fed WT group. **, p<0.05
compared to the WT versus GPR120 KO on NC. See also
Figure S7. Primer sequences are shown in Table S1.

11-coupled receptor in other contexts. This


provides further evidence demonstrating the
concept that a single GPCR can independently
signal through multiple pathways. In previous
studies, we have demonstrated that Gaq/11
activation can lead to stimulation of GLUT4
translocation in adipocytes. Since GPR120
was expressed in mature adipocytes, but not
preadipocytes, we explored the potential role
of GPR120 in glucose transport control. Inter-
estingly, we found that DHA stimulation of
GPR120 in 3T3-L1 adipocytes increased
GLUT4 translocation to the cell surface with
a subsequent increase in glucose transport
into the cells. RNA interference studies showed
putative b-arrestin2 binding motifs [(S/T)X4-5(S/T); Cen et al., that the DHA effect on glucose uptake was GPR120, GLUT4, and
2001], but whether b-arrestins play any role in GPR120 function Gaq/11 dependent, but independent of b-arrestin2. This effect
was unknown. Here we find that activation of GPR120 by DHA was about 30%–50% as great as the effect of insulin and the
stimulation leads to association of the receptor with b-arrestin2, actions of DHA on glucose uptake were additive to those of
but not b-arrestin1, and that the anti-inflammatory effects of a submaximally stimulating concentration of insulin. From this,
GPR120 are completely b-arrestin2 dependent. Functional it is possible to propose that these insulinometic effects con-
immunocytochemical studies showed that DHA stimulation tribute to the overall insulin sensitizing actions of u-3 FAs.
leads to recruitment of b-arrestin2 to the plasma membrane However, muscle glucose uptake accounts for the great majority
where it colocalizes with GPR120. This is followed by receptor (70%–80%) of insulin stimulated glucose disposal. Furthermore,
and b-arrestin2 internalization, where the two are now colocal- GPR120 is not expressed in muscle, and DHA did not stimulated
ized in the cytoplasmic compartment. TAB1 is the activating glucose uptake in L6 myocytes (Figures S4C and S4D). In addi-
protein for TAK1 and our results show that following DHA-stim- tion, acute administration of DHA had no stimulatory effects on
ulated internalization of the GPR120/b-arrestin2 complex, IS-GDR (Figure S4E). This reports the conclusion that the
b-arrestin2 can now associate with TAB1, as measured in coim- in vivo stimulatory effects of DHA on GDR are related to
munoprecipitation experiments; only full-length b-arrestin2 was anti-inflammation, and that the glucose transport stimulatory
capable of interacting with GPR120 and TAB1. This apparently effects in adipocytes contribute little to the overall phenotype.
blocks the association of TAB1 with TAK1, inhibiting TAK1 Since chronic, low grade tissue inflammation is an important
activation and downstream signaling to the IKKb/NFkB and cause of obesity-related insulin resistance, we reasoned that
JNK/AP1 system. These results provide a mechanism for the the anti-inflammatory effects of GPR120 stimulation should be
b-arrestin2-mediated inhibition of TLR4, TNF-a, and TLR2/3 coupled to insulin sensitizing actions in vivo. This idea was
action. Other studies in the literature are consistent with these confirmed in studies of WT and GPR120 KO mice. On a chow
findings, since it has been shown that b-arrestin2 can inhibit diet, the lean GPR120 KO mice were glucose intolerant, hyperin-
NFkB signaling in other systems (Gao et al., 2004; Wang et al., sulinemic and displayed decreased skeletal muscle and hepatic
2006b). Furthermore, Lefkowitz’ group has recently published insulin sensitivity, as measured during glucose clamp studies.
an extensive proteomics analysis of b-arrestin2 interacting They also displayed increased ATM content, relative to WT
partners, and among the 266 proteins they identified, TAB1 mice, and a 2- to 5-fold higher expression level of the M1 proin-
was represented on the list (Xiao et al., 2007). flammatory markers, MCP-1, iNOS, and IL-6 (Figure 7A). On
Interestingly, the anti-inflammatory effects mediated by HFD, GPR120 KO and WT mice became equally obese and
GPR120 were entirely dependent on b-arrestin2, but indepen- insulin resistant. Importantly, u-3 FA supplementation markedly
dent of Gaq/11, despite the fact that GPR120 can be a Gaq/ increased insulin sensitivity in WT mice but was without effect in

Cell 142, 687–698, September 3, 2010 ª2010 Elsevier Inc. 695


the GPR120 KOs. Consistent with these results, u-3 FA treat- tion versus anti-inflammatory actions are distinct processes, and
ment led to a decrease in ATM accumulation with reduced it is certainly possible that the products derived from u-3 FA
adipose tissue markers of inflammation in WT, but not in KO metabolism work on the former but not the latter.
mice. In addition to direct anti-inflammatory effects in macro- In summary, we have found that GPR120 functions as an u-3
phages, DHA treatment inhibited the ability of primary WT FA receptor/sensor and mediates robust and broad anti-inflam-
macrophages to migrate toward adipocyte CM. This could be matory effects, particularly in macrophages. After ligand stimula-
due to DHA-induced decreased chemokine secretion or down- tion, GPR120 couples to b-arrestin2 which is followed by
regulation of chemokine receptors, or both. In addition, it is receptor endocytosis and inhibition of TAB1-mediated activation
possible that DHA, by signaling through GPR120, can mediate of TAK1, providing a mechanism for inhibition of both the TLR
heterologous desensitization of other GPCR chemokine recep- and TNF-a proinflammatory signaling pathways. Since chronic
tors. We also observed a concomitant increase in M2 markers, tissue inflammation is linked to insulin resistance in obesity, we
such as IL-10, arginase 1, MGL1, Ym-1, Clec7a, and MMR. used GPR120 KO mice to demonstrate that u-3 FAs cause
This latter finding raises the possibility that u-3 FAs can redirect GPR120-mediated anti-inflammatory and insulin sensitizing
ATMs from an M1 to an M2 polarization state. Taken together, effects in vivo. Overall, these results strongly argue that anti-
these mechanisms account for the decreased inflammatory inflammatory effects can ameliorate insulin resistance in obesity.
state. The in vivo anti-inflammatory actions of u-3 FAs are Taken together, GPR120 emerges as an important control point
consistent with the insulin sensitizing effects of these agents in the integration of anti-inflammatory and insulin sensitizing
and are fully dependent on the presence of GPR120, indicating responses, which may prove useful in the future development
a causal relationship. Finally, the adoptive transfer studies of new therapeutic approaches for the treatment of insulin resis-
showed that hematopoietic cell GPR120 deletion results in tant diseases.
a comparable insulin resistant, u-3 FA non-responsive pheno-
type as seen in the global GPR120 KOs, indicating that this EXPERIMENTAL PROCEDURES
phenotype can be traced back to inflammatory events in
macrophages. Chemicals and Reagents
GW9508 was purchased from Tocris bioscience (Ellisville, MO) and DHA was
We also performed a detailed in vivo lipidomic analysis of FAs
from Cayman chemical (Ann Arbor, MI). All other chemicals were purchased
in the different lipid classes in the liver (Table S2). The results from Sigma unless mentioned otherwise.
showed that HFD leads to an increase in total TAGs, DAGs, total
SFAs, monounsaturated FAs and u-6 FAs in WT mice, while all of Animal Care and Use
these lipid changes are ameliorated with u-3 FA treatment. In the Male C57Bl/6 or GPR120 KO littermates were fed a normal chow (13.5% fat;
GPR120 KO mice, all of these lipids are elevated on HFD to the LabDiet) or high-fat diet (60% fat; Research Diet) ad libitum for 15–20 weeks
same extent as in WT mice, but, u-3 FA supplementation was from 8 weeks of age. GPR120 KO mice and WT littermates were provided
by Taconic Inc. (Hudson, NY). After 15 weeks on HFD, WT and GPR120 KO
either ineffective or much less effective. These results are
mice were switched to an isocaloric HFD-containing 27% menhaden fish oil
consistent with the view that the reversal of steatosis/non- replacement (wt/wt; menhaden fish oil: 16% EPA (C20:5n3), 9%, DHA
alcoholic fatty liver disease (NAFLD) by u-3 FA treatment is (C22:6n3), Research Diet) (Jucker et al., 1999; Neschen et al., 2007) and fed
mediated, in large part, by GPR120 and that the GPR120 KO for 5 weeks. Mice received fresh diet every 3rd day, and food consumption
mice are predisposed toward NAFLD even in the context of and body weight were monitored. Animals were housed in a specific path-
u-3 FA supplementation. ogen-free facility and given free access to food and water. All procedures
were approved by the University of California, San Diego animal care and
Dietary DHA is rapidly esterified into chylomicrons during the
use committee. In vivo metabolic studies were performed as described under
process of gastrointestinal absorption, and is also packaged
supplemental experimental procedures.
into VLDL triglycerides by the liver. DHA can also be esterified
into phospholipids and cholesterol esters associated with Data Analysis
circulating lipopoproteins and only a small proportion (5%) of Densitometric quantification and normalization were performed using the
total plasma DHA is found in the FFA pool. Through the action ImageJ 1.42q software. The values presented are expressed as the means ±
of lipoprotein lipase bound to the luminal surface of endothelial SEM. The statistical significance of the differences between various treat-
cells, u-3 FAs are cleaved from circulating triglycerides where ments was determined by one-way ANOVA with the Bonferroni correction
using GraphPad Prism 4.0 (San Diego, CA). The p < 0.05 was considered
they can act as ligands or be taken up by peripheral tissues
significant.
(Polozova and Salem Jr., 2007). Recent studies have also indi-
cated that metabolic products derived from u-3 FAs, such as
SUPPLEMENTAL INFORMATION
17S-hydroxy-DHA, resolvins, and protectins may play a role in
the long term resolution of inflammation and this might attenuate Supplemental Information includes Extended Experimental Procedures, seven
insulin resistance in the context of obesity (González-Périz et al., figures, and two tables and can be found with this article online at doi:10.1016/
2009). If this proves to be correct, then this could provide an j.cell.2010.07.041.
additional mechanism for long term u-3 FA-induced anti-inflam-
matory, insulin sensitizing effects. However, in the current ACKNOWLEDGMENTS

studies, we found that these u-3 FA derivatives were unable to


We thank Jachelle M. Ofrecio and Sarah Nalbandian for their help with animal
stimulate GPR120 activation in our reporter cell assay (data not maintenance and Elizabeth J. Hansen for editorial assistance. We are grateful
shown), indicating that GPR120 functions as a receptor for u-3 to Dr. Robert Lefkowitz (Howard Hughes Medical Institute, Duke University) for
FAs and not their biochemical products. Resolution of inflamma- the gift of FLAG-tagged serial mutant b-arrestin2 constructs and to Dr. Maziyar

696 Cell 142, 687–698, September 3, 2010 ª2010 Elsevier Inc.


Saberi at NGM Bio Inc. (San Francisco, CA) for GLP-1 measurements. We Jucker, B.M., Cline, G.W., Barucci, N., and Shulman, G.I. (1999). Differential
thank the Flow Cytometry Resource and Neal Sekiya for assistance with effects of safflower oil versus fish oil feeding on insulin-stimulated glycogen
FACS analysis at the VA San Diego hospital, the UCSD Histology Core lab synthesis, glycolysis, and pyruvate dehydrogenase flux in skeletal muscle:
for technical help with processing liver specimens, and UCSD Microscope a 13C nuclear magnetic resonance study. Diabetes 48, 134–140.
Resource for microscopy analysis. This study was funded in part by the
Kawai, T., and Akira, S. (2006). TLR signaling. Cell Death Differ. 13, 816–825.
National Institutes of Health grants NIDDK DK033651 (J.M.O.), DK063491
(J.M.O.), DK 074868 (J.M.O.), and the Eunice Kennedy Shriver NICHD/NIH Lee, J.Y., Plakidas, A., Lee, W.H., Heikkinen, A., Chanmugam, P., Bray, G., and
through a cooperative agreement U54 HD 012303-25 as part of the specialized Hwang, D.H. (2003). Differential modulation of Toll-like receptors by fatty
Cooperative Centers Program in Reproduction and Infertility Research. acids: preferential inhibition by n-3 polyunsaturated fatty acids. J. Lipid Res.
44, 479–486.
Received: January 20, 2010 Lumeng, C.N., DelProposto, J.B., Westcott, D.J., and Saltiel, A.R. (2008).
Revised: May 24, 2010 Phenotypic switching of adipose tissue macrophages with obesity is gener-
Accepted: July 19, 2010 ated by spatiotemporal differences in macrophage subtypes. Diabetes 57,
Published: September 2, 2010 3239–3246.
Luttrell, L.M., Ferguson, S.S., Daaka, Y., Miller, W.E., Maudsley, S., Della
REFERENCES Rocca, G.J., Lin, F., Kawakatsu, H., Owada, K., Luttrell, D.K., et al. (1999).
Beta-arrestin-dependent formation of beta2 adrenergic receptor-Src protein
Barak, L.S., Ferguson, S.S., Zhang, J., and Caron, M.G. (1997). A beta- kinase complexes. Science 283, 655–661.
arrestin/green fluorescent protein biosensor for detecting G protein-coupled Luttrell, L.M., and Lefkowitz, R.J. (2002). The role of beta-arrestins in the
receptor activation. J. Biol. Chem. 272, 27497–27500. termination and transduction of G-protein-coupled receptor signals. J. Cell
Baron, A.D., Brechtel, G., Wallace, P., and Edelman, S.V. (1988). Rates and Sci. 115, 455–465.
tissue sites of non-insulin- and insulin-mediated glucose uptake in humans. Miller, W.E., and Lefkowitz, R.J. (2001). Expanding roles for beta-arrestins as
Am. J. Physiol. 255, E769–E774. scaffolds and adapters in GPCR signaling and trafficking. Curr. Opin. Cell Biol.
Briscoe, C.P., Peat, A.J., McKeown, S.C., Corbett, D.F., Goetz, A.S., Littleton, 13, 139–145.
T.R., McCoy, D.C., Kenakin, T.P., Andrews, J.L., Ammala, C., et al. (2006).
Neschen, S., Morino, K., Dong, J., Wang-Fischer, Y., Cline, G.W., Romanelli,
Pharmacological regulation of insulin secretion in MIN6 cells through the fatty
A.J., Rossbacher, J.C., Moore, I.K., Regittnig, W., Munoz, D.S., et al. (2007).
acid receptor GPR40: identification of agonist and antagonist small molecules.
n-3 Fatty acids preserve insulin sensitivity in vivo in a peroxisome
Br. J. Pharmacol. 148, 619–628.
proliferator-activated receptor-alpha-dependent manner. Diabetes 56,
Buettner, R., Parhofer, K.G., Woenckhaus, M., Wrede, C.E., Kunz-Schughart, 1034–1041.
L.A., Scholmerich, J., and Bollheimer, L.C. (2006). Defining high-fat-diet rat
Nguyen, M.T., Favelyukis, S., Nguyen, A.K., Reichart, D., Scott, P.A., Jenn, A.,
models: metabolic and molecular effects of different fat types. J. Mol. Endocri-
Liu-Bryan, R., Glass, C.K., Neels, J.G., and Olefsky, J.M. (2007). A subpopula-
nol. 36, 485–501.
tion of macrophages infiltrates hypertrophic adipose tissue and is activated by
Calder, P.C. (2005). Polyunsaturated fatty acids and inflammation. Biochem. free fatty acids via Toll-like receptors 2 and 4 and JNK-dependent pathways. J.
Soc. Trans. 33, 423–427. Biol. Chem. 282, 35279–35292.
Cen, B., Xiong, Y., Ma, L., and Pei, G. (2001). Direct and differential interaction Oh, D.Y., Song, J.A., Moon, J.S., Moon, M.J., Kim, J.I., Kim, K., Kwon, H.B.,
of beta-arrestins with the intracellular domains of different opioid receptors. and Seong, J.Y. (2005). Membrane-proximal region of the carboxyl terminus
Mol. Pharmacol. 59, 758–764. of the gonadotropin-releasing hormone receptor (GnRHR) confers differential
Gao, H., Sun, Y., Wu, Y., Luan, B., Wang, Y., Qu, B., and Pei, G. (2004). signal transduction between mammalian and nonmammalian GnRHRs. Mol.
Identification of beta-arrestin2 as a G protein-coupled receptor-stimulated Endocrinol. 19, 722–731.
regulator of NF-kappaB pathways. Mol. Cell 14, 303–317. Polozova, A., and Salem, N., Jr. (2007). Role of liver and plasma lipoproteins
Gether, U. (2000). Uncovering molecular mechanisms involved in activation of in selective transport of n-3 fatty acids to tissues: a comparative study of
G protein-coupled receptors. Endocr. Rev. 21, 90–113. 14C-DHA and 3H-oleic acid tracers. J. Mol. Neurosci. 33, 56–66.
González-Périz, A., Horrillo, R., Ferre, N., Gronert, K., Dong, B., Moran- Schenk, S., Saberi, M., and Olefsky, J.M. (2008). Insulin sensitivity: modulation
Salvador, E., Titos, E., Martinez-Clemente, M., Lopez-Parra, M., Arroyo, V., by nutrients and inflammation. J. Clin. Invest. 118, 2992–3002.
and Claria, J. (2009). Obesity-induced insulin resistance and hepatic steatosis
Schulte, G., and Fredholm, B.B. (2003). Signaling from adenosine receptors to
are alleviated by omega-3 fatty acids: a role for resolvins and protectins.
mitogen-activated protein kinases. Cell Signal 15, 813–827.
FASEB J. 23, 1946–1957.
Shoelson, S.E., Herrero, L., and Naaz, A. (2007). Obesity, inflammation, and
Gotoh, C., Hong, Y.H., Iga, T., Hishikawa, D., Suzuki, Y., Song, S.H., Choi,
insulin resistance. Gastroenterology 132, 2169–2180.
K.C., Adachi, T., Hirasawa, A., Tsujimoto, G., et al. (2007). The regulation of
adipogenesis through GPR120. Biochem. Biophys. Res. Commun. 354, Solinas, G., Vilcu, C., Neels, J.G., Bandyopadhyay, G.K., Luo, J.L., Naugler,
591–597. W., Grivennikov, S., Wynshaw-Boris, A., Scadeng, M., Olefsky, J.M., and
Karin, M. (2007). JNK in hematopoietically derived cells contributes to diet-
Hirasawa, A., Tsumaya, K., Awaji, T., Katsuma, S., Adachi, T., Yamada, M.,
induced inflammation and insulin resistance without affecting obesity. Cell
Sugimoto, Y., Miyazaki, S., and Tsujimoto, G. (2005). Free fatty acids regulate
Metab. 6, 386–397.
gut incretin glucagon-like peptide-1 secretion through GPR120. Nat. Med. 11,
90–94. Takaesu, G., Surabhi, R.M., Park, K.J., Ninomiya-Tsuji, J., Matsumoto, K., and
Gaynor, R.B. (2003). TAK1 is critical for IkappaB kinase-mediated activation of
Imamura, T., Vollenweider, P., Egawa, K., Clodi, M., Ishibashi, K., Nakashima,
the NF-kappaB pathway. J. Mol. Biol. 326, 105–115.
N., Ugi, S., Adams, J.W., Brown, J.H., and Olefsky, J.M. (1999). G alpha-q/11
protein plays a key role in insulin-induced glucose transport in 3T3-L1 adipo- Tazoe, H., Otomo, Y., Kaji, I., Tanaka, R., Karaki, S.I., and Kuwahara, A. (2008).
cytes. Mol. Cell. Biol. 19, 6765–6774. Roles of short-chain fatty acids receptors, GPR41 and GPR43 on colonic func-
Itoh, Y., Kawamata, Y., Harada, M., Kobayashi, M., Fujii, R., Fukusumi, S., Ogi, tions. J. Physiol. Pharmacol. 59 (Suppl 2), 251–262.
K., Hosoya, M., Tanaka, Y., Uejima, H., et al. (2003). Free fatty acids regulate Ulloa-Aguirre, A., Stanislaus, D., Janovick, J.A., and Conn, P.M. (1999).
insulin secretion from pancreatic beta cells through GPR40. Nature 422, Structure-activity relationships of G protein-coupled receptors. Arch. Med.
173–176. Res. 30, 420–435.

Cell 142, 687–698, September 3, 2010 ª2010 Elsevier Inc. 697


Wang, J., Wu, X., Simonavicius, N., Tian, H., and Ling, L. (2006a). Medium- Xiao, K., McClatchy, D.B., Shukla, A.K., Zhao, Y., Chen, M., Shenoy, S.K.,
chain fatty acids as ligands for orphan G protein-coupled receptor GPR84. Yates, J.R., 3rd, and Lefkowitz, R.J. (2007). Functional specialization of
J. Biol. Chem. 281, 34457–34464. beta-arrestin interactions revealed by proteomic analysis. Proc. Natl. Acad.
Wang, Y., Tang, Y., Teng, L., Wu, Y., Zhao, X., and Pei, G. (2006b). Association Sci. USA 104, 12011–12016.
of beta-arrestin and TRAF6 negatively regulates Toll-like receptor-interleukin 1
receptor signaling. Nat. Immunol. 7, 139–147. Xu, H., Barnes, G.T., Yang, Q., Tan, G., Yang, D., Chou, C.J., Sole, J., Nichols,
Weisberg, S.P., McCann, D., Desai, M., Rosenbaum, M., Leibel, R.L., and A., Ross, J.S., Tartaglia, L.A., and Chen, H. (2003). Chronic inflammation in fat
Ferrante, A.W., Jr. (2003). Obesity is associated with macrophage accumula- plays a crucial role in the development of obesity-related insulin resistance. J.
tion in adipose tissue. J. Clin. Invest. 112, 1796–1808. Clin. Invest. 112, 1821–1830.

698 Cell 142, 687–698, September 3, 2010 ª2010 Elsevier Inc.


Anti-CD47 Antibody Synergizes with
Rituximab to Promote Phagocytosis and
Eradicate Non-Hodgkin Lymphoma
Mark P. Chao,1,10,* Ash A. Alizadeh,1,2,3,10 Chad Tang,1 June H. Myklebust,3,9 Bindu Varghese,3 Saar Gill,5 Max Jan,1
Adriel C. Cha,1 Charles K. Chan,1 Brent T. Tan,4 Christopher Y. Park,1,4 Feifei Zhao,1 Holbrook E. Kohrt,2,3
Raquel Malumbres,6 Javier Briones,7 Randy D. Gascoyne,8 Izidore S. Lossos,6 Ronald Levy,3 Irving L. Weissman,1,4,10
and Ravindra Majeti1,2,10
1Institutefor Stem Cell Biology and Regenerative Medicine, Stanford Cancer Center, and Ludwig Center at Stanford
2Department of Internal Medicine, Division of Hematology
3Division of Oncology
4Department of Pathology
5Division of Blood and Bone Marrow Transplantation

Stanford University, Palo Alto, CA 94304, USA


6Department of Medicine, Division of Hematology-Oncology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
7Department of Hematology, Hospital Santa Creu i Sant Pau, Autonomous University of Barcelona, Barcelona 08193, Spain
8Department of Pathology, British Columbia Cancer Agency, Vancouver V5Z 1L3, Canada
9Department of Immunology, Institute for Cancer Research, Oslo University Hospital/Centre for Cancer Biomedicine, University of Oslo,

Oslo 0310, Norway


10These authors contributed equally to this work

*Correspondence: mpchao@stanford.edu
DOI 10.1016/j.cell.2010.07.044

SUMMARY modality for cancer treatment (Adams and Weiner, 2005).


Although therapies combining a mAb with chemotherapeutic
Monoclonal antibodies are standard therapeutics for agents are effective in several human cancers, antibodies alone
several cancers including the anti-CD20 antibody are not curative. Antibodies effective against cancer are believed
rituximab for B cell non-Hodgkin lymphoma (NHL). to function by several mechanisms including antibody-depen-
Rituximab and other antibodies are not curative dent cellular cytotoxicity (ADCC), stimulation of complement-
and must be combined with cytotoxic chemotherapy dependent cytotoxicity (CDC), inhibition of signal transduction,
or direct induction of apoptosis (Cheson and Leonard, 2008).
for clinical benefit. Here we report the eradication
Non-Hodgkin lymphoma (NHL) is the fifth most common
of human NHL solely with a monoclonal antibody cancer in the United States consisting of indolent and aggressive
therapy combining rituximab with a blocking anti- subtypes with a 5 year overall survival ranging from 25%–75%
CD47 antibody. We identified increased expression (The International Non-Hodgkin’s Lymphoma Prognostic
of CD47 on human NHL cells and determined that Factors Project, 1993). The anti-CD20 antibody, rituximab (Rit-
higher CD47 expression independently predicted uxan), is a standard therapy for many CD20-positive B cell
adverse clinical outcomes in multiple NHL subtypes. lymphomas and significantly improves long-term survival in
Blocking anti-CD47 antibodies preferentially enabled combination with conventional chemotherapy (Cheson and Leo-
phagocytosis of NHL cells and synergized with ritux- nard, 2008). As a single agent or in combination with chemo-
imab. Treatment of human NHL-engrafted mice with therapy, rituximab is not curative in the majority of B cell NHL
anti-CD47 antibody reduced lymphoma burden and patients and rituximab resistance has been observed (reviewed
in Cheson and Leonard, 2008). Multiple lines of evidence indicate
improved survival, while combination treatment
that rituximab acts at least in part by engaging Fc receptors
with rituximab led to elimination of lymphoma and (FcRs) on immune effector cells, such as NK cells and macro-
cure. These antibodies synergized through a mecha- phages, and stimulating effector functions such as ADCC (Glen-
nism combining Fc receptor (FcR)-dependent and nie et al., 2007; Nimmerjahn and Ravetch, 2007). Although resis-
FcR-independent stimulation of phagocytosis that tance has been reported to occur through several mechanisms
might be applicable to many other cancers. (Cartron et al., 2004), there has been limited development of
agents that can overcome this resistance.
INTRODUCTION Immune effector cells, including NK cells and phagocytes, are
critical to the efficacy of many anticancer antibodies. Phagocytic
Emerging evidence has demonstrated that monoclonal anti- cells, including macrophages and dendritic cells, express signal
bodies (mAbs) either alone or in combination are an effective regulatory protein alpha (SIRPa), which binds CD47, a widely

Cell 142, 699–713, September 3, 2010 ª2010 Elsevier Inc. 699


expressed transmembrane protein (Brown and Frazier, 2001). based on the presumed cell of origin of tumors: normal germinal
CD47-mediated activation of SIRPa initiates a signal transduc- center B cells (GCB-like), which are associated with a favorable
tion cascade resulting in inhibition of phagocytosis (reviewed in clinical outcome, or activated blood memory B cells (ABC-like),
Jaiswal et al., 2010). In identifying a role for CD47 in cancer path- which are associated with a poor clinical outcome (Alizadeh
ogenesis, we previously demonstrated that forced expression of et al., 2000; Rosenwald et al., 2002). CD47 expression was sig-
mouse CD47 on a human leukemia cell line facilitated tumor nificantly higher in ABC-like DLBCL (Figure 1F and Figure S1E).
engraftment in immunodeficient mice through the evasion of CD47 expression was not found to have independent prog-
phagocytosis (Jaiswal et al., 2009). We further demonstrated nostic value within GCB and ABC subtypes, suggesting a strong
that this mechanism could be targeted therapeutically in human association with the cell-of-origin classification of DLBCL.
acute myeloid leukemia (AML) with a blocking anti-CD47 anti- Higher CD47 expression was also associated with unmutated
body that enabled phagocytosis and eliminated AML stem cells immunoglobulin heavy chain variable regions (IgVH) in CLL
(Majeti et al., 2009). Based on this antibody mechanism, we (Figure 1G and Figure S1E) and significantly correlated with
hypothesized that the combination of a blocking anti-CD47 the proliferative index in MCL (Figure 1H), both known adverse
antibody with a second FcR-activating antibody would both prognostic factors (Katzenberger et al., 2006; Oscier et al.,
prevent an inhibitory signal and deliver a positive stimulus, 2002; Rosenwald et al., 2003). In multivariate analyses, CD47
resulting in the synergistic phagocytosis and elimination of tar- expression remained prognostic of disease progression inde-
get cells. Here, we tested this antibody synergy hypothesis by pendent from the international prognostic index in DLBCL (The
investigating the combination of a blocking anti-CD47 mAb International Non-Hodgkin’s Lymphoma Prognostic Factors
with rituximab against human NHL. Project, 1993) and two major prognostic factors in CLL: IgVH
mutation status and ZAP-70 status (Figure S1G). Within the
RESULTS small MCL cohort, a multivariate model did not find independent
prognostic value for CD47 when considering the proliferation
CD47 Expression Is Increased on NHL Cells Compared index (data not shown).
to Normal B Cells
We examined CD47 protein expression on primary human NHL Blocking Anti-CD47 Antibodies Enable Phagocytosis
samples and normal B cells by flow cytometry. Compared to of NHL Cells by Macrophages and Synergize
both normal peripheral blood and germinal center B cells, with Rituximab In Vitro
CD47 was more highly expressed on a large subset of primary We first tested the ability of anti-human CD47 antibodies to
patient samples from multiple B cell NHL subtypes (Figure 1A enable phagocytosis of human NHL cell lines, primary NHL cells,
and Figure S1A available online), including diffuse large B cell and normal peripheral blood (NPB) cells by human macrophages
lymphoma (DLBCL), B cell chronic lymphocytic leukemia in vitro. Incubation of NHL cells in the presence of IgG1 isotype
(B-CLL), mantle cell lymphoma (MCL), follicular lymphoma (FL), control or anti-CD45 IgG1 antibody did not result in significant
marginal zone lymphoma (MZL), and pre-B acute lymphoblastic phagocytosis; however, two different blocking anti-CD47 anti-
leukemia (pre-B ALL). Across NHL subtypes, we found differing bodies (B6H12.2 and BRIC126) enabled phagocytosis of NHL
levels of CD47 expression that also varied within each NHL cells but not NPB cells (Figures 2A and 2B).
subtype (Figure 1B). Next, we repeated the in vitro phagocytosis assays with
mouse macrophages. Incubation of NHL cells in the presence
Increased CD47 Expression Correlates with a Worse of IgG1 isotype control or anti-CD45 IgG1 antibody did not result
Clinical Prognosis and Adverse Molecular Features in significant phagocytosis; however, phagocytosis of NHL cells
in Multiple NHL Subtypes was observed with blocking antibodies to CD47 (B6H12.2 and
Having previously shown a correlation between CD47 mRNA BRIC126), whereas no phagocytosis was observed with a non-
and protein expression (Majeti et al., 2009), we assessed CD47 blocking antibody (2D3) (Figure 2B). Disruption of the CD47-
mRNA expression across NHL subtypes for associations with SIRPa interaction with an anti-mouse SIRPa antibody also
morphologic and molecular subgroups using gene expression resulted in significant phagocytosis (Figure 2B).
data from previously described patient cohorts (Table S1) and Given variable expression of CD47 on primary NHL, we inves-
investigated the prognostic implications of increased CD47 tigated by two independent methods whether CD47 expression
expression in disease outcome. Higher CD47 expression was levels correlated with the degree of anti-CD47 antibody-medi-
associated with adverse prognosis in DLBCL, B-CLL, and MCL ated phagocytosis. First, lentiviral shRNA vectors were used to
(Figures 1C–1E). In patients with DLBCL, whether treated with knock down expression of CD47 in Raji cells. Several clones
or without rituximab-based combination chemotherapy (Fig- were generated with a range of CD47 knockdown (Figures S2A
ure 1C and Figure S1B), higher CD47 expression was signifi- and S2B). Those clones with a greater than 50% reduction in
cantly associated with risk of death. This increased risk was CD47 expression (shCD47-1 and shCD47-2) demonstrated
largely due to disease progression (Figure S1C), a finding vali- a significant reduction in anti-CD47 antibody-mediated phago-
dated in an independent cohort of patients using quantitative cytosis (Figure S3C). In the second approach, a statistical anal-
RT-PCR on fixed archival specimens (Figure S1D). ysis demonstrated a positive correlation between CD47 expres-
We next investigated whether increased CD47 expression was sion and degree of anti-CD47 antibody-mediated phagocytosis
associated with known adverse molecular features in NHL. with both mouse and human macrophage effector cells
In DLBCL, prior studies have identified two distinct subgroups (Figure S2D).

700 Cell 142, 699–713, September 3, 2010 ª2010 Elsevier Inc.


Figure 1. CD47 Expression Is Increased on NHL Cells Compared to Normal B Cells, Confers a Worse Clinical Prognosis, and Correlates with
Adverse Molecular Features in Multiple NHL Subtypes
(A) CD47 expression on normal peripheral blood (PB) B cells (CD19+), normal germinal center (GC) B cells (CD3CD5CD20+CD10+CD38+), and NHL B cells
(CD19+) was determined by flow cytometry, and mean fluorescence intensity was normalized for cell size. Each point represents a different patient sample:
DLBCL = 2, CLL = 15, MCL = 4, FL = 6, MZL = 2, and pre-B ALL = 1 (****p < 0.0001). Normalized mean expression (and range) for each population were: normal
PB B cells 420.9 (267.3–654.0), normal GC B cells 482.5 (441.1–519.9), and NHL 888.5 (270.1–1553).
(B) CD47 expression across NHL subtypes including DLBCL (DL, n = 15), MCL (n = 34), FL (n = 28), and B-CLL (n = 14) was determined as in (A). Normalized mean
expression (and range) for each population were: DL 725.7 (261.2–1344), MCL 1055 (444.2–2196), FL 825.1 (283.6–1546), CLL 713.6 (432.8–1086) (*p < 0.05).
(C–E) Prognostic influence of CD47 mRNA expression is shown on overall (C and E) and event-free (D) survival of patients with DLBCL, B-CLL, and MCL.
For DLBCL and CLL, stratification into low and high CD47 expression groups was based on an optimal threshold determined by microarray analysis; this cutpoint
was internally validated for both DLBCL and CLL and also externally validated in an independent DLBCL cohort. For MCL, stratification relative to the median was
employed as an optimal cutpoint could not be defined. Significance measures are based on log-likelihood estimates of the p value, when treating CD47 expres-
sion as a continuous variable, with corresponding dichotomous indices also provided in Table S1.
(F–H) CD47 mRNA expression is shown in relation to cell-of-origin classification for DLBCL (F), immunoglobulin heavy chain mutation status (IgVH) for CLL (G),
and proliferation index for MCL (H). Error bars represent upper and lower quartiles (F and G). Analyses for (C)–(H) employed publicly available datasets for NHL
patients (Table S1). NGC = normal germinal center, ABC = activated B cell-like, GCB = germinal center B cell-like.
See also Figure S1 and Table S1.

Cell 142, 699–713, September 3, 2010 ª2010 Elsevier Inc. 701


Figure 2. Blocking Antibodies against CD47 Enable Phagocytosis of NHL Cells by Macrophages and Synergize with Rituximab In Vitro
(A) CFSE-labeled NHL cells were incubated with human macrophages and the indicated antibodies and examined by immunofluorescence microscopy to detect
phagocytosis (arrows). Photomicrographs from a representative NHL sample are shown.
(B) Phagocytic indices of primary human NHL cells (blue), normal peripheral blood (NPB) cells (red), and NHL cell lines (purple, orange, and green) were deter-
mined using human (left) and mouse (middle) macrophages. Antibody-induced apoptosis (right panel) was tested by incubating NHL cells with the indicated anti-
bodies or staurosporine without macrophages and assessing the percentage of apoptotic and dead cells (% annexin V and/or PI positive).

702 Cell 142, 699–713, September 3, 2010 ª2010 Elsevier Inc.


It has been reported that immobilized or crosslinked anti- planted into SCID mice (Figure S3). In addition to these cell lines,
bodies against CD47 induce apoptosis of primary human we identified a primary NHL patient specimen that engrafted in
B-CLL cells, as well as several malignant lymphoid cell lines NSG mice in the bone marrow upon intravenous transplantation
(Kikuchi et al., 2004, 2005; Mateo et al., 1999; Uno et al., (Figures S5A and S5B). As with the cell lines, ex vivo coating of
2007). Therefore, anti-CD47 antibodies might be predicted to these primary cells with anti-CD47 antibody, but not controls
directly induce apoptosis of NHL cells that are then recognized (Figure 3G), resulted in complete inhibition of bone marrow
by macrophages and phagocytosed. Contrary to this prediction, engraftment (Figure 3H).
when NHL cells were incubated with anti-CD47 antibody in
the absence of macrophages, no induction of apoptosis was Combination Therapy with Anti-CD47 Antibody and
observed when cells were incubated in suspension for either Rituximab Eliminates Lymphoma in Both Disseminated
2 hr (Figure 2B, right) or 8 hr (Figures S2E and S2F). Incubation and Localized Human NHL Xenotransplant Models
of NHL cells with immobilized anti-CD47 antibody resulted in In the second treatment strategy, mice were first engrafted with
increased apoptosis compared to controls (Figures S2G and NHL and then administered single or combination antibody
S2H), consistent with prior reports (Mateo et al., 1999). Given therapy. To best model NHL, we established disseminated and
that phagocytosis of NHL cells occurs in the presence of soluble localized xenotransplantation models in NSG mice that are defi-
anti-CD47 mAbs, it is unlikely that these mAbs induce apoptosis cient in T, B, and NK cells (Shultz et al., 2005) but retain phago-
of NHL cells that are then secondarily phagocytosed. cyte effector cells. In the disseminated model, luciferase-
Next, we tested the ability of a blocking anti-CD47 mAb to syn- expressing Raji cells were transplanted intravenously into adult
ergize with rituximab in the phagocytosis of NHL cells. We exam- NSG mice. Two weeks later, these mice were administered daily
ined CD20 expression on NHL cells and found no difference injections of either control mouse IgG, anti-CD47 antibody, ritux-
between normal B cells and NHL cells (Figures S2I and S2J). imab, or anti-CD47 antibody and rituximab. Anti-CD47 antibody
Incubation of NHL cells with rituximab in the presence of mouse treatment decreased the lymphoma burden in these mice (Fig-
or human macrophages resulted in significant phagocytosis ures 4A and 4B) and significantly prolonged survival compared
(Figures 2D and S2E). We then tested the synergy hypothesis to control IgG, although all mice eventually died (Figure 4C and
by isobologram analysis (Chou, 2008; Tallarida, 2006). Using Table S2). Similar results were seen with rituximab and were
Raji, SUDHL4, and NHL17 cells, which express varying levels not statistically different compared to anti-CD47 antibody (Fig-
of both CD47 and CD20 (Figure S2K), anti-CD47 antibody ures 4A–4C and Table S2). In contrast, combination therapy
synergized with rituximab or anti-human CD20 (mouse IgG2a) with anti-CD47 antibody and rituximab eliminated lymphoma in
antibody, as indicated by combination indices less than 1 (Fig- 60% of mice as indicated by long-term survival (Figure 4C) and
ure 2C). In a second approach, in vitro phagocytosis assays the absence of luciferase-positive lymphoma (data not shown)
were conducted with primary NHL cells incubated with either more than 4 months after the end of treatment. In humans, ritux-
anti-CD47 antibody or rituximab alone, or both in combination imab efficacy is thought to be primarily mediated by both macro-
at half of the single agent dose. NHL cells exhibited a significant phages and NK cells (Nimmerjahn and Ravetch, 2007; Taylor
increase in phagocytosis when incubated with the combination and Lindorfer, 2008). Given that NSG mice lack NK cells, we con-
compared to either antibody alone when using mouse (Fig- ducted a similar experiment in NK cell-containing SCID mice and
ure 2D) or human (Figure 2E) macrophage effectors. No phago- observed similar therapeutic responses as in NSG mice (Figures
cytosis of NPB cells was observed with either antibody alone or S4A and S4B).
in combination with human macrophages (Figure 2E). In the localized NHL model, luciferase-expressing Raji cells
were transplanted subcutaneously in the right flank of NSG
Ex Vivo Coating of NHL Cells with an Anti-CD47 Antibody mice. Once tumors were palpable (approximately 2 weeks),
Inhibits Tumor Engraftment mice were treated with antibody therapy. Mice treated with
Next, the ability of blocking anti-CD47 antibody to eliminate NHL anti-CD47 antibody or rituximab demonstrated a decreased
in vivo either alone or in combination with rituximab was explored rate of lymphoma growth compared to control IgG-treated
by two separate treatment strategies. First, the effect of ex vivo mice as measured by both luciferase signal and tumor volume
anti-CD47 antibody coating on the engraftment of human NHL (Figures 4D–4F) but, like controls, eventually had to be sacrificed
cells was tested. Luciferase-expressing Raji and SUDHL4 cell due to enlarged tumor growth. In contrast, mice treated with the
lines were precoated ex vivo with anti-CD47, IgG1 isotype combination of anti-CD47 antibody and rituximab demonstrated
control, or anti-CD45 antibody and transplanted intravenously complete elimination of lymphoma, with 86% of treated mice
into SCID mice. Coating with anti-CD47 antibody prevented having no measurable mass or luciferase-positive lymphoma
engraftment of both cell lines (Figures 3A–3F). Coating of Raji 4 weeks after the end of therapy (Figures 4D–4F and Figures
cells with rituximab also inhibited engraftment when trans- S4C–S4E). Moreover, all showed no evidence of tumor growth,

(C) Synergistic phagocytosis by anti-CD47 antibody (B6H12.2) and anti-CD20 mAbs was examined by isobologram analysis and determination of combination
indices (CI). CIobs indicates observed results, and the dashed line indicates the expected results if antibodies were additive.
(D and E) Synergy between anti-CD47 antibody and rituximab in the phagocytosis of NHL and NPB cells was assessed by determining the phagocytic index when
incubated with a combination of both antibodies compared to either antibody alone at twice the dose, with either mouse (D) or human (E) macrophages. NHL17*:
cell line derived from primary sample NHL17.
***p < 0.001, ****p < 0.0001, *****p < 0.00001. Figure 2B p values represent comparison against IgG1 isotype control antibody. See also Figure S2.

Cell 142, 699–713, September 3, 2010 ª2010 Elsevier Inc. 703


Figure 3. Ex Vivo Coating of NHL Cells with an Anti-CD47 Antibody Inhibits Tumor Engraftment
(A–F) Luciferase-expressing Raji (A) and SUDHL4 (C) cells were assessed for ex vivo antibody coating by flow cytometry. SCID mice transplanted with Raji (B) and
SUDHL4 (D) were subject to bioluminescent imaging (1-IgG1 isotype control, 2-anti-CD45, 3 and 4-anti-CD47, 5-luciferase negative control). Bioluminescence
for Raji (E) and SUDHL4 (F) engrafted mice was quantified (n = 3 per antibody condition). No tumor engraftment was observed in mice transplanted with anti-
CD47-coated cells compared to IgG-coated cells (p < 0.05) for both Raji and SUDHL4, as assessed by bioluminescent imaging. Data are represented as
mean ± standard deviation (SD).

704 Cell 142, 699–713, September 3, 2010 ª2010 Elsevier Inc.


remained relapse free, and were alive at over 197 days after different anti-CD47 antibody potencies in distinct tissue com-
tumor engraftment. Out of six mice achieving a complete remis- partments (peripheral blood versus bone marrow (BM) versus
sion, five remained relapse free whereas one mouse died of non- soft tissue compartments).
tumor-related causes (Figure 4E). For both disseminated and To assess the ability of these two primary NHL xenotransplant
localized xenograft models, expression of CD47 and CD20 in models to model the disease, we compared histological sections
transplanted Raji cells did not differ from Raji cells in culture of the primary NHL specimen and the transplanted tumor. Similar
(Figure S4F). DLBCL and FL morphology was observed for NHL7 and NHL31,
respectively (Figure S5B). We next determined whether the per-
Combination Therapy with Anti-CD47 Antibody centages of macrophages infiltrating the tumor differed between
and Rituximab Eliminates Lymphoma in Primary Human the primary patient and the xenografted tumor. For NHL31, the
NHL Xenotransplant Mouse Models percentage of infiltrating human macrophages (CD68+) in the
NHL cell lines have been valuable for the evaluation of therapeu- primary lymph node was similar to the percentage of infiltrating
tics, but they may not accurately recapitulate the heterogeneity mouse macrophages (F4/80+) in bone marrow of transplanted
of the primary disease. We report here two new mouse xenograft mice (Figure S5C). Analyzing infiltrating macrophage frequency
models for NHL in which intravenous transplantation of cells by flow cytometry, no difference was observed between the
from a DLBCL patient (NHL7/SUNHL7) and a FL patient primary specimen and xenograft for either NHL7 or NHL31
(NHL31/SUNHL31) give rise to robust lymphoma engraftment (Figure S5D).
in the bone marrow and/or peripheral blood (Figure S1A and
Figure S5A). Primary DLBCL cells were transplanted into mice, Synergy between Anti-CD47 Antibody and Rituximab
which 2 weeks later were treated with daily injections of anti- Does Not Occur through NK Cells or Complement
bodies for 14 days. Treatment with anti-CD47 antibody either Rituximab can eliminate malignant cells via apoptosis, NK cell-
alone or in combination with rituximab eliminated human mediated ADCC, and CDC (reviewed in Smith, 2003). However,
lymphoma in the bone marrow, whereas treatment with rituxi- it is not known whether anti-CD47 antibody also enables ADCC
mab resulted in a reduction of disease in 60% of mice (Figures or CDC in addition to phagocytosis. Therefore, we investigated
5A and 5B). Mouse hematopoietic cells were unaffected by anti- whether anti-CD47 antibody alone could induce antitumor
body therapy (Figure S5E). Treatment was then discontinued, effects by macrophage-independent mechanisms, and whether
and all mice were monitored for survival. Mice treated with either synergy with rituximab could occur through these modalities.
anti-CD47 antibody or rituximab alone had a significantly longer First, to investigate possible synergy in direct apoptosis, NHL
survival compared to mice treated with control IgG, but all even- cells were incubated with either anti-CD47 antibody/rituximab
tually died secondary to disease due to widespread organ alone or in combination without macrophages, and cell death
dissemination on autopsy (Figure 5C, data not shown). Most was measured. No synergistic apoptosis was observed when
significantly, 8 out of 9 mice (89%) administered combination NHL cells were incubated with soluble (Figure 6A and Fig-
antibody treatment were cured of lymphoma, as indicated by ure S6A) or immobilized (Figures S6B and S6C) antibodies.
long-term disease-free survival more than 4 months after the Furthermore, crosslinking of soluble anti-CD47 antibody alone
end of treatment (Figure 5C and Table S3) with no detectable or in combination with rituximab by macrophages did not induce
lymphoma in the bone marrow (data not shown). In a second increased apoptosis of nonphagocytosed NHL cells compared
primary model, FL cells were transplanted intravenously in a to IgG1 isotype control, whereas rituximab induced a slight
similar manner. CD20+CD10+ lymphoma engraftment in the increase in apoptosis (Figure S6D). No synergistic apoptosis
peripheral blood and bone marrow was detected after 8 weeks. was observed in this context (Figure S6D). The small increase
At this time, mice were treated with a single intraperitoneal injec- in apoptosis upon antibody treatment was not FcR dependent
tion of either control IgG, anti-CD47 antibody, rituximab, or the given that results were similar with macrophages lacking the
combination and then followed for disease progression. A single Fcg subunit (Takai et al., 1994) (Figure S6D). Second, we inves-
dose of anti-CD47 antibody alone or in combination with rituxi- tigated whether NK cells could mediate tumor elimination by
mab eliminated lymphoma both in the peripheral blood (Fig- anti-CD47 antibody alone or in synergy with rituximab. A prior
ure 5D) and in bone marrow (Figure 5E). In contrast, a single report observed increased NK cell cytotoxicity of cancer cell
dose of rituximab enabled a partial reduction in tumor burden lines with an anti-CD47 antibody in vitro, though the mechanism
that rebounded back to baseline levels in the peripheral blood of targeting was not elucidated (Kim et al., 2008). We first deter-
with no tumor reduction observed in the bone marrow. The differ- mined whether human or mouse NK cells expressed SIRPa and
ence in anti-CD47 antibody clearance of lymphoma as a single found that both human NK cells, CD3CD56+CD7+ (Milush et al.,
dose in FL-engrafted mice (Figures 5D and 5E) compared to 2009), as well as mouse NK cells, CD3DX5+, expressed minimal
multiple dose therapy in mice engrafted with DLBCL (Fig- to no SIRPa (Figure 6B). Next, the ability of anti-CD47 antibody
ure 5B) or Raji (Figure 4) may be due to cell-intrinsic differences to induce NK cell-mediated ADCC through FcRs or by CD47-
in antibody sensitivity between different NHL subtypes or due to SIRPa blockade was investigated. Utilizing human NK cells as

(G) Bulk lymphoma cells from a human NHL patient were assessed for ex vivo antibody coating by flow cytometry.
(H) Compared to IgG1 isotype control, anti-CD47 antibody pretreatment inhibited engraftment of NHL cells (p < 0.0001) whereas anti-CD45-coated cells
engrafted similarly to controls (p = 0.54). All p values were determined using Fisher’s exact test.
Error bars represent SD (E and F). See also Figure S3.

Cell 142, 699–713, September 3, 2010 ª2010 Elsevier Inc. 705


Figure 4. Combination Therapy with Anti-CD47 Antibody and Rituximab Eliminates Lymphoma in Both Disseminated and Localized Human
NHL Xenotransplant Mouse Models
(A) NSG mice transplanted intravenously with luciferase-expressing Raji cells were treated with the indicated antibodies (n = 8 per treatment group). Luciferase
imaging of representative mice from pre- and 10 days post-treatment are shown (A) and averaged for all mice in each treatment group (B).
(C) Kaplan-Meier survival analysis was performed (Table S2). p values compare IgG control to combination antibody treatment or anti-CD47 antibody/rituximab
single antibody to combination. Arrows indicate start (day 14) and stop (day 35) of treatment.
(D) Luciferase-expressing Raji cells were transplanted subcutaneously in the flank of NSG mice. When palpable tumors (0.1 cm3) formed, treatment
began with the indicated antibodies. Luciferase imaging of representative mice from each treatment group is shown before (day 0) and during (day 14)
treatment.
(E) Quantified bioluminescence was determined and averaged for all mice in each treatment group (n = 7).
(F) Tumor volume was measured with average volume shown. p values were derived from a two-way ANOVA and compared to IgG treatment.
*p < 0.05, ***p < 0.001, ****p < 0.0001. Complete remission was defined as the number of mice with no evidence of tumor at the indicated date. Relapse was
defined as the number of mice achieving a complete remission that later developed recurrence of tumor growth. For (E), one mouse that achieved a complete
remission died of non-tumor-related causes. Data presented in (B), (E), and (F) are mean values ± SD. See also Figure S4 and Table S2.

706 Cell 142, 699–713, September 3, 2010 ª2010 Elsevier Inc.


Figure 5. Combination Therapy with Anti-CD47 Antibody and Rituximab Eliminates Lymphoma in Primary Human NHL Xenotransplant
Mouse Models
(A and B) Sublethally irradiated NSG mice were transplanted intravenously with cells from a primary DLBCL patient (NHL7) and treated with the indicated anti-
bodies. Flow cytometry of human lymphoma engraftment in the bone marrow of two representative mice is shown pre- and 14 days post-antibody treatment in
(A). Data from all mice are included in (B). **p < 0.01, comparing pre- and post-treatment values for each respective antibody treatment.
(C) Kaplan-Meier survival analysis (Table S3) of DLBCL-engrafted mice from each antibody treatment cohort is shown (n R 10 per antibody group), with p values
calculated comparing control IgG to combination antibody treatment or anti-CD47 antibody/rituximab single antibody to combination treatment. Arrows indicate
start (day14) and stop (day 28) of treatment.
(D and E) Mice engrafted intravenously with a primary FL patient sample (NHL31) were treated with a single dose of the indicated antibodies. Compared to IgG
control and rituximab, anti-CD47 antibody alone or in combination with rituximab eliminated tumor burden in the peripheral blood (p = 0.04, two-way ANOVA) and
bone marrow (p < 0.001, t test).
(E) Lyphoma engraftment in the bone marrow was determined 14 days post-treatment. Each antibody treatment group consisted of three mice.
For mice reported in (D) and (E), human lymphoma chimerism was between 5% and 25% in the peripheral blood and bone marrow. Error bars represent SD
(D and E). See also Figure S5 and Table S3.

Cell 142, 699–713, September 3, 2010 ª2010 Elsevier Inc. 707


Figure 6. Synergy between Anti-CD47 Antibody and Rituximab Does Not Occur through NK Cells or Complement
(A) NHL cells were incubated with the indicated soluble antibodies for 2 hr and the percentage of dead cells was calculated (% annexin V+ and/or 7-AAD+). No
statistically significant difference in % dead cells was observed with the combination of anti-CD47 antibody and rituximab compared to either anti-CD47 antibody
alone (p = 0.24) or rituximab alone (p = 0.95).
(B) SIRPa expression is shown for both human and mouse NK cells as determined by flow cytometry.
(C and D) Chromium release assays measuring ADCC were performed in triplicate with human (C) and mouse (D) at an effector:target ratio of 17.5:1, and percent-
specific lysis is reported. Antibodies were incubated at 10 mg/ml except anti-CD47 full-length or F(ab0 )2 antibody+rituximab (5 mg/ml).
(E) CDC assay with human complement was performed in duplicate. Compared to IgG1 isotype control, anti-CD47 antibody did not enable CDC (p > 0.2),
whereas rituximab did (p < 0.001) by two-way ANOVA for both SUDHL4 and NHL17*. Combination treatment with anti-CD47 antibody and rituximab did not
enable greater levels of CDC compared to rituximab (p = 0.78).
(F) CDC assay with mouse complement was performed in duplicate. Compared to IgG1 isotype control, anti-CD47 antibody did not enable CDC (p > 0.25)
whereas rituximab did (p = 0.03, p = 0.08, respectively) for both SUDHL4 and NHL17*. No difference in CDC between CD47 antibody+rituximab and rituximab
alone was observed (p > 0.13) for both SUDHL4 and NHL17*.
*p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001. Error bars represent SD (C–F). NHL17* = primary NHL17 cells expanded in culture. See also Figure S6.

708 Cell 142, 699–713, September 3, 2010 ª2010 Elsevier Inc.


effectors, anti-CD47 antibody did not induce increased ADCC of (Takai et al., 1994), but still expressing SIRPa (Figure S7A),
Raji or SUDHL4 cells compared to IgG1 isotype control (Fig- were utilized as effector cells for phagocytosis of NHL cells incu-
ure 6C). Although rituximab did enable ADCC of these two bated with either anti-CD47 antibody, anti-SIRPa antibody, ritux-
NHL cell lines, no synergistic effect between anti-CD47 antibody imab, or anti-CD47/anti-SIRPa antibody in combination with
and rituximab was observed (p = 0.77, Figure 6C). As anti-CD47 rituximab. As predicted, anti-CD47 antibody and anti-SIRPa
antibody (B6H12.2) is a mouse IgG1 isotype, we repeated these antibody, but not rituximab, enabled phagocytosis of NHL cells,
assays with mouse NK cells. Anti-CD47 antibody caused without evidence of synergistic phagocytosis (Figure 7C).
increased ADCC of these two NHL cell lines compared to isotype Third, F(ab0 )2 fragments of both anti-CD47 antibody and
control, whereas rituximab induced ADCC to a lesser degree rituximab were generated (Figures S7B–S7I) and utilized in
(Figure 6D). To test whether anti-CD47 antibody-mediated phagocytosis assays with NHL cells and wild-type macro-
ADCC was Fc dependent, we generated a F(ab0 )2 fragment of phages. Anti-CD47 F(ab0 )2 antibody synergized with rituximab
the anti-CD47 antibody (Figures S7B–S7I). The F(ab0 )2 fragment as demonstrated by isobologram analysis (Figure 7D). Addition-
did not enable ADCC, indicating that the increased ADCC was ally, anti-CD47 F(ab0 )2, but not rituximab F(ab0 )2, enabled phago-
likely mediated through FcRs (Figure 6D). The combination of cytosis of NHL cells (Figure 7E). Consistent with the proposed
anti-CD47 antibody or F(ab0 )2 fragment with rituximab did not mechanism, synergistic phagocytosis was observed with the
induce a statistically significant increase in ADCC compared to combination of either full-length anti-CD47 or anti-CD47 F(ab0 )2
single agent therapy, indicating no synergistic effect. Third, we with full-length rituximab, but not with any combination involving
investigated the role of complement in anti-CD47 antibody- rituximab F(ab0 )2 (Figure 7E).
mediated cytotoxicity. Using either human (Figure 6E) or mouse Fourth, synergistic phagocytosis was investigated in vivo
(Figure 6F) complement, anti-CD47 antibody did not induce CDC using GFP+ Raji cells engrafted in NSG mice. As single agents,
of either an NHL cell line or a primary NHL sample, whereas anti-CD47 antibody and rituximab enabled phagocytosis of
rituximab did induce significant CDC against both of these Raji cells engrafted in the liver as evidenced by an increased
samples. Moreover, the combination of anti-CD47 antibody percentage of mouse macrophages containing phagocytosed
and rituximab did not induce increased CDC compared to ritux- GFP+ Raji cells (Figure 7F). Most significantly, combination
imab alone. Fourth, we investigated the relative contribution of anti-CD47 antibody and rituximab treatment enabled signifi-
major components of macrophages, NK cells, and complement cantly increased phagocytosis compared to either single agent
in mediating the therapeutic effects of anti-CD47 antibody and demonstrating that synergistic phagocytosis occurred in vivo
rituximab in vivo. Luciferase-labeled Raji cells were engrafted (Figure 7F).
intravenously into SCID mice, which have functional macro-
phages, NK cells, and complement. Mice were then separated DISCUSSION
into cohorts receiving selective depletion of either macrophages
by clodronate, NK cells by anti-asialoGM1 antibody, comple- In this report, we identify a distinct mechanism of synergy
ment by cobra venom factor, or a vehicle control. These cohorts between mAbs in cancer therapy leading to cures in the absence
were then treated with combination anti-CD47 antibody and rit- of chemotherapy. Specifically, we utilized a blocking anti-CD47
uximab therapy for 3 days, and tumor burden was measured antibody in combination with the anti-CD20 antibody rituximab
by bioluminescent imaging pre- and post-treatment. Compared to eradicate human NHL through a mechanism of synergy
to vehicle control, NK cell and complement depletion had no involving FcR-independent enabling of phagocytosis by anti-
effect on tumor elimination by combination antibody therapy CD47 antibody and FcR-dependent stimulation of phagocytosis
(Figure S6E). In contrast, macrophage depletion significantly by rituximab. In addition, the identification of CD47 expression
abrogated the therapeutic effect, indicating that macrophages, as a prognostic factor could be incorporated into standard clin-
and not NK cells or complement, are required for combination ical prognostic considerations across multiple subtypes of NHL
anti-CD47 antibody and rituximab-mediated elimination of NHL and may be useful in risk-adapted therapy decision-making.
in vivo. Although it is thought that many therapeutic mAbs for human
malignancies, including rituximab, function primarily through
Anti-CD47 Antibody Synergizes with Rituximab through NK cell-mediated ADCC, several lines of evidence indicate that
FcR-Independent and FcR-Dependent Mechanisms the therapeutic effect of anti-CD47 antibody alone or in combi-
We hypothesize that the observed synergy between an anti- nation with rituximab is mediated primarily through macrophage
CD47 antibody and rituximab occurs through the combination phagocytosis. First, synergistic macrophage phagocytosis was
of two separate mechanisms for stimulating phagocytosis: (1) observed with combination anti-CD47 antibody and rituximab
FcR-independent through blockade of an inhibitory phagocytic in vitro, whereas no synergy was observed for direct apoptosis,
signal by anti-CD47 antibody, and (2) FcR-dependent through ADCC, or CDC (Figures 2C–2E, Figure S6, Figure 6A, and Figures
delivery of a positive phagocytic signal by rituximab. We utilized 6C–6F). Second, phagocytosis of NHL cells in vivo was observed
four independent methods to investigate this hypothesis. First, with either anti-CD47 antibody or rituximab alone and, most
synergistic phagocytosis was observed with the combination importantly, significantly increased with combination therapy
of anti-SIRPa antibody and rituximab by isobologram analysis (Figure 7F). Third, the therapeutic effect of combination antibody
(Figure 7A), and with a large panel of primary NHL samples (Fig- treatment was similar in an NHL xenotransplant model using
ure 7B). Second, mouse macrophages lacking the Fcg receptor, complement and NK cell-deficient NSG mice (Figure 4C) as in
thereby incapable of enabling FcR-dependent phagocytosis complement and NK cell-competent SCID mice (Figures S4A

Cell 142, 699–713, September 3, 2010 ª2010 Elsevier Inc. 709


Figure 7. Anti-CD47 Antibody Synergizes with Rituximab through FcR-Independent and FcR-Dependent Mechanisms
(A) Isobologram analysis of phagocytosis induced by anti-SIRPa antibody and rituximab is shown for Raji cells and mouse macrophages.
(B and C) NHL cells were incubated in vitro with the indicated antibodies in the presence of wild-type (B) or Fcgr/ (C) mouse macrophages, and the phagocytic
index was determined.
(D) Isobologram analysis of phagocytosis induced by anti-CD47 F(ab0 )2 antibody and rituximab is shown for Raji cells and mouse macrophages.
(E) NHL cells were incubated with wild-type mouse macrophages in the presence of the indicated full-length or F(ab0 )2 antibodies (single antibodies at 10 mg/ml,
combination antibodies at 5 mg/ml each) and the phagocytic index was determined.
(F) The level of in vivo phagocytosis measured as the percentage of mouse macrophages containing phagocytosed GFP+ Raji cells (hCD45GFP+F4/80+) was
determined by flow cytometry of livers from mice engrafted with GFP+ Raji cells and then treated with the indicated antibodies (see Experimental Procedures),
with each treatment group performed in duplicate. Compared to IgG control, anti-CD47 antibody and rituximab enabled increased levels of phagocytosis.
Compared to anti-CD47 antibody alone, combination anti-CD47 antibody and rituximab enabled higher levels of phagocytosis.
*p < 0.05, **p < 0.01, ****p < 0.0001. Error bars represent SD (E and F). See also Figure S7.

710 Cell 142, 699–713, September 3, 2010 ª2010 Elsevier Inc.


and S4B), suggesting that macrophages alone are sufficient to targeting of several human cancers including AML (Majeti
mediate the therapeutic effect. Fourth, depletion of macro- et al., 2009), bladder cancer (Chan et al., 2009), and now NHL,
phages, but not complement or NK cells, abrogated the syner- leading us to speculate that CD47 targeting will be effective
gistic effect of anti-CD47 antibody in combination with rituximab against a wide range of human cancers.
(Figure S6E). These studies highlight the importance of macro-
phages as effectors of anti-CD47 antibody therapy in human EXPERIMENTAL PROCEDURES
NHL.
This study describes a mechanism of antibody synergy in the Cell Lines
elimination of NHL in the absence of chemotherapy. Combina- A Burkitt’s lymphoma cell line (Raji) and a DLBCL cell line (SUDHL4) were
obtained from the American Type Culture Collection or generated in the lab.
tion antibody therapies for NHL have previously been investi-
The NHL17* cell line was generated from a patient with DLBCL by culturing
gated, mostly in combination with rituximab, with some pro- bulk cells in vitro with IMDM supplemented with 10% human AB serum for
gressing to clinical trials. These include a humanized antibody 1.5 months.
targeting the B cell antigen CD22 (epratuzumab) and galiximab,
a chimeric antibody targeting the costimulatory ligand CD80. Human Samples
Phase I/II studies with either epratuzumab or galiximab in combi- Normal human peripheral blood and human NHL samples were obtained from
nation with rituximab demonstrate relative safety and clinical the Stanford University Medical Center (Stanford, CA, USA) with informed
consent, according to an IRB-approved protocol (Stanford IRB# 13500) or
responses equal to or greater than single agent therapy alone
with informed consent from the Norwegian Radium Hospital (Oslo, Norway)
(Leonard et al., 2005, 2007, 2008). Based on these results, phase according to a Regional Ethic Committee (REK)-approved protocol (REK#
III trials are underway. Antibody combinations involving anti- 2.2007.2949). Normal tonsils for germinal center B cell analysis were obtained
CD20 antibodies and antibodies to proapoptotic receptors are from discarded tonsillectomy specimens from consented pediatric patients at
also being explored preclinically (Daniel et al., 2007; Maddipatla Stanford University Medical Center according to an IRB-approved protocol
et al., 2007). These studies highlight the clinical potential of (Stanford IRB# 13500).
combination antibody approaches in NHL patients.
Combination therapy with two or more mAbs possesses Flow Cytometry Analysis
For analysis of normal peripheral blood cells, germinal center B cells, and
several advantages compared to monotherapies in NHL or other
primary NHL cells, the following antibodies were used: CD19, CD20, CD3,
malignancies. First, therapy solely with monoclonal antibodies CD10, CD45, CD5, CD38 (Invitrogen, Carlsbad, CA, USA and BD Biosciences,
targeting cancer-specific antigens could result in decreased San Jose, CA, USA). Analysis of CD47 expression was performed with an anti-
off-target toxicity compared to current therapeutic regimens human CD47 FITC antibody (clone B6H12.2, BD Biosciences). Cell staining
that utilize chemotherapy. Second, synergy between two distinct and flow cytometry analysis was performed as previously described (Majeti
antibody effector mechanisms, FcR-independent and FcR- et al., 2009).
dependent as shown here, would result in increased therapeutic
efficacy. Third, antibody targeting of two distinct cell-surface Evaluation of Prognostic Value of CD47 in NHL
Gene expression and clinical data were analyzed for eight previously
antigens would be more likely to eliminate cancer cells with
described cohorts of adult NHL patients, including four studies of patients
pre-existing epitope variants or epitope loss, such as those with DLBCL, three with B-CLL, and one with MCL (detailed in Table S1). The
reported in rituximab-refractory/resistant NHL patients (Foran clinical end points analyzed included overall (OS), progression free (PFS),
et al., 2001; Hiraga et al., 2009; Kennedy et al., 2004). Fourth, and event-free survival (EFS), with events defined as the interval between
a bispecific FcR-engaging antibody with one arm binding and study enrollment and need for therapy or death from any cause, with data
blocking CD47 and the other binding to a validated cancer anti- censored for patients who did not have an event at the last follow-up visit.
See Extended Experimental Procedures for a detailed description of the
body target (CD20) could reduce potential antibody toxicity,
analyses.
while retaining the synergy effect, especially as CD47 is
expressed in multiple normal tissue types. Although we demon-
Therapeutic Antibodies
strated that an anti-mouse CD47 antibody is relatively nontoxic Rituximab (anti-CD20, human IgG1) was obtained from the Stanford University
to wild-type mice (Majeti et al., 2009), a clinical anti-human Medical Center, mouse anti-human CD20, IgG2a from Beckman Coulter
CD47 antibody may have a different human toxicity profile that (Miami, FL, USA), and anti-CD47 antibody BRIC126, IgG2b from AbD Serotec
could be overcome by a bispecific antibody. (Raleigh, NC, USA). Other anti-CD47 antibodies were used as in Majeti et al.
In addition to its application in NHL, the reported mechanism (2009). All in vivo antibody experiments were performed using the anti-CD47
B6H12.2 antibody.
of antibody synergy provides proof-of-principle that a blocking
mAb directed against CD47 can synergize with an FcR-acti-
In Vitro Isobologram Studies
vating antibody to provide superior therapeutic efficacy. This In vitro phagocytosis assays were conducted with NHL cells incubated with
finding raises the possibility of potential synergy between an anti-CD47 antibody (B6H12.2), anti-CD20 IgG2a, or rituximab either alone or
anti-CD47 antibody and other clinically approved therapeutic in combination at concentrations from 1 mg/ml to 10 mg/ml. The concentration
antibodies that may activate FcRs on immune effector cells for of each antibody required to produce a defined single-agent effect (phagocytic
the treatment of diverse human malignancies, including trastu- index) was determined for each cell type. Concentrations of the two antibodies
zumab (Herceptin) for HER2-positive breast carcinomas, cetux- combined to achieve this same phagocytic index were then plotted on an
isobologram and the combination index (CI) determined. The CI was calcu-
imab (Erbitux) for colorectal carcinomas and head and neck
lated from the formula CI = (d1/D1) + (d2/D2), whereby d1 = dose of drug
squamous cell carcinomas, alemtuzumab (Campath) for CLL 1 in combination to achieve the phagocytic index, d2 = dose of drug 2 in
and T cell lymphoma, and others in development (Finn, 2008). combination to achieve the phagocytic index, D1 = dose of drug 1 alone to
To date, we have demonstrated effective anti-CD47 antibody achieve the phagocytic index, D2 = dose of drug 2 alone to achieve the

Cell 142, 699–713, September 3, 2010 ª2010 Elsevier Inc. 711


phagocytic index. A CI of less than, equal to, and greater than 1 indicates SUPPLEMENTAL INFORMATION
synergy, additivity, and antagonism, respectively.
Supplemental Information includes Extended Experimental Procedures, seven
Annexin V Apoptosis Assays figures, and three tables and can be found with this article online at doi:10.
Assays were performed as previously described (Majeti et al., 2009). 1016/j.cell.2010.07.044.

ACKNOWLEDGMENTS
Preparation of Human and Mouse Immune Effector Cells, Immune
Effector Cytotoxicity Assays, and In Vivo Depletion of Immune
The authors acknowledge Dr. Christopher Contag for providing luciferase
Effector Cells
constructs, Dr. Robert Negrin for assistance, Dr. Yaso Natkunam for immuno-
See Extended Experimental Procedures.
histochemistry, Libuse Jerabek and Theresa Storm for lab management, and
Adriane Mosely for animal husbandry. We also acknowledge the patients and
Preparation of F(ab0 )2 Fragments surgeons including Drs. Wapnir, Chang, Cheng, Janisiewicz, Koltai, Liebowitz,
See Extended Experimental Procedures. and Messner for providing research specimens. M.P.C. is supported by the
HHMI and the Stanford Cancer Biology Program, A.A.A. by an NIH T32 Ruth
Generation of Luciferase-Positive Cell Lines and Luciferase Imaging L. Kirschstein National Research Service Award (HL007970), R. Malumbres
Analysis by a fellowship from Fundación Caja Madrid, I.S.L. by NIH grant CA122105,
See Extended Experimental Procedures. and R. Majeti by a grant from the AACR. R. Majeti holds a Career Award for
Medical Scientists from the Burroughs Wellcome Fund. I.L.W., M.P.C.,
A.A.A., and R. Majeti have filed U.S. Patent Application Serial No. 12/321,215
In Vivo Precoating Engraftment Assay entitled ‘‘Methods For Manipulating Phagocytosis Mediated by CD47.’’ This
Assay were performed as previously described (Majeti et al., 2009). Precoated research is supported by NIH grant P01CA139490 to I.L.W. and funding from
cells were transplanted intravenously into SCID mice or sublethally irradiated the Smith Family Fund.
(200 rads) NOD.Cg-PrkdcscidIl2rgtm1Wjl/SzJ (NSG). All experiments involving M.P.C., A.A.A., I.L.W., and R. Majeti designed the experiments, and M.P.C.,
mice were performed according to Stanford University institutional animal A.A.A., I.L.W., and R. Majeti wrote the manuscript. M.P.C., A.A.A., C.T.,
guidelines. J.H.M., S.G., M.J., A.C.C., C.K.C., B.T.T., C.Y.P, F.Z., R. Malumbres, and
I.S.L. performed the experiments and analyzed data. J.B., R.D.G., R.L., and
In Vivo Antibody Treatment in a Disseminated Lymphoma I.S.L provided patient samples and clinical data. H.E.K. provided reagents.
Xenograft Model B.V. generated F(ab0 )2 fragments of anti-CD47 antibody and rituximab. All
1.5 3 106 luciferase-labeled Raji cells were injected intravenously into the authors endorse the full content of this work.
retro-orbital sinus of 6- to 10-week-old SCID or NSG mice. Those mice with
luciferase-positive lymphoma were given daily intraperitoneal injections of Received: January 7, 2010
200 mg mouse IgG control, anti-CD47 antibody, rituximab, or 200 mg anti- Revised: April 23, 2010
CD47 antibody + 200 mg rituximab for 3 weeks. Antibody treatment was Accepted: July 6, 2010
then stopped and mice were followed for survival analysis. A complete remis- Published: September 2, 2010
sion (CR) was defined as no evidence of lymphoma by bioluminescence at the
end of treatment. A relapse was defined as evidence of lymphoma by biolumi- REFERENCES
nescence after the end of treatment in a mouse with a prior CR.
Adams, G.P., and Weiner, L.M. (2005). Monoclonal antibody therapy of cancer.
In Vivo Antibody Treatment in a Localized Lymphoma Nat. Biotechnol. 23, 1147–1157.
Xenograft Model Alizadeh, A.A., Eisen, M.B., Davis, R.E., Ma, C., Lossos, I.S., Rosenwald, A.,
3 3 106 luciferase-labeled Raji cells were injected subcutaneously into the Boldrick, J.C., Sabet, H., Tran, T., Yu, X., et al. (2000). Distinct types of diffuse
right flank of 6- to 10-week-old NSG mice. Those mice with luciferase-positive large B-cell lymphoma identified by gene expression profiling. Nature 403,
lymphoma were given daily intraperitoneal injections of 400 mg mouse IgG 503–511.
control, 400 mg anti-CD47, 200 mg rituximab, or 400 mg anti-CD47 + 200 mg rit- Brown, E.J., and Frazier, W.A. (2001). Integrin-associated protein (CD47) and
uximab for 4 weeks. Tumor volume was measured every 3–4 days using the its ligands. Trends Cell Biol. 11, 130–135.
formula (length*width)/2. Antibody treatment was then stopped and mice Cartron, G., Watier, H., Golay, J., and Solal-Celigny, P. (2004). From the bench
were followed for survival analysis. to the bedside: ways to improve rituximab efficacy. Blood 104, 2635–2642.
Chan, K.S., Espinosa, I., Chao, M., Wong, D., Ailles, L., Diehn, M., Gill, H.,
In Vivo Antibody Treatment of Primary NHL-Engrafted Mice Presti, J., Jr., Chang, H.Y., van de Rijn, M., et al. (2009). Identification, molec-
2 3 106 NHL cells were transplanted intravenously via retro-orbital plexus into ular characterization, clinical prognosis, and therapeutic targeting of human
sublethally irradiated NSG mice. Two to ten weeks later, bone marrow was bladder tumor-initiating cells. Proc. Natl. Acad. Sci. USA 106, 14016–14021.
aspirated from these mice and those mice with evidence of human lymphoma
Cheson, B.D., and Leonard, J.P. (2008). Monoclonal antibody therapy for
engraftment (hCD45+CD19/CD10+ bone marrow cells) were then treated with
B-cell non-Hodgkin’s lymphoma. N. Engl. J. Med. 359, 613–626.
the same antibody regimen as in the disseminated lymphoma model. After
14 days, bone marrow cells from these mice were aspirated and antibody Chou, T.C. (2008). Preclinical versus clinical drug combination studies. Leuk.
treatment was stopped and mice followed for survival analysis. A CR was Lymphoma 49, 2059–2080.
defined as no evidence of lymphoma in the BM at end of treatment. A relapse Daniel, D., Yang, B., Lawrence, D.A., Totpal, K., Balter, I., Lee, W.P., Gogineni,
was defined as evidence of lymphoma in the BM after end of treatment in A., Cole, M.J., Yee, S.F., Ross, S., and Ashkenazi, A. (2007). Cooperation of the
a mouse with a prior CR. proapoptotic receptor agonist rhApo2L/TRAIL with the CD20 antibody rituxi-
mab against non-Hodgkin lymphoma xenografts. Blood 110, 4037–4046.
In Vivo Phagocytosis Finn, O.J. (2008). Cancer immunology. N. Engl. J. Med. 358, 2704–2715.
In vivo phagocytosis was performed as previously described (Majeti et al., Foran, J.M., Norton, A.J., Micallef, I.N., Taussig, D.C., Amess, J.A., Rohatiner,
2009) analyzing mice transplanted with GFP+ Raji cells into adult NSG mice. A.Z., and Lister, T.A. (2001). Loss of CD20 expression following treatment with
Mice were given a single dose of antibody and analyzed 4 hr later for in vivo rituximab (chimaeric monoclonal anti-CD20): a retrospective cohort analysis.
phagocytosis. Br. J. Haematol. 114, 881–883.

712 Cell 142, 699–713, September 3, 2010 ª2010 Elsevier Inc.


Glennie, M.J., French, R.R., Cragg, M.S., and Taylor, R.P. (2007). Mechanisms tion of monoclonal antibodies targeting TRAIL-R1 and CD20. Clin. Cancer Res.
of killing by anti-CD20 monoclonal antibodies. Mol. Immunol. 44, 3823–3837. 13, 4556–4564.
Hiraga, J., Tomita, A., Sugimoto, T., Shimada, K., Ito, M., Nakamura, S., Kiyoi, Majeti, R., Chao, M.P., Alizadeh, A.A., Pang, W.W., Jaiswal, S., Gibbs, K.D.,
H., Kinoshita, T., and Naoe, T. (2009). Down-regulation of CD20 expression in Jr., van Rooijen, N., and Weissman, I.L. (2009). CD47 is an adverse prognostic
B-cell lymphoma cells after treatment with rituximab-containing combination factor and therapeutic antibody target on human acute myeloid leukemia stem
chemotherapies: its prevalence and clinical significance. Blood 113, 4885– cells. Cell 138, 286–299.
4893. Mateo, V., Lagneaux, L., Bron, D., Biron, G., Armant, M., Delespesse, G., and
Jaiswal, S., Jamieson, C.H., Pang, W.W., Park, C.Y., Chao, M.P., Majeti, R., Sarfati, M. (1999). CD47 ligation induces caspase-independent cell death in
Traver, D., van Rooijen, N., and Weissman, I.L. (2009). CD47 is upregulated chronic lymphocytic leukemia. Nat. Med. 5, 1277–1284.
on circulating hematopoietic stem cells and leukemia cells to avoid phagocy- Milush, J.M., Long, B.R., Snyder-Cappione, J.E., Cappione, A.J., 3rd, York,
tosis. Cell 138, 271–285. V.A., Ndhlovu, L.C., Lanier, L.L., Michaelsson, J., and Nixon, D.F. (2009). Func-
Jaiswal, S., Chao, M.P., Majeti, R., and Weissman, I.L. (2010). Macrophages tionally distinct subsets of human NK cells and monocyte/DC-like cells identi-
as mediators of tumor immunosurveillance. Trends Immunol. 31, 212–219. fied by coexpression of CD56, CD7, and CD4. Blood 114, 4823–4831.
Katzenberger, T., Petzoldt, C., Holler, S., Mader, U., Kalla, J., Adam, P., Ott, Nimmerjahn, F., and Ravetch, J.V. (2007). Antibodies, Fc receptors and
M.M., Muller-Hermelink, H.K., Rosenwald, A., and Ott, G. (2006). The Ki67 cancer. Curr. Opin. Immunol. 19, 239–245.
proliferation index is a quantitative indicator of clinical risk in mantle cell Oscier, D.G., Gardiner, A.C., Mould, S.J., Glide, S., Davis, Z.A., Ibbotson, R.E.,
lymphoma. Blood 107, 3407. Corcoran, M.M., Chapman, R.M., Thomas, P.W., Copplestone, J.A., et al.
Kennedy, A.D., Beum, P.V., Solga, M.D., DiLillo, D.J., Lindorfer, M.A., Hess, (2002). Multivariate analysis of prognostic factors in CLL: clinical stage,
C.E., Densmore, J.J., Williams, M.E., and Taylor, R.P. (2004). Rituximab infu- IGVH gene mutational status, and loss or mutation of the p53 gene are inde-
sion promotes rapid complement depletion and acute CD20 loss in chronic pendent prognostic factors. Blood 100, 1177–1184.
lymphocytic leukemia. J. Immunol. 172, 3280–3288. Rosenwald, A., Wright, G., Chan, W.C., Connors, J.M., Campo, E., Fisher, R.I.,
Kikuchi, Y., Uno, S., Yoshimura, Y., Otabe, K., Iida, S., Oheda, M., Fukushima, Gascoyne, R.D., Muller-Hermelink, H.K., Smeland, E.B., Giltnane, J.M., et al.
N., and Tsuchiya, M. (2004). A bivalent single-chain Fv fragment against CD47 (2002). The use of molecular profiling to predict survival after chemotherapy
induces apoptosis for leukemic cells. Biochem. Biophys. Res. Commun. 315, for diffuse large-B-cell lymphoma. N. Engl. J. Med. 346, 1937–1947.
912–918. Rosenwald, A., Wright, G., Wiestner, A., Chan, W.C., Connors, J.M., Campo,
Kikuchi, Y., Uno, S., Kinoshita, Y., Yoshimura, Y., Iida, S., Wakahara, Y., E., Gascoyne, R.D., Grogan, T.M., Muller-Hermelink, H.K., Smeland, E.B.,
Tsuchiya, M., Yamada-Okabe, H., and Fukushima, N. (2005). Apoptosis et al. (2003). The proliferation gene expression signature is a quantitative inte-
inducing bivalent single-chain antibody fragments against CD47 showed anti- grator of oncogenic events that predicts survival in mantle cell lymphoma.
tumor potency for multiple myeloma. Leuk. Res. 29, 445–450. Cancer Cell 3, 185–197.

Kim, M.J., Lee, J.C., Lee, J.J., Kim, S., Lee, S.G., Park, S.W., Sung, M.W., and Shultz, L.D., Lyons, B.L., Burzenski, L.M., Gott, B., Chen, X., Chaleff, S., Kotb,
Heo, D.S. (2008). Association of CD47 with natural killer cell-mediated cytotox- M., Gillies, S.D., King, M., Mangada, J., et al. (2005). Human lymphoid and
icity of head-and-neck squamous cell carcinoma lines. Tumour Biol. 29, myeloid cell development in NOD/LtSz-scid IL2R gamma null mice engrafted
28–34. with mobilized human hemopoietic stem cells. J. Immunol. 174, 6477–6489.
Smith, M.R. (2003). Rituximab (monoclonal anti-CD20 antibody): mechanisms
Leonard, J.P., Coleman, M., Ketas, J., Ashe, M., Fiore, J.M., Furman, R.R.,
of action and resistance. Oncogene 22, 7359–7368.
Niesvizky, R., Shore, T., Chadburn, A., Horne, H., et al. (2005). Combination
antibody therapy with epratuzumab and rituximab in relapsed or refractory Takai, T., Li, M., Sylvestre, D., Clynes, R., and Ravetch, J.V. (1994). FcR
non-Hodgkin’s lymphoma. J. Clin. Oncol. 23, 5044–5051. gamma chain deletion results in pleiotrophic effector cell defects. Cell 76,
519–529.
Leonard, J.P., Friedberg, J.W., Younes, A., Fisher, D., Gordon, L.I., Moore, J.,
Czuczman, M., Miller, T., Stiff, P., Cheson, B.D., et al. (2007). A phase I/II study Tallarida, R.J. (2006). An overview of drug combination analysis with isobolo-
of galiximab (an anti-CD80 monoclonal antibody) in combination with rituxi- grams. J. Pharmacol. Exp. Ther. 319, 1–7.
mab for relapsed or refractory, follicular lymphoma. Ann. Oncol. 18, 1216– Taylor, R.P., and Lindorfer, M.A. (2008). Immunotherapeutic mechanisms of
1223. anti-CD20 monoclonal antibodies. Curr. Opin. Immunol. 20, 444–449.
Leonard, J.P., Schuster, S.J., Emmanouilides, C., Couture, F., Teoh, N., The International Non-Hodgkin’s Lymphoma Prognostic Factors Project.
Wegener, W.A., Coleman, M., and Goldenberg, D.M. (2008). Durable complete (1993). A predictive model for aggressive non-Hodgkin’s lymphoma. N. Engl.
responses from therapy with combined epratuzumab and rituximab: final J. Med. 329, 987–994.
results from an international multicenter, phase 2 study in recurrent, indolent, Uno, S., Kinoshita, Y., Azuma, Y., Tsunenari, T., Yoshimura, Y., Iida, S., Kiku-
non-Hodgkin lymphoma. Cancer 113, 2714–2723. chi, Y., Yamada-Okabe, H., and Fukushima, N. (2007). Antitumor activity of
Maddipatla, S., Hernandez-Ilizaliturri, F.J., Knight, J., and Czuczman, M.S. a monoclonal antibody against CD47 in xenograft models of human leukemia.
(2007). Augmented antitumor activity against B-cell lymphoma by a combina- Oncol. Rep. 17, 1189–1194.

Cell 142, 699–713, September 3, 2010 ª2010 Elsevier Inc. 713


A C-Type Lectin Collaborates with a
CD45 Phosphatase Homolog to Facilitate
West Nile Virus Infection of Mosquitoes
Gong Cheng,1 Jonathan Cox,1 Penghua Wang,1 Manoj N. Krishnan,1 Jianfeng Dai,1 Feng Qian,2 John F. Anderson,3
and Erol Fikrig1,4,*
1Sectionof Infectious Diseases
2Sectionof Rheumatology
Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520, USA
3Department of Entomology, Connecticut Agricultural Experiment Station, New Haven, CT 06504, USA
4Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA

*Correspondence: erol.fikrig@yale.edu
DOI 10.1016/j.cell.2010.07.038

SUMMARY wide (Nir et al., 1968; Turell et al., 2000). In the United States,
the most important vectors are Culex pipiens in the east, Culex
West Nile virus (WNV) is the most common tarsalis in the midwest and west, and Culex quinquefasciatus in
arthropod-borne flavivirus in the United States; the southeast (Hayes et al., 2005). WNV has also been isolated
however, the vector ligand(s) that participate in infec- from Aedes, Ochlerotatus, and Culisetam mosquitoes (http://
tion are not known. We now show that an Aedes www.cdc.gov/ncidod/dvbid/westnile/mosquitoSpecies.htm).
aegypti C-type lectin, mosGCTL-1, is induced by Aedes aegypti, a member of the Culicinae subfamily, is a major
vector for numerous flaviviruses (Gould and Solomon, 2008).
WNV, interacts with WNV in a calcium-dependent
A. aegypti is ideal for viral pathogenesis studies because these
manner, and facilitates infection in vivo and in vitro. mosquitoes are easy to cultivate and the genome has been char-
A mosquito homolog of human CD45 in A. aegypti, acterized (Gubler, 1998; Nene et al., 2007; Halstead, 2008).
designated mosPTP-1, recruits mosGCTL-1 to A. aegypti is readily susceptibility to infection with WNV in
enable viral attachment to cells and to enhance viral the laboratory, and the virus rapidly disseminates throughout
entry. In vivo experiments show that mosGCTL-1 and most of the mosquito after the blood meal. As with Culex spp.,
mosPTP-1 function as part of the same pathway A. aegypti is a threat for the transmission of WNV to humans
and are critical for WNV infection of mosquitoes. (Vanlandingham et al., 2007).
A similar phenomenon was also observed in Culex C-type lectins are a group of carbohydrate-binding proteins
quinquefasciatus, a natural vector of WNV, further (Zelensky and Gready, 2005). Several members of this family
demonstrating that these genes participate in WNV are highly expressed by immune cells, including monocytes,
macrophages, and dendritic cells (DCs), and play a central
infection. During the mosquito blood-feeding pro-
role in activating host defenses (Robinson et al., 2006). Human
cess, WNV infection was blocked in vivo with mannose-binding lectin (MBL) is a pattern recognition molecule
mosGCTL-1 antibodies. A molecular understanding of the innate immune system that binds to sugars on the surface
of flaviviral-arthropod interactions may lead to strat- of invading pathogens, leading to opsonization, phagocytosis,
egies to control viral dissemination in nature. and activation of the complement pathway (Neth et al., 2002).
In contrast, some C-type lectins are recruited to facilitate flavivi-
INTRODUCTION ral infection. In mammals, two membrane C-type lectins,
DC-SIGN (CD209) and L-SIGN (CD209L), interact with flavivi-
West Nile virus (WNV) is maintained in a bird-mosquito transmis- ruses via high mannose glycans on viral glycoproteins (Klimstra
sion cycle and has become the most common arthropod-borne et al., 2003) and are essential host cell factors exploited by
flavivirus in the United States. Humans, horses, and other nona- dengue virus (DENV) and WNV to invade immature DCs and
vian vertebrates are incidental hosts (Monath and Heinz, 1996). macrophages (Geijtenbeek et al., 2000; Soilleux et al., 2002;
Infection in man can result in fever, meningitis, or encephalitis, Tassaneetrithep et al., 2003; Davis et al., 2006). Another
among other symptoms (Hubálek and Halouzka, 1999; Leis C-type lectin, the mannose receptor (MR), also interacts with
et al., 2002). Approved human vaccines or therapeutics are not the DENV envelope protein and may enhance viral attachment
available and preventive measures largely focus upon mosquito to phagocytes (Miller et al., 2008). A recent study identified
control (Reisen and Brault, 2007). C-type lectin domain family 5, member A (CLEC5A), as
The ability of different mosquito species to transmit WNV a DENV receptor. The association between CLEC5A and
varies widely. Culex spp. are the major vectors for WNV world- DENV does not result in viral entry, but rather stimulates the

714 Cell 142, 714–725, September 3, 2010 ª2010 Elsevier Inc.


Figure 1. Effect of WNV Infection on mosGCTL-1 Expression
(A) Viral distribution in A. aegypti. WNV (1 3 103 M.I.D50) was inoculated into the mosquito thorax.
(B–E) mosGCTL-1 messenger RNA (mRNA) was induced by viral infection in whole A. aegypti (B), salivary glands (C), hemolymph (D), and midgut (E). Total RNA
was isolated from various tissues or whole mosquitoes at 5 time points after viral infection.
(A–E) The viral load and mosGCTL-1 mRNA levels were determined by Taqman RT-QPCR and normalized with A. aegypti actin (AAEL011197). WNV (1 3 103
M.I.D50) was microinjected into each mosquito. Data are shown as the mean ± standard error (SEM). The experiments were repeated three times.
(F) Immunoblot to detect mosGCTL-1 in WNV-infected mosquitoes. Three WNV infected or control mosquitoes were pooled and homogenized. The supernatant
was then isolated, separated by SDS-PAGE, and probed with rabbit mosGCTL-1 antisera. UI, uninfected mosquitoes; I, WNV-infected mosquitoes; dpi, days
postinfection. Fifty micrograms of protein from mosquito lysates was loaded into each lane.
See also Figure S1 and Table S1.

release of proinflammatory cytokines, potentially contributing to well-known PTP, protein tyrosine phosphatase receptor type C
the pathogenesis of dengue hemorrhagic fever (Chen et al., (PTPRC, CD45), is important for thymocyte development and
2008). T cell activation (Byth et al., 1996; Trowbridge and Thomas,
Protein tyrosine phosphatases (PTPs) remove phosphate 1994) and is expressed on all nucleated cells of hemopoietic
groups from phosphorylated tyrosine residues and play critical origin (Thomas, 1989). The association between MBL and
roles in cell communication, shape, motility, proliferation, and CD45 in immature T cells influences thymocyte development
differentiation (Alonso et al., 2004; Mustelin et al., 2005). One (Baldwin and Ostergaard, 2001).

Cell 142, 714–725, September 3, 2010 ª2010 Elsevier Inc. 715


Figure 2. The Function of mosGCTL-1 in WNV Infection
(A and B) mosGCTL-1 silencing. The mock group was treated with the same amount of GFP dsRNA. mosGCTL-1- or GFP-dsRNA-treated mosquitoes were used
to isolate total RNA at several time points post dsRNA injection. mRNA levels were determined by SYBR Green RT-QPCR (A). mosGCTL-1-dsRNA-treated or
mock-treated mosquitoes were collected at 6 days after gene silencing. The supernatant was separated by SDS-PAGE and probed with rabbit mosGCTL-1
antisera (B).
(C and D) Silencing mosGCTL-1 impairs WNV (C), but not dengue virus (D), infection. The viral burden was examined at day 6 after infection. WNV or dengue virus
(10 M.I.D50) was used to challenge mosquitoes. The viral load was determined by Taqman RT-QPCR and normalized with A. aegypti actin. The result shown was
representative of four independent experiments.
(E) The role of the mosGCTL paralogues in WNV infection. The sample number was no less than 12 mosquitoes in each group. The viral burden was determined by
Taqman RT-QPCR and normalized with A. aegypti actin. *p < 0.05. The results were pooled from two independent experiments.
(F) WNV induces mosGCTL-1 homolog expression in C. quinquefasciatus. WNV-infected and mock mosquitoes were collected at 6 days after infection. Culex
mosGCTL-1 mRNA was determined by SYBR Green RT-QPCR and normalized with C. quinquefasciatus actin (CPIJ012570). The experiment was repeated three
times with similar results.
(G) Efficiency of Culex mosGCTL-1 RNA interference. The mock group was treated with the same amount of GFP dsRNA. mRNA levels were determined by SYBR
Green RT-QPCR.
(H) Silencing of Culex mosGCTL-1 decreases the WNV burden in C. quinquefasciatus. The viral burden was determined by Taqman RT-QPCR. The results were
combined from three independent experiments.

716 Cell 142, 714–725, September 3, 2010 ª2010 Elsevier Inc.


Genome-wide RNA interference (RNAi) screening studies mosquitoes. Total RNA from mock-injected and mosGCTL-1
have revealed several hundred host factors that influence WNV dsRNA-injected mosquitoes was analyzed by RT-QPCR. Com-
or DENV infection in human or Drosophila cell lines and have pared to the mock group, the target gene was silenced 6- to
identified cellular pathways that have a role in viral internaliza- 10-fold from day 3 through 9 (Figures 2A and 2B). WNV was
tion, replication, assembly, or secretion (Krishnan et al., 2008; therefore inoculated into mosquitoes on day 3 after dsRNA treat-
Sessions et al., 2009). However, the relationship between flavivi- ment, and the viral load was quantified on day 9. There was
ruses and mosquitoes is not well understood. We now examine a 3-fold reduction in the WNV burden in mosGCTL-1 silenced
WNV-mosquito interactions by using A. aegypti and Culex quin- A. aegypti (Figure 2C, p < 0.0001) compared to controls.
quefasciatus, characterize the expression profile of mosquito In contrast, the burden of a related flavivirus, dengue virus,
homologs of human susceptibility proteins (Krishnan et al., was unaffected by mosGCTL-1 silencing (Figure 2D), suggesting
2008) in response to viral infection, and identify a lectin-based that mosGCTL-1 has a specific role in WNV infection of
pathway that is critical for viral infection of mosquitoes. mosquitoes.
As mosGCTL-1 is part of a multigene family with 25 members
RESULTS and shares 22%–48% identity with many of these genes, we
examined the role of the mosGCTL paralogues in WNV and
A recent RNAi screening study characterized 283 human DENV infection. Twenty-one of these 25 genes were expressed
proteins that facilitate WNV infection (Krishnan et al., 2008). in adult female A. aegypti (Tables S1 and S2). Since AAEL014382
We have now identified 215 homologs of these genes in and AAEL014390 share greater than 95% identity with
A. aegypti. The expression of 32 genes was altered by the pres- AAEL011607 and AAEL011619, respectively, AAEL011607 and
ence of WNV in mosquitoes, potentially suggesting a role in viral AAEL011619 were selected for functional assays. We silenced
infection of the arthropod (Table S1 available online). To examine each of the expressed mosGCTL genes and then infected the
their function, we therefore silenced each of these 32 genes in mosquitoes with virus. In the WNV challenge, two additional
mosquitoes with RNAi and assessed the effect on viral load. mosGCTL subtypes (AAEL000556 and AAEL011610) showed
Knockdown of 13 genes significantly altered the viral burden in the similar phenotype to mosGCTL-1 (Figure 2E), and in DENV
the vector (Table S1 and Figure S1). One of these genes, infection, silencing of 5 mosGCTL-related genes reduced the
AAEL000563, exhibited the most dramatic reduction in viral DENV burden (Figure S2).
load after silencing and was selected as the target for further Culex mosquitoes are a common vector for WNV in nature.
investigation. AAEL000563 belongs to the A. aegypti galac- We therefore extended the studies to Culex quinquefasciatus,
tose-specific binding C-type lectin family, shares 26% amino a example of this species that is abundant in the southeast
acid identity with human mannose-binding lectin, and was U.S. We identified a mosGCTL-1 homolog (CPIJ010995, Culex
designated as mosGCTL-1 (mosquito galactose-specific binding mosGCTL-1) in C. quinquefasciatus that has a higher degree
C-type lectin). of similarity (64%) with mosGCTL-1 than other homologs.
We then determined whether Culex mosGCTL-1 had a similar
role in WNV infection. Culex mosGCTL-1 was upregulated by
mosGCTL-1 Expression Increases during WNV Infection WNV infection (Figure 2F). Silencing of Culex mosGCTL-1 in
of A. aegypti C. quinquefasciatus reduced the WNV burden compared to the
To further investigate the relationship between WNV infection control group (Figures 2G and 2H, p < 0.0001), suggesting that
and mosGCTL-1 expression, we determined the viral load and Culex mosGCTL-1 plays a role in facilitating WNV infection in
mosGCTL-1 level in selected A. aegypti tissues after the inocula- Culex mosquitoes.
tion of WNV into female mosquitoes. WNV was readily detect-
able 4 days after infection, and the viral level subsequently
increased. The salivary glands and hemolymph had the highest mosGCTL-1 Interacts with WNV
levels of WNV, while the viral load in the midgut was substantially Our results show that silencing mosGCTL-1 in A. aegypti and
lower (Figure 1A). mosGCTL-1 expression was induced in C. quinquefasciatus reduces WNV infection. The mechanism
diverse tissues over time, including the salivary glands, hemo- by which mosGCTL-1 facilitates viral infection in mosquitoes
lymph, and midgut (Figures 1B–1E). Immunoblots also demon- was, therefore, investigated. We first generated A. aegypti
strated an increased amount of mosGCTL-1 in WNV-infected mosGCTL-1 protein in a Drosophila expression system (Fig-
mosquitoes (Figure 1F). ure 3A) and investigated whether mosGCTL-1 associates with
WNV envelope (E) protein. Immunoprecipitation experiments
mosGCTL-1 Facilitates WNV Infection in A. aegypti revealed that these two proteins strongly interacted in a
Double-stranded RNA (dsRNA)-mediated gene silencing studies calcium-dependent manner (Figure 3B). We then determined,
then assessed the role of mosGCTL-1 in WNV infection of by ELISA, that mosGCTL-1 bound to WNV virions. mosGCTL-1

(A, C, D and H) Statistical analysis was done with the Mann-Whitney test in all experiments. Each dot represents the mRNA levels in an individual mosquito.
The horizontal line depicts the medians.
(E–G) The Mann-Whitney test was used for statistical analysis. Data are shown as the mean ± standard error (SEM).
See also Figure S2 and Table S2.

Cell 142, 714–725, September 3, 2010 ª2010 Elsevier Inc. 717


Figure 3. mosGCTL-1 Interacts with WNV
(A) Recombinant mosGCTL-1, produced in Drosophila cells, and purified with a Ni-His column (left). Glycosylated mosGCTL-1 was detected with a V5-HRP mAb
(right). The control was the supernatant of mock-infected S2 cells.
(B) mosGCTL-1 interacted with WNV E protein in a coimmunoprecipitation assay. The protein complex was pulled down with a flavivirus E mAb and probed with
anti-V5-HRP mAb. The experiments were repeated three times.
(C) mosGCTL-1 captured West Nile virions by ELISA. The interaction was determined with a flavivirus E mAb. Data are expressed as the mean ± standard error
from three independent experiments.
(D) Comicroinjection of the purified mosGCTL-1 with WNV enhanced virus infection in A. aegypti. The various amount of purified mosGCTL-1 were premixed with
WNV (10 M.I.D50 per mosquito) for 1 hr at 4 C. The mixture was microinjected into mosquito and compared to the control group inoculated with the same amount
WNV. The mosquitoes were collected at 3 days (i), 6 days (ii), and 9 days (iii) after WNV inoculation. Total RNA was isolated to determine the viral burden by
Taqman RT-QPCR and normalized with A. aegypti actin. Each dot represents the mRNA level in one mosquito. The horizontal line represents the medians.
n.s., nonsignificance (p > 0.05). The Mann-Whitney test was used for analysis. Three independent experiments yielded similar data.
(E) The association between mosGCTL-1 and WNV in A .aegypti hemolymph. Hemolymph was collected from WNV-infected or mock-infected mosquitoes
for immunofluorescence staining. WNV E protein was stained with anti-mouse IgG Alexa-488 (green), and mosGCTL-1 was identified with anti-rabbit
IgG Alexa-546 (red). Nuclei were stained blue with To-Pro-3 iodide. The white arrow represents the induced expression of mosGCTL-1 in WNV-infected

718 Cell 142, 714–725, September 3, 2010 ª2010 Elsevier Inc.


efficiently captured virus in the presence of calcium, and high levels of mosPTP-1 in the cytoplasm also had substantial
this interaction was inhibited by EDTA (Figure 3C). Since this mosGCTL-1 on the cell surface (Figure 4F). These results
association suggests a role during infection, we premixed suggest that mosGCTL-1 binds mosPTP-1.
mosGCTL-1 and WNV and then coinjected the combination To test whether mosPTP-1 has a conserved role in Culex spp.,
into A. aegypti and determined the viral burden. mosGCTL-1 we identified the mosPTP-1 homolog from the Culex quinquefas-
protein significantly enhanced the viral load at days 3 (Fig- ciatus genome (CPIJ014098, Culex mosPTP-1). Silencing of this
ure 3D, i, p = 0.0001) and 6 (Figure 3D, ii, p < 0.0001) after viral gene in Culex also influenced the viral burden (Figures S4B and
challenge. S4C), similar to mosPTP-1 in A. aegypti (Figure 4B), suggesting
To further characterize the association between mosGCTL-1 that mosPTP-1 in both these mosquito species have a similar
and WNV infection in vivo, we used immunofluorescence to role in WNV infection.
examine the hemolymph and salivary glands of WNV-infected
A. aegypti at different time points. Colocalization of mosGCTL-1 The mosGCTL-1/mosPTP-1 Pathway Has a Dominant
and WNV was clearly observed in both tissues at various inter- Role in WNV Infection
vals after WNV infection (Figure 3E and Figure S3). In the hemo- mosGCTL-1 and mosPTP-1 each facilitate WNV infection of
lymph, some hemocytes were highly infected by WNV, and A. aegypti in vivo and in vitro. We therefore assessed whether
mosGCTL-1 was induced in these infected hemocytes mosGCTL-1 and mosPTP-1 cooperate to enable viral infection.
(Figure 3E). We counted the number of mosGCTL-1-positive WNV E protein, mosGCTL-1, and mosPTP-1-Ex form a complex
and WNV-infected hemocytes by microscopy. All these infected in which mosGCTL-1 is the key factor linking the other two
cells stained positive for mosGCTL-1. The virus spread rapidly to proteins (Figure 5A). WNV E protein did not interact directly
the salivary glands after inoculation (Figure S3), consistent with with mosPTP-1-Ex (Figure 5A). We then examined whether
the Q-PCR data (Figure 1A). mosGCTL-1 was identified on the mosPTP-1-expressing cells could recruit more WNV in the pres-
basement membrane of salivary glands in uninfected mosqui- ence of mosGCTL-1. mosGCTL-1 and WNV were added to cells
toes, and throughout this tissue after infection, suggesting that and incubated at 4 C for membrane attachment. After washing,
mosGCTL-1 was induced by WNV infection of the salivary the cells were transferred to room temperature and collected at
glands (Figure S3). different time points to determine the viral burden (Krishnan
et al., 2007). mosPTP-1-expressing cells incorporated up to
mosPTP-1 Captures mosGCTL-1 onto the Cell Surface 5- to 10-fold more WNV in the presence of mosGCTL-1, com-
Human mannose-binding lectin (MBL) (Zelensky and Gready, pared to control groups (Figure 5B). We then examined the role
2005) and mosGCTL-1 are both secreted. Human MBL is of the mosGCTL-1/mosPTP-1 pathway for WNV infection in
thought to interact with several surface receptors to exert pleo- A. aegypti. We knocked down the mosPTP-1 gene with dsRNA
trophic effects (Baldwin and Ostergaard, 2001; Arnold et al., and then inoculated the mosGCTL-1/WNV mixture at 3 days
2006). We therefore postulated that secreted mosGCTL-1 after RNAi silencing. Silencing of mosPTP-1 interfered with the
captures WNV and presents it to a ligand on the cell surface, ability of mosGCTL-1 to facilitate WNV infection (shown in Fig-
thereby facilitating viral entry. To assess this, we identified 11 ure 3D) at 3 and 6 days after challenge (Figure 5C). The viral
A. aegypti homologs of human proteins that putatively interact burden of the mosPTP-1 RNAi/mosGCTL-1/WNV group was
with human MBL (Table S3) from the Human Protein Reference 2- to 3-fold lower than that of the mock/mosGCTL-1/WNV group
Database (http://www.hprd.org/) and examined their roles in (p < 0.001) and decreased to a level similar to that of the group
WNV infection of A. aegypti (Figure S4A). One mosquito CD45 that did not receive mosGCTL-1, suggesting that mosGCTL-1
homolog (AAEL013105, mosPTP-1) exhibited a phenotype and mosPTP-1 cooperate in the same pathway to enhance
similar to that of mosGCTL-1. The viral burden was significantly WNV infection. To further determine the relationship between
decreased in the mosPTP-1 dsRNA-treated mosquitoes (p < mosGCTL-1 and mosPTP-1 in infection, we silenced both of
0.002) (Figures 4A and 4B). To further determine whether these genes with dsRNA. These genes were successfully
mosPTP-1 interacts with mosGCTL-1, we cloned and expressed knocked down in mosquitoes in the cosilenced group (Figures
the extracellular region of mosPTP-1 (mosPTP-1-Ex) in a S5A and S5B). The decrease in the viral burden was similar in
Drosophila cell line (Figure 4C). mosGCTL-1 strongly interacted the cosilenced and individually silenced groups (Figure S5C),
with mosPTP-1-Ex in a coimmunoprecipitation assay (Figure 4D). suggesting that mosPTP-1 is the dominant downstream
We then expressed the mosPTP-1 gene, including the trans- receptor for mosGCTL-1 in the process of WNV infection of
membrane region (91 bp–2202 bp), in S2 cells (Figure 4C) and A. aegypti.
examined the ability of mosPTP-1 to bind mosGCTL-1 on the To better understand the association between mosGCTL-1,
cell surface. A mock DNA vector transfected stable S2 cell line mosPTP-1, and WNV in vivo, we examined the distribution of
was used as control. More than 40% of the mosPTP-1-express- mosPTP-1 in A. aegypti. mosPTP-1 was highly expressed in
ing cells bound mosGCTL-1 and showed double-positive stain- various mosquito tissues but not induced by WNV infection
ing in FACS (Figure 4E, i) compared with controls (Figure 4E, ii, (Figure S4D). The salivary glands and hemolymph were sites of
iii). Confocal microscopy demonstrated that cells expressing abundant mosPTP-1 expression, while expression in midgut

hemocytes. The yellow arrows show the infected hemocytes. Images were examined with a Zeiss LSM 510 Meta Confocal Microscope 633 objective
lens.
See also Figure S3.

Cell 142, 714–725, September 3, 2010 ª2010 Elsevier Inc. 719


Figure 4. mosPTP-1 Captures mosGCTL-1 onto the Cell Surface
(A) RNAi efficiency for the mosPTP-1 gene at 6 days after dsRNA treatment. The amount of mosPTP-1 mRNA was determined by SYBR Green RT-QPCR and
normalized with A. aegypti actin. Data are represented as the mean ± standard error.
(B) Silencing mosPTP-1 decreased WNV infection. The viral burden was measured at day 6. WNV (10 M.I.D50) was injected into each mosquito. The viral load was
determined by Taqman RT-QPCR and normalized with A. aegypti actin. Statistical analysis was done with the Mann-Whitney test. The horizontal line depicts the
medians. The result shown is a combination of three independent experiments.
(C) Expression of recombinant mosPTP-1 and mosPTP-1-Ex in S2 cells. mosPTP-1 and mosPTP-1-Ex genes were isolated from complementary DNA (cDNA)
library of A. aegypti and expressed as recombinant proteins with an HA tag at the N terminus. The left panel is mosPTP-1, and the right panel is mosPTP-1-EX,
probed by anti-HA tag mAb in western blot. The control was the products from S2 cells transfected with mock DNA vector.
(D) mosGCTL-1 interacts with mosPTP-1-Ex peptide by coimmunoprecipitation. The protein complex was pulled down with a rabbit HA antibody and probed with
a V5-HRP mAb. The experiments were repeated three times.

720 Cell 142, 714–725, September 3, 2010 ª2010 Elsevier Inc.


was comparatively lower (Figures S4E–S4G). We next generated into mosquitoes binds to secreted mosGCTL-1 in the hemo-
mosPTP-1 antibody in mice, which recognized native mosPTP-1 lymph, thereby forming a complex in the extracellular milieu
protein (Figure S5D) and the mosPTP-1-Ex expressed by S2 that has the ability to interact with the membrane protein,
cells (Figure S5E). We then determined the relationship between mosPTP-1, to facilitate cellular entry. The virus rapidly replicates
mosGCTL-1, mosPTP-1, and WNV in salivary glands by immu- in the mosquito thorax. This induces additional mosGCTL-1
nofluorescence. mosPTP-1 was copiously expressed on the expression, which accelerates formation of the mosGCTL-1/
cell surface of salivary glands (Figure 5D), thereby providing WNV complex—enabling WNV to invade different mosquito
additional data to complement and extend the initial QPCR tissues and enhancing viral spread throughout the mosquito
expression data (Figure S4E). Several regions in the salivary body. This mechanism, which involves WNV associating with
glands with substantial mosPTP-1 also demonstrated staining mosGCTL-1 and then being captured by mosPTP-1 onto the
for mosGCTL-1 and WNV (Figure 5D). cell surface in mosquitoes, suggests that an extracellular soluble
protein is an important receptor for flavivirus in arthropods.
mosGCTL-1 Antisera Interferes with WNV Infection mosGCTL-1 shares homology with human MBL. In mammals,
of Mosquitoes MBL is a pattern recognition molecule that recognizes carbohy-
Disruption of the transfer of WNV from the vertebrate to drate moieties on invading microbes (Neth et al., 2002). As exam-
arthropod host could theoretically diminish viral dissemination ples, MBL interacts with HIV envelope protein (gp120) (Saifuddin
in nature. We therefore investigated whether mosGCTL-1 anti- et al., 2000) and HBV surface antigen (HBsAg) (Chong et al., 2005)
sera reduces WNV infection in mosquitoes during the blood and has a role in the opsonization of HIV (Ezekowitz et al., 1989).
meal. We generated mosGCTL-1 antisera in rabbits and showed In these processes, MBL associates with serine proteases,
that mosGCTL-1 antisera interfered with mosGCTL-1 binding to MASPs, and activates the complement system (Neth et al.,
WNV E protein in vitro (Figure 6A). Then, we examined whether 2002). Homologs of the proteins that associate with mammalian
mosGCTL-1 antisera influenced the ability of WNV to infect MBL have not been found in A. aegypti (Table S3), suggesting that
mosquitoes during a blood meal. We mixed mosGCTL-1 antisera the A. aegypti mosGCTL-1 may have different physiological func-
and WNV with fresh whole blood and performed membrane tions than mammalian MBL. Invertebrates lack antibody- and
blood feeding with a Hemotek. Seven days later, mosquitoes interferon-based immune responses (Cheng et al., 2009). Since
were sacrificed to determine the infectivity rate. mosGCTL-1 lectin expression is significantly upregulated by microbial infec-
antisera efficiently blocked WNV infection of A. aegypti. The tion, these molecules are presumed to participate in nonself
number of infected mosquitoes was reduced in mosGCTL-1 recognition and pathogen resistance (Wilson et al., 1999; Tanji
antisera treated groups, compared to mock group, by QPCR et al., 2006). Indeed, recent studies have shown that a comple-
(Figure 6B) or a 50% tissue culture infective doses (TCID50) assay ment-like system exists in the hemolymph of Anopheles gambiae
(Figure 6C). Hence, a humoral response against mosGCTL-1 in and mediates parasite killing (Blandin et al., 2004; Povelones
a vertebrate host may alter WNV infection of mosquitoes during et al., 2009). It is possible that mosGCTL-1 and other subtypes
the feeding process. This hypothetically affords a strategy to in this family, similar to their mammalian homologs, may normally
develop a transmission blocking vaccine to control WNV recognize most pathogens and be involved in the arthropod
dissemination in nature. complement-like system. Nevertheless, our studies showed
that the expression of mosGCTL-1 is induced by WNV infection.
DISCUSSION The induced mosGCTL-1 that then binds to virus amplifies WNV
infection. Overall, these suggest a critical role for mosGCTL-1 in
As mosquitoes are prominent vectors for flaviviruses, specific WNV infection of mosquitoes.
interactions between the virus and arthropod likely enhance In the mammalian host, the association between MBL and the
pathogen survival. In the mammalian host, C-type lectins such CD45 external domain primarily occurs in immature T cells and
as DC-SIGN and the mannose receptor augment viral entry affect the development of thymocytes (Baldwin and Ostergaard,
into specific DCs and macrophages (Tassaneetrithep et al., 2001). Mammalian CD45 is expressed on the hemopoietic-orig-
2003; Davis et al., 2006; Miller et al., 2008). Our results show inated nucleated cells (Thomas, 1989); however, the mosquito
that a secreted mosquito C-type lectin, mosGCTL-1, binds to CD45 homolog, mosPTP-1, does not appear to be restricted to
WNV in a calcium-dependent manner and enhances viral infec- particular cells. As a transmembrane protein, mosPTP-1 was
tion. A mosquito homolog of human CD45 (mosPTP-1) recruits abundantly detected in the salivary glands and hemolymph of
mosGCTL-1 to facilitate viral attachment to cells. Based on our mosquitoes. The pattern of mosPTP-1 expression correlated
findings, we envision a model whereby WNV that is inoculated with the distribution of WNV in A. aegypti. In our model, after

(E) mosPTP-1 captured mosGCTL-1 to the cell surface by flow cytometry. A stable cell line was generated to express mosPTP-1 in S2 cells. The purified
mosGCTL-1 was inoculated with mosPTP-1-expressing cells at 4 C. An empty DNA vector transfected stable S2 cell line was used as the control. The interaction
between mosPTP-1 and mosGCTL-1 was investigated by FACS. mosPTP-1 was stained by Alexa-488; mosGCTL-1 was stained by Phycoerythrin (PE). Three
independent experiments yielded similar results, and one representative study is shown in this figure.
(F) Confocal microscopy to examine for mosPTP-1 and mosGCTL-1. mosGCTL-1 was stained with Alexa-488 (green) and mosPTP-1 was identified with Alexa-
546 (red). Nuclei were stained by To-Pro-3 iodide (blue). The images were collected using a Zeiss LSM 510 Meta Confocal Microscope 633 objective lens.
The arrows represent the overlap between mosPTP-1 and mosGCTL-1.
See also Figure S4 and Table S3.

Cell 142, 714–725, September 3, 2010 ª2010 Elsevier Inc. 721


Figure 5. The mosGCTL-1/mosPTP-1 Pathway in WNV Infection
(A) mosGCTL-1 associated with WNV E protein bound to mosPTP-1-Ex. The protein complex was pulled down with an HA antibody and probed with V5-HRP and
flavivirus E protein antibody. The experiment was repeated three times.
(B) mosPTP-1-expressing cells recruit WN virions in the presence of mosGCTL-1. mosGCTL-1 and WNV were added to cells and incubated at 4 C for 1 hr, for
membrane attachment. Cells were gently washed three times with cold PBS and were then moved to room temperature. At different time points—0, 15, and
60 min—cells were collected for total RNA isolation. Control cells were the empty DNA vector transfected stable S2 cell line. Viral attachment was determined
by SYBR Green RT-QPCR and normalized with Drosophila actin 5C (CG4027). Statistical analysis was done with ANOVA. Data are represented as the mean ±
standard error. The results are representative of three independent experiments.
(C) Silencing of mosPTP-1 impairs the function of mosGCTL-1. The mosPTP-1 gene was knocked down with dsRNA treatment. Then the mosGCTL-1/WNV
mixture was inoculated at 3 days after RNAi silencing. The virus burden was measured at 3 (i) and 6 (ii) days after the introduction of virus by Taqman RT-QPCR.
WNV (10 M.I.D50) and mosGCTL-1 (100 pg) were inoculated into each mosquito. Each dot represents the mRNA level in an individual mosquito. Statistical analysis
was done with the Mann-Whitney test. The horizontal line depicts the medians. The result shown is the combination of five independent experiments.
(D) Immunostaining of mosGCTL-1, mosPTP-1, and WNV in A. aegypti salivary glands. mosPTP-1 was stained with anti-mouse IgG Alexa-488 (green),
mosGCTL-1 was identified by anti-rabbit IgG Alexa-546 (red), and WNV E protein was probed with horse anti-E protein IgG and detected by anti-horse IgG
Alexa-633 (blue). The arrows show the sites of overlap between mosGCTL-1, mosPTP-1, and WNV at 6 days after infection. LL, lateral lobe; ML, median lobe
in female A.aegypti salivary glands. Images were examined with a Zeiss LSM 510 Meta Confocal Microscope 253 objective lens.
See also Figure S5.

722 Cell 142, 714–725, September 3, 2010 ª2010 Elsevier Inc.


Figure 6. mosGCTL-1 Antiserum Interferes
with WNV Infection of Mosquitoes
(A) mosGCTL-1 antisera blocked the interaction
between mosGCTL-1 and WNV E protein.
mosGCTL-1 antisera or mock sera was diluted
1000-fold. The protein complex was pulled down
with a V5 mAb, and WNV E protein was detected
with an E protein antibody.
(B and C) mosGCTL-1 antisera interrupted WNV
infection during the blood meal. The antisera or
control sera were diluted 100- or 2000-fold with
fresh whole blood containing 5 3 106 pfu/ml
WNV. Membrane blood-feeding was then per-
formed with a Hemotek. Seven days later, mosqui-
toes were sacrificed to determine the infectivity
rate by Taqman RT-QPCR (B) and TCID50 (C).
Each group included 50 mosquitoes in the QPCR
assay and 32 mosquitoes in TCID50 assay. One
dot corresponds to a mosquito. n, the number of
mosquitoes in each group. The result is represen-
tative of three independent experiments.

our understanding of flavivirus-arthropod


interactions and may aid in the develop-
ment of strategies to target selected
points in the flaviviral life cycle and inter-
fere with these pathogens in nature.

EXPERIMENTAL PROCEDURES

Mosquitoes and Viruses


A. aegypti and C. quinquefasciatus mosquitoes
were maintained in a sugar solution at 27 C and
80% humidity according to standard rearing
procedures (Keene et al., 2004; Xi et al., 2008).
binding to mosGCTL-1, WNV binds to membrane bound WNV strain 2471 and DENV-2 (DENV New Guinea C strain) were passaged
mosPTP-1. This implies that mosGCTL-1 and mosPTP-1 are in mosquito C6/36 cells (Hanna et al., 2005; Lin et al., 2007; Krishnan et al.,
recruited as receptors to facilitate cellular invasion by WNV. 2008). The titer of WNV for cell culture was determined by a plaque formation
assay as described previously (Bai et al., 2007). The viruses for in vivo exper-
Mosquito control is a common strategy to influence
iments were titrated in mosquitoes through thoracic microinjection. The dose
WNV numbers in nature (van der Meulen et al., 2005; Dauphin of viruses was determined by 10-fold serial dilutions (i.e., 10 4, 10 5, 10 6, and
and Zientara, 2007). The increase in viral spread and fatalities 10 7) in PBS. The mosquitoes (12 in each group) were inoculated in the thorax
over the last decade (Reisen and Brault, 2007; Lindsey et al., by microinjection with 300 nl of diluted virus. On day 6, the mosquitoes were
2009) (http://www.cdc.gov/ncidod/dvbid/westnile/index.htm) sacrificed, total RNA was isolated, and the viral load determined by RT-
suggests that additional strategies could assist in combating QPCR (Figure S1). The 50% mosquito infective dose (M.I.D50) was estimated
by the Reed-Muench method (Pizzi, 1950).
WNV. For arthropod-borne microbes, vector ligands that interact
with pathogens are potential targets for interfering with Gene Silencing and Viral Challenge in Mosquitos
the successful acquisition of the microbe from the vertebrate dsRNA synthesis was performed as described previously (Brackney et al.,
host. As an example, blocking the tick gut receptor for the 2008). The primers are shown in Table S4. For silencing the target genes, adult
Lyme disease agent limits the colonization of ticks by Borrelia female mosquitoes were kept on ice for 15 min and then transferred to a cold
burgdorferi (Pal et al., 2004). Our studies show that blocking tray to receive a systemic injection of dsRNA into the hemocoele. Two micro-
grams of dsRNA/300 nl in PBS was microinjected into the thorax of each
mosGCTL-1 within A. aegypti reduced the vector competence
mosquito. After a 3 day recovery period, the mosquitoes were microinjected
for WNV and interrupted the infective cycle of WNV. These with either WNV 10 M.I.D50/ 300 nl for functional studies or 1000 M.I.D50/
results indicate that it is theoretically possible to develop a trans- 300 nl for expression profile assays.
mission-blocking vaccine to interfere with the migration of WNV
from vertebrates to mosquito, thereby restricting viral dissemi- Purification of mosGCTL-1 with the Drosophila Expression System
mosGCTL-1 was amplified by RT-PCR from adult female A. aegypti. The
nation in the environment.
primers are shown in Table S4. The PCR product was subcloned into the
In summary, we identified a lectin-based pathway to facilitate
pMT/BiP/V5-His A vector (Invitrogen, Carlsbad, CA) and transfected into
flaviviral entry, in which mosGCTL-1 and mosPTP-1 are cascade Drosophila S2 cells in combination with the hygromycin selection vector
receptors in WNV infection of A. aegypti and C. quinquefascia- pCo-Hygro for stable transfection. The cells were selected through the
tus. Characterization of mosquito ligands for WNV enhances use of 300 mg/ml Hygromycin-B (Invitrogen) for 4 weeks. The resistant cells

Cell 142, 714–725, September 3, 2010 ª2010 Elsevier Inc. 723


were grown in spinner flasks, switched to Express Five serum-free medium Received: September 12, 2009
(GIBCO, Invitrogen) for 3 days, and induced with copper sulfate at a final Revised: April 5, 2010
concentration of 500 mM for 4 days. The culture medium was cleared by Accepted: July 27, 2010
centrifugation at 1000 3 g for 5 min and collected for protein purification Published online: August 26, 2010
with the Talon metal affinity resin (Clontech, Mountain View, CA). The
protein was eluted with 150 mM imidazole, extensively dialyzed against REFERENCES
PBS (pH 7.8), and concentrated by centrifugal filtration through a 5 kDa
filter (Millipore, Bedford, MA). The protein purity was checked by SDS- Alonso, A., Sasin, J., Bottini, N., Friedberg, I., Friedberg, I., Osterman, A., God-
PAGE, and immunoblots were performed with an anti-V5-HRP mouse zik, A., Hunter, T., Dixon, J., and Mustelin, T. (2004). Protein tyrosine phospha-
mAb (Invitrogen). tases in the human genome. Cell 117, 699–711.
Arnold, J.N., Dwek, R.A., Rudd, P.M., and Sim, R.B. (2006). Mannan binding
lectin and its interaction with immunoglobulins in health and in disease. Immu-
Isolation and Image of Mosquito Tissues
nol. Lett. 106, 103–110.
Salivary glands were dissected as the previously described (Coleman et al.,
2007). Tissues were isolated, placed on sialylated slides (PGC Scientific, Bai, F.W., Town, T., Pradhan, D., Cox, J., Ashish, Ledizet, M., Anderson, J.F.,
Gaithersburg, MD), washed in PBS, and fixed in 4% PFA at 37 C for 1 hr. Flavell, R.A., Krueger, J.K., Koski, R.A., and Fikrig, E. (2007). Antiviral peptides
For hemolymph isolation, the mosquitoes were anesthetized on ice tray, and targeting the west nile virus envelope protein. J. Virol. 81, 2047–2055.
then the proboscis was removed with forceps. Hemolymph was expelled in Baldwin, T.A., and Ostergaard, H.L. (2001). Developmentally regulated
a droplet using the tip of the proboscis upon pressure to the thorax. Only clear changes in glucosidase II association with, and carbohydrate content of, the
droplets were collected, to avoid contamination by the fat body (Han et al., protein tyrosine phosphatase CD45. J. Immunol. 167, 3829–3835.
1999). The liquid was allowed to be dried on sialylated slides and fixed in Blandin, S., Shiao, S.H., Moita, L.F., Janse, C.J., Waters, A.P., Kafatos, F.C.,
4% PFA. Samples were blocked in PBS with 1% BSA and 0.1% Triton and Levashina, E.A. (2004). Complement-like protein TEP1 is a determinant
X-100 at room temperature for 2 hr before antibody incubations. After staining of vectorial capacity in the malaria vector Anopheles gambiae. Cell 116,
by primary and secondary antibodies, slides were imaged with the Multi-Track 661–670.
mode of a Zeiss LSM 510 Meta Confocal Microscope.
Brackney, D.E., Foy, B.D., and Olson, K.E. (2008). The effects of midgut serine
proteases on dengue virus type 2 infectivity of Aedes aegypti. Am. J. Trop.
Membrane Blood Feeding Med. Hyg. 79, 267–274.
Fresh whole blood was obtained from C57BL/6J mice, placed in tubes with Byth, K.F., Conroy, L.A., Howlett, S., Smith, A.J., May, J., Alexander, D.R., and
anticoagulant, and centrifuged at 3000 rpm for 10 min to separate the serum Holmes, N. (1996). CD45-null transgenic mice reveal a positive regulatory role
and cells. The serum was collected and heat inactivated at 57 C for 1 hr. for CD45 in early thymocyte development, in the selection of CD4+CD8+
The blood cells were washed in PBS three times to remove the anticoagulant. thymocytes, and B cell maturation. J. Exp. Med. 183, 1707–1718.
The cells were resuspended with serum. Five million plaque forming units per Chen, S.T., Lin, Y.L., Huang, M.T., Wu, M.F., Cheng, S.C., Lei, H.Y., Lee, C.K.,
milliliter WNV and antisera (or preimmune sera) were added to the treated Chiou, T.W., Wong, C.H., and Hsieh, S.L. (2008). CLEC5A is critical for
blood. About 1 ml of the virus/sera/blood mixture was used on each Hemotek dengue-virus-induced lethal disease. Nature 453, 672–676.
feeder. The feeder was placed on the mosquito container and the mosquitoes Cheng, G., Zhao, X., Li, Z., Liu, X., Yan, W., Zhang, X., Zhong, Y., and Zheng, Z.
were allowed to feed for 1 hr. The container was transferred to 4 C for 20 min to (2009). Identification of a putative invertebrate helical cytokine similar to the
anesthetize the mosquitoes. Carefully selected fed mosquitoes were placed ciliary neurotrophic factor/leukemia inhibitory factor family by PSI-BLAST-
into new containers and reared for 7 days. The mosquitoes were sacrificed; based approach. J. Interferon Cytokine Res. 29, 461–468.
the whole mosquitoes were homogenized in RLT buffer (RNeasy Mini kit,
Chong, W.P., To, Y.F., Ip, W.K., Yuen, M.F., Poon, T.P., Wong, W.H., Lai, C.L.,
QIAGEN) to measure the viral load by RT-QPCR, or grinded in PBS for virologic
and Lau, Y.L. (2005). Mannose-binding lectin in chronic hepatitis B virus infec-
assays.
tion. Hepatology 42, 1037–1045.
Coleman, J., Juhn, J., and James, A.A.A. (2007). Dissection of midgut and
SUPPLEMENTAL INFORMATION salivary glands from Ae. aegypti mosquitoes. J. Vis. Exp. 5, 228.
Dauphin, G., and Zientara, S. (2007). West Nile virus: recent trends in diagnosis
Supplemental Information includes Extended Experimental Procedures, five and vaccine development. Vaccine 25, 5563–5576.
figures, and four tables and can be found with this article online at doi:10.
Davis, C.W., Nguyen, H.Y., Hanna, S.L., Sánchez, M.D., Doms, R.W., and Pier-
1016/j.cell.2010.07.038.
son, T.C. (2006). West Nile virus discriminates between DC-SIGN and
DC-SIGNR for cellular attachment and infection. J. Virol. 80, 1290–1301.
ACKNOWLEDGMENTS Ezekowitz, R.A., Kuhlman, M., Groopman, J.E., and Byrn, R.A. (1989).
A human serum mannose-binding protein inhibits in vitro infection by the
This work was supported by National Institutes of Health grants AI 50031 and human immunodeficiency virus. J. Exp. Med. 169, 185–196.
AI 070343. We thank Yue Zhang, Lei Liu, Lili Zhang, and Sukanya Narasimhan Geijtenbeek, T.B., Torensma, R., van Vliet, S.J., van Duijnhoven, G.C., Adema,
for help with the biochemical studies, Debbie Beck for technical assistance, G.J., van Kooyk, Y., and Figdor, C.G. (2000). Identification of DC-SIGN, a novel
and Michel Ledizet for providing the purified WNV E protein and the flaviviral dendritic cell-specific ICAM-3 receptor that supports primary immune
E protein antibodies. G.C. is a James Hudson Brown-Alexander Brown responses. Cell 100, 575–585.
Coxe Postdoctoral Fellow at the Yale School of Medicine. P.W. is supported Gould, E.A., and Solomon, T. (2008). Pathogenic flaviviruses. Lancet 371,
by a Career Development Award from Northeast Biodefense Center (U54- 500–509.
AI057158-Lipkin). E.F. is an investigator with the Howard Hughes Medical
Gubler, D.J. (1998). Dengue and dengue hemorrhagic fever. Clin. Microbiol.
Institute. G.C. and E.F. designed the experiments and wrote the manuscript;
Rev. 11, 480–496.
G.C. performed the majority of the experiments and analyzed data; J.C. assis-
ted the mosquito techniques; J.F.A. provided the A. aegypti and C. quinque- Halstead, S.B. (2008). Dengue virus-mosquito interactions. Annu. Rev. Ento-
fasciatus mosquitoes and helped with the mosquito feeding; J.D. and F.Q. mol. 53, 273–291.
assisted in FACS and confocal microscopy; and P.W., M.N.K. and J.D. Han, Y.S., Chun, J., Schwartz, A., Nelson, S., and Paskewitz, S.M. (1999).
provided suggestions for the project. All authors reviewed, critiqued, and Induction of mosquito hemolymph proteins in response to immune challenge
provided comments on the text. and wounding. Dev. Comp. Immunol. 23, 553–562.

724 Cell 142, 714–725, September 3, 2010 ª2010 Elsevier Inc.


Hanna, S.L., Pierson, T.C., Sanchez, M.D., Ahmed, A.A., Murtadha, M.M., and Pal, U., Li, X., Wang, T., Montgomery, R.R., Ramamoorthi, N., Desilva, A.M.,
Doms, R.W. (2005). N-linked glycosylation of west nile virus envelope proteins Bao, F., Yang, X., Pypaert, M., Pradhan, D., et al. (2004). TROSPA, an Ixodes
influences particle assembly and infectivity. J. Virol. 79, 13262–13274. scapularis receptor for Borrelia burgdorferi. Cell 119, 457–468.
Hayes, E.B., Komar, N., Nasci, R.S., Montgomery, S.P., O’Leary, D.R., and Pizzi, M. (1950). Sampling variation of the fifty percent end-point, determined
Campbell, G.L. (2005). Epidemiology and transmission dynamics of West by the Reed-Muench (Behrens) method. Hum. Biol. 22, 151–190.
Nile virus disease. Emerg. Infect. Dis. 11, 1167–1173. Povelones, M., Waterhouse, R.M., Kafatos, F.C., and Christophides, G.K.
Hubálek, Z., and Halouzka, J. (1999). West Nile fever—a reemerging mosquito- (2009). Leucine-rich repeat protein complex activates mosquito complement
borne viral disease in Europe. Emerg. Infect. Dis. 5, 643–650. in defense against Plasmodium parasites. Science 324, 258–261.
Keene, K.M., Foy, B.D., Sanchez-Vargas, I., Beaty, B.J., Blair, C.D., and Olson, Reisen, W., and Brault, A.C. (2007). West Nile virus in North America: perspec-
K.E. (2004). RNA interference acts as a natural antiviral response to O’nyong- tives on epidemiology and intervention. Pest Manag. Sci. 63, 641–646.
nyong virus (Alphavirus; Togaviridae) infection of Anopheles gambiae. Proc. Robinson, M.J., Sancho, D., Slack, E.C., LeibundGut-Landmann, S., and Reis
Natl. Acad. Sci. USA 101, 17240–17245. e Sousa, C. (2006). Myeloid C-type lectins in innate immunity. Nat. Immunol. 7,
Klimstra, W.B., Nangle, E.M., Smith, M.S., Yurochko, A.D., and Ryman, K.D. 1258–1265.
(2003). DC-SIGN and L-SIGN can act as attachment receptors for alphaviruses Saifuddin, M., Hart, M.L., Gewurz, H., Zhang, Y., and Spear, G.T. (2000). Inter-
and distinguish between mosquito cell- and mammalian cell-derived viruses. action of mannose-binding lectin with primary isolates of human immunodefi-
J. Virol. 77, 12022–12032. ciency virus type 1. J. Gen. Virol. 81, 949–955.
Krishnan, M.N., Sukumaran, B., Pal, U., Agaisse, H., Murray, J.L., Hodge, Sessions, O.M., Barrows, N.J., Souza-Neto, J.A., Robinson, T.J., Hershey,
T.W., and Fikrig, E. (2007). Rab 5 is required for the cellular entry of dengue C.L., Rodgers, M.A., Ramirez, J.L., Dimopoulos, G., Yang, P.L., Pearson,
and West Nile viruses. J. Virol. 81, 4881–4885. J.L., and Garcia-Blanco, M.A. (2009). Discovery of insect and human dengue
Krishnan, M.N., Ng, A., Sukumaran, B., Gilfoy, F.D., Uchil, P.D., Sultana, H., virus host factors. Nature 458, 1047–1050.
Brass, A.L., Adametz, R., Tsui, M., Qian, F., et al. (2008). RNA interference Soilleux, E.J., Morris, L.S., Leslie, G., Chehimi, J., Luo, Q., Levroney, E., Trow-
screen for human genes associated with West Nile virus infection. Nature sdale, J., Montaner, L.J., Doms, R.W., Weissman, D., et al. (2002). Constitutive
455, 242–245. and induced expression of DC-SIGN on dendritic cell and macrophage
Leis, A.A., Stokic, D.S., Polk, J.L., Dostrow, V., and Winkelmann, M. (2002). subpopulations in situ and in vitro. J. Leukoc. Biol. 71, 445–457.
A poliomyelitis-like syndrome from West Nile virus infection. N. Engl. J. Med. Tanji, T., Ohashi-Kobayashi, A., and Natori, S. (2006). Participation of a galac-
347, 1279–1280. tose-specific C-type lectin in Drosophila immunity. Biochem. J. 396, 127–138.
Lin, C.C., Yang, C.F., Tu, C.H., Huang, C.G., Shih, Y.T., Chuang, C.K., and Tassaneetrithep, B., Burgess, T.H., Granelli-Piperno, A., Trumpfheller, C.,
Chen, W.J. (2007). A novel tetraspanin C189 upregulated in C6/36 mosquito Finke, J., Sun, W., Eller, M.A., Pattanapanyasat, K., Sarasombath, S., Birx,
cells following dengue 2 virus infection. Virus Res. 124, 176–183. D.L., et al. (2003). DC-SIGN (CD209) mediates dengue virus infection of human
Lindsey, N.P., Hayes, E.B., Staples, J.E., and Fischer, M. (2009). West Nile dendritic cells. J. Exp. Med. 197, 823–829.
virus disease in children, United States, 1999-2007. Pediatrics 123, e1084– Thomas, M.L. (1989). The leukocyte common antigen family. Annu. Rev.
e1089. Immunol. 7, 339–369.
Miller, J.L., de Wet, B.J., deWet, B.J., Martinez-Pomares, L., Radcliffe, C.M., Trowbridge, I.S., and Thomas, M.L. (1994). CD45: an emerging role as a protein
Dwek, R.A., Rudd, P.M., and Gordon, S. (2008). The mannose receptor medi- tyrosine phosphatase required for lymphocyte activation and development.
ates dengue virus infection of macrophages. PLoS Pathog. 4, e17. Annu. Rev. Immunol. 12, 85–116.
Monath, T.P., and Heinz, F.X. (1996). Flaviviruses (Philadelphia: Lippincott- Turell, M.J., O’Guinn, M., and Oliver, J. (2000). Potential for New York mosqui-
Raven Publishers). toes to transmit West Nile virus. Am. J. Trop. Med. Hyg. 62, 413–414.
Mustelin, T., Vang, T., and Bottini, N. (2005). Protein tyrosine phosphatases van der Meulen, K.M., Pensaert, M.B., and Nauwynck, H.J. (2005). West Nile
and the immune response. Nat. Rev. Immunol. 5, 43–57. virus in the vertebrate world. Arch. Virol. 150, 637–657.
Nene, V., Wortman, J.R., Lawson, D., Haas, B., Kodira, C., Tu, Z.J., Loftus, B., Vanlandingham, D.L., McGee, C.E., Klinger, K.A., Vessey, N., Fredregillo, C.,
Xi, Z., Megy, K., Grabherr, M., et al. (2007). Genome sequence of Aedes and Higgs, S. (2007). Relative susceptibilties of South Texas mosquitoes to
aegypti, a major arbovirus vector. Science 316, 1718–1723. infection with West Nile virus. Am. J. Trop. Med. Hyg. 77, 925–928.

Neth, O., Jack, D.L., Johnson, M., Klein, N.J., and Turner, M.W. (2002). Wilson, R., Chen, C.W., and Ratcliffe, N.A. (1999). Innate immunity in insects:
Enhancement of complement activation and opsonophagocytosis by the role of multiple, endogenous serum lectins in the recognition of foreign
complexes of mannose-binding lectin with mannose-binding lectin-associ- invaders in the cockroach, Blaberus discoidalis. J. Immunol. 162, 1590–1596.
ated serine protease after binding to Staphylococcus aureus. J. Immunol. Xi, Z., Ramirez, J.L., and Dimopoulos, G. (2008). The Aedes aegypti toll
169, 4430–4436. pathway controls dengue virus infection. PLoS Pathog. 4, e1000098.
Nir, Y., Goldwasser, R., Lasowski, Y., and Margalit, J. (1968). Isolation of West Zelensky, A.N., and Gready, J.E. (2005). The C-type lectin-like domain super-
Nile virus strains from mosquitoes in Israel. Am. J. Epidemiol. 87, 496–501. family. FEBS J. 272, 6179–6217.

Cell 142, 714–725, September 3, 2010 ª2010 Elsevier Inc. 725


The ATAC Acetyltransferase Complex
Coordinates MAP Kinases
to Regulate JNK Target Genes
Tamaki Suganuma,1 Arcady Mushegian,1,2 Selene K. Swanson,1,3 Susan M. Abmayr,1,3 Laurence Florens,1
Michael P. Washburn,1,4 and Jerry L. Workman1,*
1Stowers Institute for Medical Research, 1000 East 50th Street, Kansas City, MO 64110, USA
2Department of Microbiology, Molecular Genetics, and Immunology
3Department of Anatomy and Cell Biology
4Department of Pathology and Laboratory Medicine

University of Kansas Medical Center, Kansas City, KS 66160, USA


*Correspondence: jlw@stowers.org
DOI 10.1016/j.cell.2010.07.045

SUMMARY complex (Lee and Workman, 2007). However, it was unknown


whether ATAC functions as a transcription cofactor like SAGA.
In response to extracellular cues, signal transduction Mitogen-activated protein kinases (MAPKs) and their target
activates downstream transcription factors like kinases in the pathway lead to phosphorylation of target
c-Jun to induce expression of target genes. We transcription factors (Edmunds and Mahadevan, 2004; Thomson
demonstrate that the ATAC (Ada two A containing) et al., 1999). Exposure of cells to increases in extracellular osmo-
histone acetyltransferase (HAT) complex serves as lality results in rapid activation of stress-activated MAPKs
(SAPKs), including c-Jun-NH2-terminal kinase (JNK/basket in
a transcriptional cofactor for c-Jun at the Jun
Drosophila) and p38 MAPKs (de Nadal et al., 2002; Edmunds
N-terminal kinase (JNK) target genes Jra and chick- and Mahadevan, 2004; Kyriakis and Avruch, 2001). Osmotic
adee. ATAC subunits are required for c-Jun occu- stress causes activation of JNK by phosphorylation which, in
pancy of these genes and for H4K16 acetylation at turn, phosphorylates c-Jun and enhances its transcriptional
the Jra enhancer, promoter, and transcribed se- activity (Kayali et al., 2000; Thomson et al., 1999). MAPKs are
quences. Under conditions of osmotic stress, ATAC more intimately involved in the regulation of downstream target
colocalizes with c-Jun, recruits the upstream kinases genes than just phosphorylation of target transcription factors
Misshapen, MKK4, and JNK, and suppresses further (Dioum et al., 2009). For example, the yeast SAPK Hog1 is
activation of JNK. Relocalization of these MAPKs recruited to target genes in chromatin and interacts with tran-
and suppression of JNK activation by ATAC are scription factors, cofactors, and RNA polymerase II (Alepuz
dependent on the CG10238 subunit of ATAC. Thus, et al., 2001, 2003; de Nadal et al., 2004; Mas et al., 2009; Pokho-
lok et al., 2006; Proft et al., 2006; Zapater et al., 2007). Recently,
ATAC governs the transcriptional response to MAP
the ERK2 MAPK was found to bind with sequence specificity
kinase signaling by serving as both a coactivator to DNA and act as a transcriptional repressor of interferon-g-
of transcription and as a suppressor of upstream induced genes (Hu et al., 2009). Moreover, multiple MAPKs,
signaling. including ERK1/2, p38, and JNK, bind to and regulate transcrip-
tion of the insulin gene (Lawrence et al., 2009). Thus, MAPKs play
INTRODUCTION important chromatin-associated functions in the regulation of
gene expression.
Histone acetyltransferase complexes have been isolated from MBIP (MAPK upstream kinase [MUK] binding inhibitory
multiple organisms and shown to be involved in nuclear events protein), a component of the human ATAC complex, was
that relate to chromatin biology (Kimura et al., 2005; Lee and identified as a MUK binding partner in a yeast two-hybrid screen
Workman, 2007). The Drosophila ATAC (Ada two A containing) (Fukuyama et al., 2000). The Drosophila ATAC component
complex consists of 13 proteins and includes two distinct CG10238 encodes the Drosophila homolog of MoaE, a subunit
histone acetyltransferases, Gcn5/KAT2 and Atac2/KAT14 (Fig- of molybdopterin (MPT) synthase, an essential enzyme involved
ure 1A). Whereas Gcn5/KAT2 preferentially acetylates histones in the synthesis of the molybdenum cofactor (Moco). Moco binds
H3K9 and H3K14, Atac2/KAT14 acetylates H4K16 (Ciurciu molybdenum in the active site of molybdenum enzymes, which
et al., 2006; Guelman et al., 2006; Suganuma et al., 2008). The catalyze redox reactions as part of ancient and conserved
Gcn5/KAT2, Ada3, and CG30390 (Sgf29 in yeast) subunits of biosynthetic pathways (Iyer et al., 2006; Leimkuhler et al.,
ATAC are shared with SAGA (Spt-Ada-Gcn5 acetyltransferase) 2003; Rudolph et al., 2001; Schwarz and Mendel, 2006).
(Suganuma et al., 2008), an important transcriptional coactivator However, CG10238 also contains C-terminal sequences that

726 Cell 142, 726–736, September 3, 2010 ª2010 Elsevier Inc.


A Figure 1. Peptides from Proteins Related to
the JNK Pathway Were Detected in Purifica-
Purifications
tions of ATAC Subunits, and CG10238
CG10238 Atac2 CHRAC14 Ada2a WDS D12 Control
Proteins PEP (SC%) PEP (SC%) PEP (SC%) PEP (SC%) PEP (SC%) PEP (SC%) PEP (SC%) Inhibits JNK Activation by Osmotic Stress
ATAC CG10238 29 (72.21) 5 (21.25) 2 (9.81) 7 (25.61) 6 (30.25) 16 (53.13) 0 (0)
Gcn5/KAT 35 (46.69) 4 (6.15) 4 (6.4) 5 (8.24) 4 (7.38) 33 (33.95) 0 (0) in Drosophila Schneider’s S2 Cells
Atac1 14 (53.37) 2 (5.34) 2 (7.58) 6 (16.3) 0 (0) 9 (20.71) 0 (0) (A) Peptides of MAPK pathway proteins in addition
Ada3 21 (58.89) 1 (1.98) 1 (1.98) 2 (7.19) 1 (3.24) 10 (16.37) 0 (0)
Ada2a 20 (48.2) 2 (4.93) 2 (4.93) 9 (18.6) 0 (0) 18 (36.43) 0 (0) to ATAC subunits detected by MudPIT analysis for
HCF 66 (68.87) 11 (11.93) 16 (17.6) 17 (19.87) 19 (18.07) 54 (49.4) 1 (2.6)
D12 37 (45.86) 7 (9.08) 11 (19.81) 10 (13.62) 5 (9.7) 31 (38.29) 0 (0) affinity purifications of ATAC subunits with FLAG-
CG30390 14 (56.60) 1 (5.88) 0 (0) 6 (26.64) 0 (0) 13 (55.36) 0 (0)
Atac2/KAT14 26 (47.16) 10 (22.09) 7 (16.28) 5 (9.95) 5 (13.18) 19 (39.41) 0 (0)
HA-tagged CG10238, Atac2/KAT14, Ada2a,
CHARC14 5 (62.5) 1 (10.16) 1 (10.16) 3 (27.34) 1 (14.84) 7 (71.88) 0 (0) CHRAC14, WDS, and D12. Parental S2 cells
Atac3 14 (40.88) 1 (3.89) 0 (0) 3 (7.61) 0 (0) 9 (20.71) 0 (0)
NC2 beta 6 (60.66) 2 (14.21) 1 (7.1) 0 (0) 2 (14.21) 5 (27.32) 0 (0) were used in mock purification as a control. PEP,
WDS 18 (66.48) 7 (25.76) 5 (18.28) 8 (26.59) 20 (63.43) 13 (53.19) 1 (3.6)
Proteins Jra 2 (10.03) 0 (0) 3 (19.03) 0 (0) 0 (0) 2 (13.84) 0 (0)
peptide count; SC%, sequence coverage (%).
related to Misshapen 16 (21.34) 0 (0) 5 (6.12) 0 (0) 0 (0) 5 (5.85) 0 (0) (B) Homology domains of CG10238 and experi-
JNK pathway MKK4 4 (15.8) 0 (0) 0 (0) 0 (0) 0 (0) 0 (0) 0 (0)
slik 5 (4.82) 0 (0) 0 (0) 0 (0) 0 (0) 0 (0) 0 (0) mental design of truncation mutants. The diagram
MEK3/MKK3 2 (8.08) 0 (0) 0 (0) 0 (0) 0 (0) 0 (0) 0 (0)
Chickadee 2 (20.63) 0 (0) 0 (0) 0 (0) 0 (0) 0 (0) 0 (0)
shows the domains of CG10238 that are homolo-
gous to human MOCS2B (hMoaE) and to human
MBIP, which are separate proteins. The full-length
B (FL), N-terminal that contains only MoaE domain
Drosophila melanogaster
(N), and C-terminal that contains only MBIP protein
1 137 171 367 a.a. sequences (C) were stably expressed in S2 cells
MoaE MBIP
(Figure 2; see Figure S1B).
Homo sapiens
1 188 a.a.
MoaE
(C) CG10238 inhibited the activation of JNK stimu-
Mocs2B
1 344 a.a. lated by osmotic stress in S2 cells. Stably ex-
MBIP pressed FLAG-HA-tagged CG10238 was induced
Design of deletion mutants of CG10238 by 0, 50, or 150 mM CuSO4 in S2 cells. Cells were
FL
1 367a.a.
stimulated with osmotic stress by addition of
1 137a.a.
-HF
500 mM sorbitol for 12 min before harvesting cells
N
137a.a. 367a.a. (C, lanes 6–10; Figure 2C, lanes 5–14; Figure 2D,
C -HF
lanes 1–12; see Figures S2A–S2C). The nuclear
extracts were examined by western blot probing
with anti-HA, anti-Active JNK, and anti-JNK
C (C; Figure 2C). Parental S2 cells of stable cell lines
CuSO4 (x10µM) - 15 - 5 15 - 15 0 5 15
cultured with 0 or 150 mM CuSO4 were used as
HA-CG10238 - - - - controls (C, lanes 1, 2, 6, and 7; see Figure S3).
Parental cells + + - - - + + - - - See also Figures S1, S2, and S3.
Sorbitol - - - - - + + + + +
1 2 3 4 5 6 7 8 9 10

anti-HA 53.9

53.9
anti-ActiveJNK

53.9
anti-JNK

are homologous to human MBIP. This observation, coupled with genetically with the JNK pathway (Jasper et al., 2001; Morrison
the presence of MAPK signaling proteins in ATAC purifications, et al., 2000).
led us to ask whether CG10238 and ATAC play a role in MAPK
signaling. The ATAC Subunit CG10238 Functions as an Inhibitor
of JNK Activation in Response to Osmotic Stress
RESULTS We tested whether CG10238 plays a role in MAPK signaling like
MBIP (Fukuyama et al., 2000). MAPK cascades can be activated
ATAC Interacts with Proteins Related to the MAPK by osmotic stress, resulting in activation of JNK by phosphoryla-
Pathway tion (Kayali et al., 2000; Yang et al., 2003). We therefore exam-
Affinity purifications of the ATAC complex revealed proteins that ined whether expression of CG10238 affected the activation of
are part of the MAPK signaling pathway (Figure 1A; see Fig- JNK under conditions of osmotic stress (Kayali et al., 2000).
ure S1A available online). Peptides from these proteins were We first titrated the cellular response to osmotic stress
found in purifications via the CG10238, CHRAC14, and D12 stimulated by sorbitol in S2 cells by western blot (Figures S2A
ATAC subunits. Peptides were identified from the transcription and S2B). Maximum activation of JNK was observed between
factor Jra (Jun-related antigen), the Drosophila homolog of 7 and 30 min after treatment with a minimum concentration of
c-Jun, and Misshapen (MSN), the Drosophila homolog of 500 mM sorbitol (Figure S2A, lane 5; Figure S2B, lanes 2–4).
Ste-20 kinase (Figure 1A) (Morrison et al., 2000; Su et al., 1998; We then examined the level of JNK activation in cells expressing
Treisman et al., 1997). We also found peptides from other STE CG10238 in the presence or absence of osmotic stress induced
kinases, such as MKK4, slik, and MEK3/MKK3, as well as by 500 mM sorbitol for 12 min. Expression of CG10238 was
peptides from Chickadee, which has been shown to interact inducible by CuSO4, and parental S2 cells were treated with

Cell 142, 726–736, September 3, 2010 ª2010 Elsevier Inc. 727


A B Figure 2. MBIP-Related Sequences Incor-
Input nuclear extracts Elution
porate CG10238 into ATAC; However, the
- F N C F N C - (KDa)
CG10238-Domain N C MoaE Domain Inhibits JNK Activation
ATAC subunits PEP( SC%) PEP( SC%) 54.8
(A) FLAG-HA-tagged N terminus that contains only
CG10238-PA 7 (25.6) 17 (50.95)
Gcn5/KAT 0 (0) 8 (12.67)
the MoaE domain (N) and C terminus that contains
37.7
anti-HA only MBIP protein sequences (C) were stably ex-
Atac1 0 (0) 3 (9.55)
Ada3 0 (0) 3 (8.99) 29.4 pressed in S2 cells purified by FLAG affinity beads.
Ada2a 0 (0) 5 (13.09) The copurified proteins were examined by MudPIT
HCF 5 (1.47) 24 (26.8) 17.5
D12 0 (0) 15 (21.26) 97.3 analysis (see Figures S1A and S1B). The observed
anti-Atac1
CG30390 0 (0) 3 (13.15) PEP and SC (%) of each sample are indicated as in
Atac2/KAT14 0 (0) 5 (10.98) 118.2
anti-Atac2 Figure 1.
CHRAC14 0 (0) 0 (0)
anti-Atac3
97.3 (B) The copurified proteins of CG10238-full-length
Atac3 0 (0) 2 (6.55)
NC2beta 0 (0) 3 (14.21) 206.3 (F), -N, or -C (Figure 1B; Figure S1) were analyzed
anti-D12
118.2
WDS 1 (3.88) 6 (27.42) 118.2 by western blotting with antibodies against HA tag
anti-Gcn5
and ATAC subunits. As a control, untransfected
anti-Ada2a 54.8
parental S2 cells were mock purified and analyzed
anti-Hcf
97.3
by western blotting (-).
29.4 (C and D) The MoaE domain inhibits JNK activa-
anti-NC2 beta tion. The ability to inhibit JNK activation was
54.8 compared between FLAG-HA-tagged CG10238
anti-WDS
(FL), CG10238-N (MoaE domain), and CG10238-
37.6
anti-CG30390 C (MBIP domain) after the expression levels were
normalized (see also Figure S1B). Parental S2 cells
cultured with 150 mM CuSO4 were used as a posi-
tive control (C, lanes 1 and 5; D, lanes 1, 5, and 9).
C D Osmotic stress was induced as in Figure 1C
HA-C - - - + - - - - - - - (see also Figures S2A and S2B). For sorbitol-stim-
HA-N - - + - - - - - - - -
Active JNK ulated samples, the intensity of each band for
HA-FL - + - - - - - - - - -
Parental cells + - - - + - - - - - - - - - Active JNK in western blotting was quantified,
160
Sorbitol - - - - + + + + + + + + + +
1 2 3 4 5 6 7 8 9 10 11 12 13 14 140
and each sample from CG10238 (FL), CG10238-N,
120
and CG10238-C was individually compared with
100
the intensity of parental cells (P), and the ratios
Ratio (%)

80
(%) are shown in (D). The average of four indepen-
anti-HA
60
dent experiments is graphed. Error bars represent
40 standard deviation.
20 See also Figures S1 and S2.
0
anti-ActiveJNK 1 2 3 4 5 6 7 8 9 10 11 12

P P P
anti-JNK HA-FL HA-N HA-C

the same amount of CuSO4 and sorbitol as controls. We first examined the two parts of CG10238 to determine which might
confirmed by western blot that the expression level of HA- incorporate it into the ATAC complex. We generated S2 cell lines
tagged CG10238 in the stable cell line without induction was that stably expressed tagged truncated forms of CG10238 that
similar to that of endogenous CG10238 in the parental cells included only the N-terminal MoaE domain or the C-terminal
(Figure S2C). Active JNK was not detected in the absence of MBIP domain (Figure 1B). These tagged proteins were affinity
sorbitol treatment, or upon induction of CG10238 by addition purified from the stable cell lines after their expression levels
of CuSO4 (Figure 1C, lanes 1 and 2). In the presence of sorbitol, were normalized (Figure S1B). Proteins that copurified with
JNK was activated in the parental cells (Figure 1C, lanes 6 and 7). each domain of CG10238 were identified by multidimensional
However, induction of CG10238 expression by CuSO4 inhibited protein identification technology (MudPIT) analysis and
the activation of JNK in a dose-dependent manner (Figure 1C, confirmed by western blots (Figures 2A and 2B; Figure S1A).
lanes 8–10). Thus, CG10238 inhibits JNK activation by osmotic All ATAC subunits except CHRAC14 associated with the MBIP
stress in vivo. Because JNK is also activated by ultraviolet light domain. A few peptides from two of the ATAC subunits were de-
(UV), we examined whether the inhibitory activity of CG10238 tected by purification with the MoaE domain and three subunits
extended to this JNK activation mechanism (Angel et al., 1988; were detected weakly by westerns (Figure 2B). Thus, the MBIP
Devary et al., 1991; Rozek and Pfeifer, 1995). Expression of domain of CG10238 is responsible for incorporating the protein
CG10238 also inhibited activation of JNK by UV (Figure S3). into the ATAC complex.

The MBIP Domain of CG10238 Is Required The MoaE Domain of CG10238 Inhibits JNK Activation
for Incorporation into the ATAC Complex Analysis of the domain structure of MoaE and MBIP orthologs in
The finding that CG10238 prevented JNK activation when a variety of organisms indicates that many insects encode these
expressed in vivo led us to ask whether CG10238 served this two domains as a translational fusion like that of Drosophila
function in isolation or as part of the ATAC complex. We first CG10238. By contrast, orthologs in nematodes, plants, fungi,

728 Cell 142, 726–736, September 3, 2010 ª2010 Elsevier Inc.


B S2 Cells Figure 3. ATAC Inhibits JNK Signaling
A S2 Cells ta ta
nt nt c2 c2 t t be be S2 cells were transfected with dsRNA-Atac2, dsRNA-NC2
on Con NC2 NC2
Co A-Co -Ata A-Ata -C
A- A -
NA RNA RNA RNA
- -
b, or dsRNA-Control (dsRNA-Cont) (A and B) (see
RN sRN sRN sRN ds
R
ds ds ds
Transfected with ds d d d Transfected with
- + - + (KDa) Sorbitol - + - + Figure S4). 293T cells were transfected with dsRNA-
Sorbitol (KDa)
118.1
anti-NC2beta 29.4 hAtac2/CSRP2BP, dsRNA-MBIP, or dsRNA-Control
anti-Atac2
(dsRNA-Cont) (C and D). The knockdown cells were then
54.8 54.8
anti-ActiveJNK anti-ActiveJNK stimulated with or without 500 mM (for S2 cells) or
54.8 54.8
400 mM (for 293T cells) sorbitol as in Figure 1C (see
anti-JNK anti-JNK
Figure S2D). The nuclear extracts were examined by
anti-tubulin anti-tubulin western blot probing with anti-Active JNK, anti-JNK,
54.8 54.8
anti-Atac2 (A and C), anti-NC2 b (B), anti-MBIP (D), or
anti-tubulin antibodies as a loading control (A–D, top).
dsRNA-NC2beta
dsRNA-Atac2 The intensities of each band of JNK, Active JNK, Atac2,
800 120 3000 120
NC2 b, hAtac2, or MBIP in western blotting were quantified
Ratio-JNK or Ratio-ActJNK

Ratio-JNK or Ratio-ActJNK
700
100 2500 100
600
from four independent experiments and plotted as ratios to

Ratio-NC2 beta
80 2000 80 the untreated control cells (A–D, bottom). Dark bars show
Ratio-Atac2

500
JNK JNK
400 60 ActJNK 1500 60 ActJNK
the ratio of JNK intensities, light bars show the ratio of
300
Atac2 NC2beta
Active JNK intensities, and the dotted line shows the ratio
40 1000 40
200 of Atac2 (A), NC2 b (B), Atac2/CSRP2BP (C), or MBIP (D)
100
20 500 20
intensities. Error bars represent standard deviation.
0 0 0 0 See also Figures S2 and S4.
1 2 3 4 1 2 3 4

Sorbitol - + - + Sorbitol - + - +
dsRNA-Cont. dsRNA-Atac2 dsRNA-Cont. dsRNA-NC2beta

the inhibitory activities of the full-length protein


C 293T Cells BP 2BP D 293T Cells to that of the MoaE and MBIP domains (Fig-
t t P2 P nt nt IP BIP
on on SR SR Co Co MB ures 2C and 2D). Full-length CG10238 and the
A -C A-C A-C A-C A- NA- NA- NA-M
R N RN R N RN N
Transfected with ds ds ds ds Transfected with d sR ds
R
ds
R
ds
R MoaE domain alone inhibited activation of JNK
Sorbitol - + - + (KDa) Sorbitol - + - + (KDa)
46
(Figure 2C, lanes 6–11; Figure 2D, bars 1–8).
anti-Atac2/CSRP2BP anti-MBIP
80
However, the MBIP domain of CG10238 failed
anti-Active JNK
46
anti-Active JNK
46 to prevent the activation of JNK (Figure 2C,
46
lanes 12–14; Figure 2D, bars 9–12). Thus, the
46
anti-JNK anti-JNK active portion of CG10238 in inhibition of JNK
anti-tubulin anti-tubulin
activation is the MoaE domain, whereas the
46
46
C-terminal sequences connect this activity to
the ATAC complex.
dsRNA-CSRP2BP dsRNA-MBIP

600 140 350 140

ATAC Inhibits JNK Activation


Ratio-JNK or Ratio-ActJNK

Ratio-JNK or Ratio-ActJNK

120 300 120


500
The MoaE domain of CG10238 was sufficient to
Ratio-CSRP2BP

100 250 100


400
prevent JNK activation when expressed in vivo,
Ratio-MBIP

80 JNK 200 80 JNK


300
60
whereas the MBIP domain was required to
ActJNK
CSRP2BP 150 60
ActJNK
MBIP

200
40 incorporate CG10238 into ATAC (Figures 2A– 100 40

100
20 2D). These data raised the question of whether 50 20

0 0 ATAC itself plays a role in inhibition of the JNK 0 0


1 2 3 4 1 2 3 4

Sorbitol - + - +
pathway. To address this question, we exam-
Sorbitol - + - +
dsRNA-Cont dsRNA-CSRP2BP dsRNA-Cont dsRNA-MBIP ined JNK activation in S2 cells where endoge-
nous subunits of ATAC were knocked down by
and all prokaryotes except parasitic bacteria have a MoaE dsRNA interference. The expression level of Atac2 was reduced
homology domain but are missing the MBIP domain. Sequence 60% in cells expressing dsRNA-Atac2 (Figure 3A). Interestingly,
database searches using the PSI-BLAST program (Altschul JNK was partially activated upon reduction of Atac2 even in the
et al., 1997) with human MBIP (GenBank accession number absence of osmotic stress (Figure 3A). Moreover, activation of
119586267) as query revealed sequence matches between the JNK by osmotic stress was enhanced in the Atac2 knockdown
N-terminal portion of MBIP orthologs from Metazoa and MoaE cells (Figure 3A). We observed similar results upon knockdown
proteins. Thus, the N-terminal sequences of mammalian MBIPs of NC2 b or CG10238 subunits of ATAC by dsRNA (Figure 3B;
are related to the MoaE sequences. Hence, our bioinformatic Figure S4A). Although JNK activation was not observed in D12
analysis suggests the N-terminal sequences of MBIP evolved knockdown cells without osmotic stress, its activation was
from MoaE and raises the possibility that the MoaE domain of also increased in these cells under conditions of osmotic stress
CG10238 contributes to the JNK inhibition activity. (Figure S4B). Because human MBIP was recently shown to be a
We generated S2 cell lines that stably expressed tagged component of the human ATAC complex (Fukuyama et al., 2000;
truncated forms containing the MoaE domain (N-terminal) or Wang et al., 2008), it was of interest to determine whether human
MBIP domain (C-terminal) (Figure S1B), and then compared ATAC inhibited the activation of JNK by osmotic stress in human

Cell 142, 726–736, September 3, 2010 ª2010 Elsevier Inc. 729


A B Figure 4. ATAC Is Required for the Transcription
Input IP Input IP Regulation of JNK Target Genes
N
IP with - IgG Jra IP with - IgG M
S (A and B) Extracts from S2 cells endogenously expressing
1 2 3 1 2 3 ATAC subunits and JNK (B) were immunoprecipitated with
anti-Atac2 anti-Atac2 antibodies to endogenous Jra (A) or Misshapen (MSN) (B).
The presence of ATAC subunits, MSN, and JNK in the
anti-CG10238 anti-CG10238 input and immunoprecipitates (IP) was detected by
anti-NC2beta western blots.
anti-NC2 beta (C) Quantitative real-time RT-PCR (qRT-PCR) of Jra
anti-D12
and Chickadee mRNA in S2 cells transfected with
anti-Jra anti-MSN dsRNA-LacZ (Control), dsRNA-Atac2, dsRNA-CG10238,
dsRNA-NC2 b (NC2b), dsRNA-Jra, or dsRNA-MSN (see
anti-JNK
Figure S5A). Gene-specific mRNA levels from the cells
without sorbitol stimulation (C, top) and 30 min after
500 mM sorbitol stimulation (C, bottom) were measured
C
Jra Sorbitol (-) Chickadee Sorbitol (-)
and normalized to RpL-32 expression (Figure S5B), and
are represented as ratios by qRT-PCR measurements.
2 2
1.8 1.8 The average of three independent experiments is graphed.
Chickadee/Control

1.6 1.6
1.4 1.4
Error bars represent standard deviation.
Jra/Control

1.2 1.2 (D) ChIP assays were performed with an antibody against
1 1
0.8 0.8 Jra from S2 cells without sorbitol stimulation (D, left) or with
0.6 0.6
0.4 0.4
sorbitol stimulation (D, right) expressing dsRNA-LacZ
0.2 0.2 (Cont), dsRNA-Atac2, dsRNA-CG10238, or dsRNA-MSN
0 0
1 2 3 4 5 6 1 2 3 4 5 6 (D) (Figures 5B–5M). The association of Jra on the probed
RNAi b a RNAi
cZ c2 38 2 Jr MSN cZ c2 38 2b Jra MSN region of the Jra gene as indicated in Figure 5A, and input
La Ata 102 NC La Ata 102 NC
CG CG
chromatin was measured by qRT-PCR and normalized to
Jra Sorbitol (+) Chickadee Sorbitol (+)
input, and the ratios of quantities are represented (see
4 1.6 Figure S6). The average of three independent experiments
1.4
is graphed. Error bars represent standard deviation.
Chickadee/Control

3 1.2
See also Figures S2, S5, and S6.
Jra/Control

1
2 0.8
0.6
1 0.4
0.2
0 0

RNAi
1 2 3 4 5 6
RNAi
in vivo. Endogenous Atac2, CG10238, NC2 b,
1 2 3 4 5 6
2 8 2b Jra SN a
cZ tac 23 cZ c2 38 2b Jr MSN
La A G10 N C M La Ata 102 NC
and D12 coimmunoprecipitated with endoge-
C CG
nous MSN (Figure 4B).
If ATAC serves as a cofactor for Jra, then
D
ChIP-Jra on Jra Sorbitol (-) ChIP-Jra on Jra Sorbitol (+)
Jra-dependent transcription would require
dsRNA-Cont dsRNA-Atac2 dsRNA-CG10238 dsRNA-MSN dsRNA-Cont dsRNA-Atac2 dsRNA-CG10238 dsRNA-MSN ATAC and ATAC should be localized to Jra target
100.00%
90.00%
genes. Because the gene encoding c-Jun, the
80.00%

70.00%
80.00%
70.00%
mammalian homolog of Jra, is positively
60.00%
ChIP/Input

ChIP/Input

50.00%
60.00%
50.00%
regulated by c-Jun protein (Angel et al., 1988),
40.00%
40.00%
30.00% we examined whether ATAC functions in Jra
30.00%

20.00% 20.00%

10.00% transcription. We measured the levels of Jra


10.00%
0.00%

Probe A B C D E Probe A B C D E
transcripts by quantitative real-time RT-PCR
0.00%

(qRT-PCR) of control and ATAC knockdown S2


cells. As a control, we confirmed that Jra tran-
293T cells (Figure S2D). Indeed, JNK was activated to higher script levels were reduced in cells expressing dsRNA-Jra
levels upon knockdown of MBIP or CSRP2BP (human homolog compared with cells expressing control-dsRNA-LacZ (Figure 4C,
of Atac2) in 293T cells (Figures 3C and 3D). These data indicate upper left). Importantly, Jra transcript levels were significantly
that the ATAC complex is required for complete inhibition of JNK reduced in Atac2 and CG10238 knockdown cells and slightly
activation in both Drosophila and human cells. reduced in NC2 b knockdown cells (Figure 4C, upper left). We
did not detect effects on Jra transcripts in MSN knockdown cells.
ATAC Functions as Transcriptional Cofactor We confirmed that the transcripts of atac2, CG10238, nc2 b, and
for JNK Target Genes msn were reduced by expression of each dsRNA-Atac2, dsRNA-
The presence of peptides from the Jra transcription factor in CG10238, dsRNA-NC2 b, and dsRNA-MSN by qRT-PCR (Fig-
MudPIT analyses of the affinity-purified ATAC complex (Fig- ure S5A). We also observed that the expression of Atac2 was
ure 1A) suggests that ATAC interacts with Jra and may serve increased in Jra knockdown (see below).
as a transcriptional cofactor. Indeed, endogenous Jra was able We next examined chickadee, another Jra target gene
to coimmunoprecipitate with Atac2, CG10238, and NC2 b, sub- (Jasper et al., 2001). Transcript levels of chickadee decreased
units of ATAC (Figure 4A). We also sought to confirm the interac- significantly upon knockdown of Atac2 and somewhat more
tion of MSN (Figure 1A), the Drosophila Ste-20 kinase, with ATAC modestly upon knockdown of NC2 b or Jra (Figure 4C).

730 Cell 142, 726–736, September 3, 2010 ª2010 Elsevier Inc.


Transcript levels of chickadee were not reduced significantly in knockdown of Atac2 or CG10238. We next investigated
CG10238 knockdown cells. Of note, we did not observe changes whether ATAC occupancy affected acetylation of the nucleo-
in the transcript levels of target genes of a different H4K16 some on the Jra gene by a ChIP assay for H4K16 acetylation.
acetyltransferase complex (MSL) upon knockdown of ATAC, In control cells without osmotic stress, both the enhancer and
indicating that requirement for ATAC is specific for Jra and promoter regions were enriched in H4K16 acetylation, which
chickadee (Figure S5B). was significantly reduced in the Atac2 and CG10238 knock-
The JNK cascade is activated by osmotic stress and increases down cells (Figure 5D). Thus, in the absence of osmotic
c-Jun expression in mammals. Thus, we examined Jra and stress, ATAC occupied the enhancer and promoter of Jra,
chickadee transcript levels in ATAC knockdown cells upon and H4K16 acetylation at these sites was dependent on
treatment with sorbitol. Interestingly, Jra transcripts were not ATAC.
reduced in Atac2 knockdown cells upon osmotic stress but In the presence of osmotic stress, ATAC occupancy and
were increased in CG10238 knockdown cells. The expression H4K16 acetylation of the promoter were similar but were
of Chickadee was also increased in ATAC and MSN knockdown reduced at the upstream enhancer (Figures 5C and 5E). Interest-
cells under osmotic stress. Thus, ATAC positively impacts the ingly, ATAC occupancy and H4K16 acetylation of the coding
expression of JNK target genes in the absence of osmotic stress region at primer D were stimulated by osmotic stress (Figures
but plays a predominantly negative role at JNK target genes 5C and 5E). Jra was clearly localized on this region with or
under conditions of osmotic stress. Moreover, the positive without osmotic stress (Figure 4D). Jra occupancy at primer D
effects of ATAC on expression of JNK target genes are was strongly reduced in Atac2, CG10238, and MSN knockdown
mediated primarily by Atac2, whereas its negative effects are cells in the absence of sorbitol. However, under osmotic stress,
most strongly dependent on CG10238. Jra occupancy required primarily CG10238 (Figure 4D). Thus,
under conditions of osmotic stress, ATAC occupancy at the
ATAC Functions as a Cofactor for the JNK Target Gene upstream enhancer was reduced and instead increased in the
Jra, and Coordinates Upstream MAPKs onto Jra coding region where Jra was bound.
Previous studies have shown the direct association of MAPKs We next tested whether stress-activated upstream kinases
with gene loci that they regulate (de Nadal and Posas, 2010; were recruited to the Jra gene. We probed Active JNK (phos-
Dioum et al., 2009). Thus, we measured the occupancy of pho-JNK) and phospho-MKK4/SEK1 (Ser257/Thr261) occu-
ATAC and upstream kinases directly on JNK target genes. We pancy on the Jra gene. We also tested for the occupancy of
examined the occupancy of ATAC on the Jra gene in ATAC the MSN kinase. In the absence of osmotic stress in control cells,
and MSN knockdown cells by chromatin immunoprecipitation each of these kinases could be found on the Jra gene although
(ChIP) assay. Previous studies have shown that Jun can form a their locations varied. Active JNK was found at the enhancer
homodimer or heterodimerize with ATF or Fos to form the AP-1 (primer A), the promoter (primer B), and downstream of the
transcription factor. The c-Jun promoter contains AP-1-like coding region (primer E) (Figure 5F). Phospho-MKK4 was local-
sequences (TTACCTCA and TGACATCA) in addition to an ized to the enhancer, promoter, and downstream of the coding
NF-jun binding sequence (GGAGTCTCC) (Rozek and Pfeifer, region (primer E) and MSN was localized to the promoter (primer
1993). c-Jun was found to occupy the c-Jun promoter without B) (Figures 5H and 5J). Strikingly, like ATAC upon osmotic stress,
external stimulus (Angel and Karin, 1991). We confirmed that occupancy of each of these kinases was increased in the coding
Jra actually bound to the Jra gene by ChIP assay (Figure 4D, region (primer D), the site of Jra binding (Figures 5G, 5I, and 5K).
left). Jra was found to significantly occupy a site in the middle This was most significant for Active JNK, which bound strongly
of the coding region that contains an NF-jun recognition to the primer D region upon osmotic stress (note the difference
sequence (Figure 4D, left). We analyzed regions of the Jra in scale between Figures 5F and 5G), and for MSN, which relo-
gene that are comparable to the regulatory regions of the human calized from the promoter to the coding region (primer D)
c-Jun gene (Figure 5A). We probed ATAC occupancy, with and (Figures 5J and 5K).
without osmotic stress, at an upstream enhancer (primer A), The ChIP results from the knockdown cell lines indicate a
the promoter containing the AP-1 binding sequence (primer B), dynamic interplay between ATAC and the upstream kinases.
the 50 region of the coding sequence (primer C), the middle For example, in the absence of osmotic stress, H4K16 acetyla-
of the coding sequence that contained an NF-jun binding tion at the promoter (primer B) was independent of MSN (Fig-
sequence (GGAGGCACC) (primer D), and a region 30 of the ure 5D); however, upon osmotic stress, MSN suppressed
coding sequence (primer E) (FlyBase and UCSC Genome H4K16 acetylation at the promoter but was required for
Browser) (Figure 5A) (Rozek and Pfeifer, 1993). maximum acetylation in the coding region (primer D) (Figure 5E).
In the absence of osmotic stress, Atac2 was found to In the absence of osmotic stress, the binding of Active JNK to the
occupy the enhancer and promoter regions of Jra (Figure 5B, enhancer, promoter, and downstream of the coding region
dsRNA-Cont). As expected, Atac2 occupancy of these regions required ATAC but was suppressed by MSN (Figure 5F). By
was reduced in the Atac2 knockdown cells (Figure 5B). Atac2 contrast, under osmotic stress, the strong binding of Active
occupancy of the promoter was reduced in CG10238 but not JNK to the coding region (primer D) required both ATAC and
MSN knockdown cells. As controls for the specificity of ATAC MSN (Figure 5G). In the absence of osmotic stress, binding of
occupancy of Jra, we performed an ATAC-ChIP assay on two phospho-MKK4 to the enhancer was suppressed by ATAC and
target genes of the MSL complex (Figures S5 and S6). Back- MSN (Figure 5H) but upon osmotic stress, binding to the
ground signals on these genes were not affected by the promoter required MSN and CG10238 specifically (Figure 5I).

Cell 142, 726–736, September 3, 2010 ª2010 Elsevier Inc. 731


A Figure 5. Occupancy of ATAC, H4K16
Jra gene span Jra CDS Acetylation, MAPKs, and H3S10P on the
Enhancer Promoter Coding Coding E
Jra Gene with and without Osmotic Stress
Probes 100bp
A B C D (A) Schematic representation of the Jra gene
Enhancer
B ChIP-Atac2 on Jra Sorbitol (-) C ChIP-Atac2 on Jra Sorbitol (+) AP-1
NF-jun
indicating the position of different probes tested
dsRNA-Contt dsRNA-Atac2
dsRNA-At
A ac2 dsRNA-CG10238 dsRNA-MSN dsRNA-Cont dsRNA-Atac2
dsRNA-At
A ac2 dsRNA-CG10238 dsRNA-MSN
(A–E on x axis for B–M). The gray box indicates
1.00% 1.00%

0.80% 0.80%
the coding sequence (CDS), the gray circles indi-

ChIP/Input
cate the enhancer, the white circle indicates pre-
ChIP/Input

0.60% 0.60%

0.40% 0.40%
dicted AP-1 sites, and the black square indicates
0.20%
0.20%

0.00%
the NF-jun-like sequence.
0.00%
A B C D E A B C D E
(B–M) ChIP assays were performed with anti-
D ChIP-H4K16ac on Jra Soro
Sorobitol
r bitol (-) E ChIP-H4K16ac on Jra Sorbitol (+) bodies against Atac2, H4K16ac, Active JNK,
dsRNA-Cont dsRNA-Atac2
dsRNA-At
A ac2 dsRNA-CG10238 dsRNA-MSN

0.60%
dsRNA-Cont

6.00%
dsRNA-Atac2
dsRNA-At
A ac2 dsRNA-CG10238 dsRNA-MSN
phospho-MKK4 (S257T261) (pMKK4), MSN, and
0.50% 5.00% phospho-H3S10 (H3S10P) from ATAC and MSN
ChIP/Input

0.40%
knockdown S2 cells (as in Figure 4D) without
ChIP/Input 4.00%

0.30%
3.00%

0.20%
2.00% sorbitol stimulation (B, D, F, H, J, and L) (Figures
0.10%

0.00%
1.00%
6B, 6D, 6F, 6H, and 6J) or with sorbitol stimulation
0.00%
A B C D E
A B C D E (C, E, G, I, K, M) (Figures 6C, 6E, 6G, 6I, and 6K)
F ChIP-ActiveJNK
ChIP-Active
v JNK on Jra Sorbitol (-) G ChIP-ActiveJNK
ChIP-Active
v JNK on Jra Sorbitol (+)
on the probed region of the Jra gene as indicated
dsRNA-Cont dsRNA-Atac2
dsRNA-At
A ac2 dsRNA-CG10238 dsRNA-MSN dsRNA-Cont dsRNA-Atac2
dsRNA-At
A ac2 dsRNA-CG10238 dsRNA-MSN
in (A), and input chromatin was measured by
1.20% 70.00%

1.00% 60.00%
qRT-PCR and normalized to input. The ratios of
50.00%
measured quantities are represented (see Figures
ChIP/Input

ChIP/Input

0.80%
40.00%
0.60%

0.40%
30.00%
S5A and S6). The average of three independent
20.00%
0.20% 10.00% experiments is graphed. Error bars represent
0.00% 0.00%
A B C D E A B C D E standard deviation.
See also Figures S2, S5, and S6.
H dsRNA-Contt
ChIP-pMKK4 on Jra Sorbitol (-)
dsRNA-Atac2
dsRNA-At
A ac2 dsRNA-CG10238 dsRNA-MSN
I dsRNA-Cont
ChIP-pMKK4 on Jra Sorbitol (+)
dsRNA-Atac2
dsRNA-At
A ac2 dsRNA-CG10238 dsRNA-MSN
0.30% 0.45%
0.40%
0.25%
0.35%
ChIP/Input

0.20% 0.30%
ChIP/Input

0.25%
0.15%
0.20%

0.10% 0.15%
0.10%
0.05% 0.05%
0.00%
the promoter and strongly suppressed
0.00%

A B C D E
A B C D E
downstream of the coding region
J dsRNA-Contt
ChIP-Misshapen on Jra Sorbitol (-)
dsRNA-Atac2
dsRNA-At
A ac2 dsRNA-CG10238 dsRNA-MSN
K dsRNA-Cont
ChIP-Missahpen on Jar Sorbitol (+)
dsRNA-Atac2
dsRNA-At
A ac2 dsRNA-CG10238 dsRNA-MSN
(Figure 5L). Osmotic stress substantially
0.35% 0.30% increased H3S10 phosphorylation levels
0.30%
0.25%
0.25%
across the Jra gene except for the
ChIP/Input

ChIP/Input

0.20%
0.20%

0.15%

0.10%
0.15%

0.10%
upstream enhancer (note the different
0.05%

0.00%
0.05%
scales in Figures 5L and 5M). Induction
0.00%
A B C D E
A B C D E
of H3S10 phosphorylation at the
L ChIP-H3S10P on Jra Sorbitol (-) M ChIP-H3S10P on Jra Sorbitol (+) promoter, the coding region (primer D),
dsRNA-Contt dsRNA-Atac2
dsRNA-At
A ac2 dsRNA-CG10238 dsRNA-MSN dsRNA-Cont dsRNA-Atac2
dsRNA-At
A ac2 dsRNA-CG10238 dsRNA-MSN

2.50% 20.00%
18.00%
and downstream of the coding region de-
pended on ATAC (especially CG10238);
2.00% 16.00%
14.00%
ChIP/Input

ChIP/Input

1.50% 12.00%

1.00%
10.00%
8.00% however, MSN suppressed H3S10 phos-
6.00%
0.50%

0.00%
4.00%
2.00% phorylation across the gene (Figure 5M).
0.00%
0 00%

A B C D E A B C D E
These results suggest that the relation-
ship between H3S10 phosphorylation
and H4K16 acetylation by ATAC at Jra
Finally, binding of MSN to the promoter is suppressed by ATAC differs from what was observed with H3S10 phosphorylation
in the absence of osmotic stress (Figure 5J); however, when it and H4K16 acetylation by MOF at the FOSL1 gene (Zippo
moves to the coding region where Jra is bound (primer D), et al., 2009). Acetylation of H4K16 at the enhancer of FOSL1
upon osmotic stress its binding is independent of ATAC by MOF required H3S10 phosphorylation; however, ATAC is
(Figure 5K). required for H3S10 phosphorylation in the presence of osmotic
A previous study linked the acetylation of H4K16 with stress. Moreover, ATAC-dependent H4K16 acetylation at the
H3S10 phosphorylation in the activation of the FOSL1 gene Jra enhancer in the absence of osmotic stress (Figure 5D)
(Zippo et al., 2009). At this gene, H3S10 phosphorylation was occurred independently of significant H3S10 phosphorylation
reported to recruit MOF-dependent H4K16 acetylation, which (Figure 5L).
then recruited the elongation factor P-TEFb to release paused
polymerase (Zippo et al., 2009). As ATAC is also an H4K16 ace- ATAC Acetylates Nucleosomes and Coordinates
tyltransferase, we tested for a relationship between H3S10 the JNK Upstream Kinases with Jra on chickadee
phosphorylation and H4K16 acetylation, ATAC, or MSN at Jra. The most striking aspect of our ChIP analysis of the Jra gene is the
H3S10P was enriched in the enhancer and promoter regions movement upon osmotic stress of ATAC and MAPKs to the
(primer B) in the absence of osmotic stress (Figure 5L). H3S10 coding region (primer D), which contains an NF-jun binding
phosphorylation levels were suppressed by ATAC and MSN at sequence and is occupied by the Jra transcription factor. This

732 Cell 142, 726–736, September 3, 2010 ª2010 Elsevier Inc.


A Figure 6. Occupancy of ATAC, H4K16
Chickadee gene span Acetylation, MAPKs, and H3S10P on the
chic CDS chickadee Gene with and without Osmotic
Probes ChicA ChicB ChicC Stress
1K bp
Enhancer (A) Schematic representation of the chickadee
B ChIP-Atac2 on Chickadee Sorbitol (-) C ChIP-Atac2 on Chickadee Sorbitol (+) AP-1 gene including 7 kb upstream of the transcription
dsRNA-Cont dsRNA-Atac2 dsRNA-Cont dsRNA-Atac2
dsRNA-CG10238
dsRNA-
A CG10238
dsRNA-Jra
dsRNA-
A Jra
dsRNA-MSN
dsRNA-
A MSN dsRNA-CG10238
dsRNA-
A CG10238
dsRNA-
A Jra
dsRNA-Jra
dsRNA-MSN
dsRNA-
A MSN
start site indicating the position of different probes
0.14%
4% 1.00%
0.90%
on the gene (ChicA-C on x axis for B–K). The gray
0.12%
2%

0.10%
0%
0.80%
0.70%
box indicates the coding sequence (CDS), the gray
ChIP/Input

ChIP/Input
0.08%
8% 0.60%

0.06%
6%
0.50% circles indicate the enhancer, and the white circles
0.40%
4%
0.04% 0.30%
0.20%
indicate predicted AP-1 sites.
2%
0.02%

0.00%
0%
0.10%
0.00%
(B–K) ChIP assays were performed with antibodies
ChicA ChicB ChicC ChicA ChicB ChicC
against Atac2, H4K16ac, pMKK4 (S257T261),
D ChIP-H4K16ac on Chickadee Sorbitol (-) E ChIP-H4K16ac on Chickadee Sorbitol (+) MSN, and phospho-H3S10 (H3S10P) from ATAC
dsRNA-Cont
dsRNA-
A Cont
dsRNA-
dsRNA-CG10238
A CG10238
dsRNA-Atac2
dsRNA-
A At
dsRNA-
A ac2
dsRNA-MSN
A MSN
dsRNA-Cont
dsRNA-
A Cont
dsRNA-
dsRNA-CG10238
A CG10238
dsRNA-Atac2
dsRNA-
A At
dsRNA-
A ac2
dsRNA-MSN
A MSN and MSN knockdown S2 cells with or without
dsRNA-
A Jra
dsRNA-Jra dsRNA-Jra
dsRNA-
A Jra
.90%
0.90% 0.90% sorbitol stimulation (as in Figure 4D) on the probed
.80%
0.80% 0.80%
.70%
0.70% 0.70% region of the chickadee gene and upstream region
ChIP/Input

0.60%
.60% 0.60%
ChIP/Input

.50%
0.50% 0.50% as indicated in (A), and input chromatin was
0.40%
.40% 0.40%

.30%
0.30% 0.30%
0.20%
measured by qRT-PCR and normalized to input
.20%
0.20%
.10%
0.10%
0.00%
.00%
0.10%
0.00%
(see Figure S7A). The ratios of the measured
ChicA ChicB ChicC ChicA ChicB ChicC quantities are represented. The average of three
F ChIP-pMKK4 on Chicadee Sorbitol (-) G ChIP-pMKK4 on Chickadee Sorbitol (+)
independent experiments is graphed. Error bars
dsRNA-Cont
dsRNA-
A Cont
dsRNA-
dsRNA-CG10238
A CG10238
dsRNA-Atac2
dsRNA-
A At
dsRNA-
A ac2
dsRNA-MSN
A MSN
dsRNA-Cont
dsRNA-
A Cont
dsRNA-
dsRNA-CG10238
A CG10238
dsRNA-Atac2
dsRNA-
A At
dsRNA-
A ac2
dsRNA-MSN
A MSN represent standard deviation.
dsRNA-Jra dsRNA-
A Jra
dsRNA-Jra
0.60%
.60% 1.20% See also Figures S2, S5, S6, and S7.
0.50%
.50% 1.00%

0.80%
ChIP/Input

0.40%
.40%
ChIP/Input

0.30%
.30% 0.60%

0.20%
.20% 0.40%

0.10%
.10% 0.20%

0.00%
0.00%
.00%
ChicA ChicB ChicC ChicA ChicB ChicC MSN knockdown cells but not in Atac2
H ChIP-Misshapen on Chickadee Sorbitol (-) I ChIP-Misshapen on Chickadee Sorbitol (+)
knockdown cells (Figure S7A). These
dsRNA-Cont
dsRNA-
A Cont
dsRNA-CG10238
dsRNA-
A CG10238
dsRNA-
A Jra
dsRNA-Jra
dsRNA-
dsRNA-Atac2
A At
A ac2
dsRNA-MSN
dsRNA-
A MSN
dsRNA-
dsRNA-Cont
A Cont
dsRNA-CG10238
dsRNA-
A CG10238
dsRNA-Jra
dsRNA-
A Jra
dsRNA-
dsRNA-Atac2
A At
A ac2
dsRNA-MSN
dsRNA-
A MSN two regions were co-occupied by ATAC
0.30% 0.25%
and H4K16 acetylation (Figures 6B and
0.25% 0.20%
6D). The presence of Atac2 and H4K16
ChIP/Input

0.20%
ChIP/Input

0.15%
0.15%

0.10%
0.10% acetylation at these regions was not
0.05%
0.05%
affected by the knockdown of Jra, sug-
0.00% 0.00%

ChicA ChicB ChicC ChicA ChicB ChicC gesting ATAC does not require Jra to
J K bind the chickadee gene in the absence
ChIP-H3S10P on Chickadee Sorbitol (-) ChIP-H3S10P on Chickadee Sorbitol (+)

dsRNA-Cont
dsRNA-
A Cont
dsRNA-
dsRNA-CG10238
A CG10238
dsRNA-
dsRNA-Atac2
A At
dsRNA-
A ac2
dsRNA-MSN
A MSN
dsRNA-Cont
dsRNA-
A Cont
dsRNA-
dsRNA-CG10238
A CG10238
dsRNA-Atac2
dsRNA-
A At
dsRNA-
A ac2
dsRNA-MSN
A MSN
of osmotic stress. Interestingly, in the
dsRNA-
A Jra
dsRNA-Jra dsRNA-Jra
dsRNA-
A Jra
2.00% 18.00%
presence of osmotic stress, ATAC occu-
1.80% 16.00%
1.60%
1.40%
14.00% pancy and H4K16 acetylation were
ChIP/Input

ChIP/Input

12.00%
1.20%
1.00%
0.80%
10.00%
8.00%
increased at the ChicA region, and this
6.00%
0.60%
0.40% 4.00%
2.00%
increased occupancy was reduced in
0.20%
0.00%
0 00%

ChicA ChicB ChicC


0.00%
0 00%

ChicA ChicB ChicC


Jra knockdown cells (Figures 6C and 6E).
Phospho-MKK4 and MSN also occupied
the ChicA and ChicB regions, and these
raised the question as to whether ATAC and MAPKs move to kinases were also found in the open reading frame (ChicC) in
this location because Jra is bound there or because they need the absence of osmotic stress (Figures 6F and 6H). In contrast
to associate with the coding region. Thus, we performed a ChIP to what was seen at the Jra gene, on chickadee the kinases
assay on another JNK target gene, chickadee. Chickadee does move to the upstream ChicA region upon osmotic stress and their
not have an apparent NF-jun binding site in the coding region. It occupancy is largely (MKK4) or slightly (MSN) dependent on Jra
does, however, have several AP-1-like sequences upstream of (Figures 6G and 6I). Occupancy of ChicA by the kinases under
the promoter and at a far-upstream enhancer (Figure 6A). We these conditions is slightly suppressed by Atac2 but completely
analyzed 7 kb upstream of the chickadee gene and found five dependent on CG10238. Indeed, CG10238 was crucial for the
AP-1-like sequences. We probed a region containing an AP-1- occupancy of ATAC, MKK4, and MSN on the far-upstream
like site, which was also near two CCAAT motifs that are 6.4 kb ChicA region under conditions of osmotic stress (Figures 6C,
upstream from the coding sequence (ChicA primer). We also 6G, and 6I). At the Jra gene, occupancy of the primer D region
probed adjacent to the AP-1-like site 2 kb upstream from the containing the NF-Jun binding site by ATAC, Active JNK, and
coding sequence close to the predicted promoter (ChicB primer) phospho-MKK4 upon osmotic stress was dependent on
and the middle of the coding region (ChicC primer) (Figure 6A). CG10238. As seen on Jra, H3S10P on chickadee was sup-
ChIP analysis revealed that Jra was localized at the ChicA and pressed by ATAC and MSN in the absence of osmotic stress
ChicB regions, suggesting Jra binds AP-1 sites on chickadee and, although H3S10P increased upon osmotic stress, these
(Figure S7A). Jra occupancy was reduced in CG10238 and levels were suppressed by MSN specifically (Figures 6J and 6K).

Cell 142, 726–736, September 3, 2010 ª2010 Elsevier Inc. 733


A The models presented in Figure 7 summarize the molecular
ATAC events occurring on Jra and chickadee as revealed by the ChIP
CG10238
JNK data. Although differing in details having to do with the architec-
JNK
Atac2 Jra Jra-P MKK4 ture of each gene, comparison of the expression and ChIP data
Jra K16ac
S10P
Osmotic stress from Jra and chickadee reveals strikingly common features
E AP-1 NF-jun TATA-like
about the role of ATAC at these genes. First, in the absence of
MSN
MKK4
MSN osmotic stress, ATAC is required for the basal expression of
these genes, which is most highly dependent on the Atac2 ace-
B MKK4-P
tyltransferase subunit. Second, upon osmotic stress, ATAC
suppresses induced transcription levels of these genes, which
CG10238 JNK-P
is most highly dependent on the CG10238 subunit. Third, upon
ATAC
Atac2 osmotic stress, ATAC and the MAPKs move to the region of
Jra-P
Jra
K16ac K16ac the gene where Jra is bound. And fourth, the relocalization of
S10P
E AP-1 NF-jun TATA-like ATAC and MAPKs is dependent on CG10238, the MAP kinase
C MSN upstream kinase binding inhibitory protein. Thus, ATAC coordi-
P-1 nates the occupancy of Jra and upstream kinases while it
E E A 16ac
K controls the levels of JNK activation and target gene induction.
Jra

ATAC MSN
CG10238
MKK4
DISCUSSION
Osmotic stress
JNK
Jra
K16ac
ATAC shares four subunits with the SAGA transcription coactiva-
MSN
S10P
Chickadee AP-1 tor complex and was anticipated to function as a coactivator.
MKK4-P However, preliminary studies indicated that ATAC did not
AP-1c interact with the activation domains of VP16 or p53 under
E E K160aP JNK-P
D S10P S10P S1
conditions in which SAGA bound (Kusch et al., 2003). Prompted
Jra-P
by the identification of peptides from the Jra (Drosophila c-Jun)
ATAC MSN
CG10238
MKK4
transcription factor in ATAC affinity purifications, we demon-
strated that Jra coimmunoprecipitates with ATAC subunits. We
Jra JNK
K16ac further demonstrate that ATAC is localized to Jra target genes
S10P
Chickadee AP-1 (Jra and chickadee) and is required for H4K16 acetylation at
the Jra and chickadee enhancer and promoter. ATAC was
required for basal levels of expression of these genes, which
Figure 7. Models Summarizing ATAC Functions and the Molecular depended strongly on the Atac2 acetyltransferase subunit.
Events Occurring on Jra and chickadee as Revealed by the ChIP Thus, to our knowledge, this is the first report to demonstrate
Data
that ATAC functions as a transcription cofactor.
Summaries of ChIP data on the Jra gene without and with osmotic stress are
shown in (A) and (B), respectively. Summaries of ChIP data on the chickadee
Our pursuit of the functions of the CG10238 subunit of ATAC
gene without and with osmotic stress are shown in (C) and (D), respectively. and the potential role of its MoaE domain led us to discover
(A) In the absence of osmotic stress, ATAC occupies and acetylates H4K16 that ATAC is intimately integrated into MAP kinase signaling
(K16ac) at the enhancer (E) and promoter regions, containing AP-1 sites and target gene expression. CG10238 was found to inhibit
(AP-1), of the Jra gene and is required for basal levels of Jra transcription (small JNK activation by osmotic stress, an activity mediated through
arrow). ATAC blocks the recruitment of MKK4 to the enhancer and MSN to the
the MoaE domain. Thus, structural features of this protein that
promoter while inhibiting the activation of JNK. The phosphorylation of H3S10
(S10P) on the promoter region is suppressed by MSN.
are utilized for molybdopterin synthesis have been conscripted
(B) When cells are exposed to osmotic stress, the stress-activated kinases in in ATAC to function in the regulation of MAP kinase signaling.
the JNK cascade are recruited to the Jra binding motif (NF-jun) on the Jra gene The MSN kinase is the most likely direct target of CG10238, as
by ATAC, especially dependent on CG10238. In addition to interacting with the it coimmunoprecipitates with ATAC subunits. However, we
promoter region, ATAC also interacts with the NF-jun site by interaction with cannot rule out potential interactions with other kinases; for
Jra and further acetylates H4K16 at this site. ATAC continues to limit the extent example, peptides from MKK4 have been found in ATAC purifi-
of JNK activation. Phosphorylation of H3S10 is increased across the Jra gene
cations (Figure 1). Under conditions of osmotic stress, ATAC
by osmotic stress but is still suppressed by MSN. Jra transcription is rapidly
and transiently induced (large arrow). suppresses the level of target gene expression in a manner
(C) The chickadee gene does not have an apparent NF-jun site. In the absence strongly dependent on CG10238. We believe this is due to inhi-
of osmotic stress, Jra, ATAC, and acetyl-H4K16 are found at two AP-1 sites, bition of JNK activation rather than a direct negative effect of
which are 2 and 7 kb upstream from the coding sequence. Upstream kinases ATAC on transcription. In fact, ATAC may still function as a posi-
occupy these sites and the downstream coding region. tive cofactor for induced Jra expression, as the levels of H3S10P
(D) In the presence of osmotic stress, ATAC preferentially relocalizes to the
on the gene, considered a positive mark for transcription
far-upstream AP-1 site and recruits Jra to this location. Phospho-MKK4
(MKK4-P) and MSN are also relocalized to this far-upstream AP-1 site in
a CG10238- and Jra-dependent manner. The CG10238 subunit of ATAC is the levels of phosphorylated H3S10 are suppressed by MSN across the
required for ATAC, Jra, MKK4-P, and MSN relocalization to the far-upstream chickadee gene.
enhancer. Transcription of chickadee is activated (large arrow) whereas See also Figure S7.

734 Cell 142, 726–736, September 3, 2010 ª2010 Elsevier Inc.


(Zippo et al., 2009), are dependent on ATAC (Figure 5M). In Multidimensional Protein Identification Technology Analysis
a manner strongly dependent on CG10238, ATAC plays a striking MudPIT analysis was performed as described in the Extended Experimental
Procedures.
role in the binding of MAPKs to the Jra and chickadee genes.
Upon osmotic stress, ATAC was required for the recruitment of
SUPPLEMENTAL INFORMATION
the JNK, MKK4, and MSN kinases to the site of Jra binding.
The only exception was the binding of MSN to the Jra gene Supplemental Information includes Extended Experimental Procedures and
(Figure 5K). However, MSN required CG10238 for binding chick- seven figures and can be found with this article online at doi:10.1016/j.cell.
adee under inducing conditions (Figure 6I). The crucial role of 2010.07.045.
CG10238 in both the recruitment of the kinases and the inhibition
of signaling is most consistent with a model by which ATAC ACKNOWLEDGMENTS

inhibits JNK activation at the downstream target genes. The


We thank the Workman Lab members, Bjoern Gaertner in the Zeitlinger Lab,
fact that ATAC and the MAPKs all move to the site of Jra binding and Hidehisa Takahashi in the Conaway Lab for advice and support during
suggests that a localized signaling network is organized at the this project. This research was supported by the Stowers Institute for Medical
site occupied by the downstream transcription factor. Research.
Our data indicate that a positive cofactor of JNK target gene
transcription, ATAC, also serves to negatively feed back for inhi- Received: December 30, 2009
Revised: May 14, 2010
bition of the JNK signaling pathway. Why would a downstream
Accepted: July 1, 2010
activator also repress upstream signaling? The JNK pathway Published: September 2, 2010
and c-Jun have been shown to suppress p53-dependent
apoptosis and cellular senescence (Alexaki et al., 2008; Butter- REFERENCES
field et al., 1997; Das et al., 2007; Fan et al., 2010; Ham et al.,
2000; Takekawa and Saito, 1998; Yang et al., 1997). Moreover, Alepuz, P.M., Jovanovic, A., Reiser, V., and Ammerer, G. (2001). Stress-
induced MAP kinase Hog1 is part of transcription activation complexes. Mol.
c-Jun has been shown to repress transcription of the p53 gene
Cell 7, 767–777.
and hence its downstream targets (Schreiber et al., 1999).
Alepuz, P.M., de Nadal, E., Zapater, M., Ammerer, G., and Posas, F. (2003).
Thus, a delicate balance of JNK signaling and p53 expression
Osmostress-induced transcription by Hot1 depends on a Hog1-mediated
controls cell growth, proliferation, and apoptosis. If ATAC recruitment of the RNA Pol II. EMBO J. 22, 2433–2442.
suppression of JNK is involved in this process, we might expect Alexaki, V.I., Javelaud, D., and Mauviel, A. (2008). JNK supports survival in
loss of ATAC to result in decreased expression of p53 due to melanoma cells by controlling cell cycle arrest and apoptosis. Pigment Cell
increased JNK activity (Schreiber et al., 1999). Indeed, whereas Melanoma Res 21, 429–438.
knockdown of Jra increased p53 expression in S2 cells, knock- Altschul, S.F., Madden, T.L., Schaffer, A.A., Zhang, J., Zhang, Z., Miller, W.,
down of ATAC subunits resulted in a decrease in p53 expression and Lipman, D.J. (1997). Gapped BLAST and PSI-BLAST: a new generation
consistent with ATAC suppression of JNK activation (Figure S7B). of protein database search programs. Nucleic Acids Res. 25, 3389–3402.
Our results reveal a mechanism for governing the transcrip- Angel, P., and Karin, M. (1991). The role of Jun, Fos and the AP-1 complex in
tional response to signaling pathways. By serving as both a cell-proliferation and transformation. Biochim. Biophys. Acta 1072, 129–157.

downstream transcriptional cofactor and as an inhibitor of Angel, P., Hattori, K., Smeal, T., and Karin, M. (1988). The jun proto-oncogene
is positively autoregulated by its product, Jun/AP-1. Cell 55, 875–885.
upstream signaling, ATAC serves as a master regulator which
Butterfield, L., Storey, B., Maas, L., and Heasley, L.E. (1997). c-Jun
administers the appropriate level of response to the inducing
NH2-terminal kinase regulation of the apoptotic response of small cell lung
signals.
cancer cells to ultraviolet radiation. J. Biol. Chem. 272, 10110–10116.
Ciurciu, A., Komonyi, O., Pankotai, T., and Boros, I.M. (2006). The Drosophila
EXPERIMENTAL PROCEDURES histone acetyltransferase Gcn5 and transcriptional adaptor Ada2a are
involved in nucleosomal histone H4 acetylation. Mol. Cell. Biol. 26, 9413–9423.
Cell Lines, Extract Preparation, and Complex Purification Das, M., Jiang, F., Sluss, H.K., Zhang, C., Shokat, K.M., Flavell, R.A., and
The nuclear extracts from S2 cell lines were generated as described in
Davis, R.J. (2007). Suppression of p53-dependent senescence by the JNK
Extended Experimental Procedures and were used for FLAG affinity purifica-
signal transduction pathway. Proc. Natl. Acad. Sci. USA 104, 15759–15764.
tion as previously described (Suganuma et al., 2008).
de Nadal, E., and Posas, F. (2010). Multilayered control of gene expression by
stress-activated protein kinases. EMBO J. 29, 4–13.
Chromatin Immunoprecipitation Assay
de Nadal, E., Alepuz, P.M., and Posas, F. (2002). Dealing with osmostress
The crosslinked S2 cell pellets were washed, resuspended, and sonicated,
through MAP kinase activation. EMBO Rep. 3, 735–740.
and the DNA was immunoprecipitated with antibodies using Dynabeads
(Invitrogen). The bound DNA and input DNA were incubated with RNaseA de Nadal, E., Zapater, M., Alepuz, P.M., Sumoy, L., Mas, G., and Posas, F.
and the crosslinking was reversed. The ethanol-precipitated DNA pellets (2004). The MAPK Hog1 recruits Rpd3 histone deacetylase to activate
were resuspended and then were analyzed by quantitative real-time RT-PCR. osmoresponsive genes. Nature 427, 370–374.
Antibodies, buffer content, and primers for ChIP-qRT-PCR are described in Devary, Y., Gottlieb, R.A., Lau, L.F., and Karin, M. (1991). Rapid and
the Extended Experimental Procedures. preferential activation of the c-jun gene during the mammalian UV response.
Mol. Cell. Biol. 11, 2804–2811.
Osmotic Stress Response in dsRNAi Knockdown S2 Cells Dioum, E.M., Wauson, E.M., and Cobb, M.H. (2009). MAP-ping unconven-
and siRNA Knockdown 293T Cells tional protein-DNA interactions. Cell 139, 462–463.
The dsRNAi was transfected in S2 cells. The siRNA was transfected in 293T Edmunds, J.W., and Mahadevan, L.C. (2004). MAP kinases as structural
cells as described in the Extended Experimental Procedures. The cells were adaptors and enzymatic activators in transcription complexes. J. Cell Sci.
incubated with 500 mM sorbitol for 12 min before harvest. 117, 3715–3723.

Cell 142, 726–736, September 3, 2010 ª2010 Elsevier Inc. 735


Fan, Y., Lee, T.V., Xu, D., Chen, Z., Lamblin, A.F., Steller, H., and Bergmann, A. Pokholok, D.K., Zeitlinger, J., Hannett, N.M., Reynolds, D.B., and Young, R.A.
(2010). Dual roles of Drosophila p53 in cell death and cell differentiation. Cell (2006). Activated signal transduction kinases frequently occupy target genes.
Death Differ. 17, 912–921. Science 313, 533–536.
Fukuyama, K., Yoshida, M., Yamashita, A., Deyama, T., Baba, M., Suzuki, A., Proft, M., Mas, G., de Nadal, E., Vendrell, A., Noriega, N., Struhl, K., and Posas,
Mohri, H., Ikezawa, Z., Nakajima, H., Hirai, S., et al. (2000). MAPK upstream F. (2006). The stress-activated Hog1 kinase is a selective transcriptional elon-
kinase (MUK)-binding inhibitory protein, a negative regulator of MUK/dual gation factor for genes responding to osmotic stress. Mol. Cell 23, 241–250.
leucine zipper-bearing kinase/leucine zipper protein kinase. J. Biol. Chem. Rozek, D., and Pfeifer, G.P. (1993). In vivo protein-DNA interactions at the c-jun
275, 21247–21254. promoter: preformed complexes mediate the UV response. Mol. Cell. Biol. 13,
Guelman, S., Suganuma, T., Florens, L., Swanson, S.K., Kiesecker, C.L., 5490–5499.
Kusch, T., Anderson, S., Yates, J.R., III, Washburn, M.P., Abmayr, S.M., Rozek, D., and Pfeifer, G.P. (1995). In vivo protein-DNA interactions at the c-jun
et al. (2006). Host cell factor and an uncharacterized SANT domain protein promoter in quiescent and serum-stimulated fibroblasts. J. Cell. Biochem. 57,
are stable components of ATAC, a novel dAda2A/dGcn5-containing histone 479–487.
acetyltransferase complex in Drosophila. Mol. Cell. Biol. 26, 871–882.
Rudolph, M.J., Wuebbens, M.M., Rajagopalan, K.V., and Schindelin, H. (2001).
Ham, J., Eilers, A., Whitfield, J., Neame, S.J., and Shah, B. (2000). c-Jun and Crystal structure of molybdopterin synthase and its evolutionary relationship to
the transcriptional control of neuronal apoptosis. Biochem. Pharmacol. 60, ubiquitin activation. Nat. Struct. Biol. 8, 42–46.
1015–1021.
Schreiber, M., Kolbus, A., Piu, F., Szabowski, A., Mohle-Steinlein, U., Tian, J.,
Hu, S., Xie, Z., Onishi, A., Yu, X., Jiang, L., Lin, J., Rho, H.S., Woodard, C., Karin, M., Angel, P., and Wagner, E.F. (1999). Control of cell cycle progression
Wang, H., Jeong, J.S., et al. (2009). Profiling the human protein-DNA interac- by c-Jun is p53 dependent. Genes Dev. 13, 607–619.
tome reveals ERK2 as a transcriptional repressor of interferon signaling. Cell
Schwarz, G., and Mendel, R.R. (2006). Molybdenum cofactor biosynthesis and
139, 610–622.
molybdenum enzymes. Annu. Rev. Plant Biol. 57, 623–647.
Iyer, L.M., Burroughs, A.M., and Aravind, L. (2006). The prokaryotic anteced-
Su, Y.C., Treisman, J.E., and Skolnik, E.Y. (1998). The Drosophila Ste20-
ents of the ubiquitin-signaling system and the early evolution of ubiquitin-like
related kinase misshapen is required for embryonic dorsal closure and acts
b-grasp domains. Genome Biol. 7, R60.
through a JNK MAPK module on an evolutionarily conserved signaling
Jasper, H., Benes, V., Schwager, C., Sauer, S., Clauder-Munster, S., Ansorge, pathway. Genes Dev. 12, 2371–2380.
W., and Bohmann, D. (2001). The genomic response of the Drosophila embryo
Suganuma, T., Gutierrez, J.L., Li, B., Florens, L., Swanson, S.K., Washburn,
to JNK signaling. Dev. Cell 1, 579–586.
M.P., Abmayr, S.M., and Workman, J.L. (2008). ATAC is a double histone
Kayali, A.G., Austin, D.A., and Webster, N.J. (2000). Stimulation of MAPK acetyltransferase complex that stimulates nucleosome sliding. Nat. Struct.
cascades by insulin and osmotic shock: lack of an involvement of p38 Mol. Biol. 15, 364–372.
mitogen-activated protein kinase in glucose transport in 3T3-L1 adipocytes.
Takekawa, M., and Saito, H. (1998). A family of stress-inducible GADD45-like
Diabetes 49, 1783–1793.
proteins mediate activation of the stress-responsive MTK1/MEKK4 MAPKKK.
Kimura, A., Matsubara, K., and Horikoshi, M. (2005). A decade of histone acet- Cell 95, 521–530.
ylation: marking eukaryotic chromosomes with specific codes. J. Biochem.
Thomson, S., Mahadevan, L.C., and Clayton, A.L. (1999). MAP kinase-
138, 647–662.
mediated signalling to nucleosomes and immediate-early gene induction.
Kusch, T., Guelman, S., Abmayr, S.M., and Workman, J.L. (2003). Two Semin. Cell Dev. Biol. 10, 205–214.
Drosophila Ada2 homologues function in different multiprotein complexes.
Treisman, J.E., Ito, N., and Rubin, G.M. (1997). misshapen encodes a protein
Mol. Cell. Biol. 23, 3305–3319.
kinase involved in cell shape control in Drosophila. Gene 186, 119–125.
Kyriakis, J.M., and Avruch, J. (2001). Mammalian mitogen-activated protein Wang, Y.L., Faiola, F., Xu, M., Pan, S., and Martinez, E. (2008). Human ATAC is
kinase signal transduction pathways activated by stress and inflammation. a GCN5/PCAF-containing acetylase complex with a novel NC2-like histone
Physiol. Rev. 81, 807–869. fold module that interacts with the TATA-binding protein. J. Biol. Chem. 283,
Lawrence, M.C., Shao, C., McGlynn, K., Naziruddin, B., Levy, M.F., and Cobb, 33808–33815.
M.H. (2009). Multiple chromatin-bound protein kinases assemble factors that Yang, D.D., Kuan, C.Y., Whitmarsh, A.J., Rincon, M., Zheng, T.S., Davis, R.J.,
regulate insulin gene transcription. Proc Natl Acad Sci USA 106, 22181–22186. Rakic, P., and Flavell, R.A. (1997). Absence of excitotoxicity-induced
Lee, K.K., and Workman, J.L. (2007). Histone acetyltransferase complexes: apoptosis in the hippocampus of mice lacking the Jnk3 gene. Nature 389,
one size doesn’t fit all. Nat. Rev. Mol. Cell Biol. 8, 284–295. 865–870.
Leimkuhler, S., Freuer, A., Araujo, J.A., Rajagopalan, K.V., and Mendel, R.R. Yang, S.H., Sharrocks, A.D., and Whitmarsh, A.J. (2003). Transcriptional
(2003). Mechanistic studies of human molybdopterin synthase reaction and regulation by the MAP kinase signaling cascades. Gene 320, 3–21.
characterization of mutants identified in group B patients of molybdenum Zapater, M., Sohrmann, M., Peter, M., Posas, F., and de Nadal, E. (2007).
cofactor deficiency. J. Biol. Chem. 278, 26127–26134. Selective requirement for SAGA in Hog1-mediated gene expression depend-
Mas, G., de Nadal, E., Dechant, R., Rodriguez de la Concepcion, M.L., Logie, ing on the severity of the external osmostress conditions. Mol. Cell. Biol. 27,
C., Jimeno-Gonzalez, S., Chavez, S., Ammerer, G., and Posas, F. (2009). 3900–3910.
Recruitment of a chromatin remodelling complex by the Hog1 MAP kinase Zippo, A., Serafini, R., Rocchigiani, M., Pennacchini, S., Krepelova, A., and
to stress genes. EMBO J. 28, 326–336. Oliviero, S. (2009). Histone crosstalk between H3S10ph and H4K16ac
Morrison, D.K., Murakami, M.S., and Cleghon, V. (2000). Protein kinases and generates a histone code that mediates transcription elongation. Cell 138,
phosphatases in the Drosophila genome. J. Cell Biol. 150, F57–F62. 1122–1136.

736 Cell 142, 726–736, September 3, 2010 ª2010 Elsevier Inc.


A Bacterial mRNA Leader that Employs
Different Mechanisms to Sense
Disparate Intracellular Signals
Sun-Yang Park,1,3,4 Michael J. Cromie,1,2,3,5 Eun-Jin Lee,1,2,5 and Eduardo A. Groisman1,2,5,*
1Department of Molecular Microbiology
2Howard Hughes Medical Institute
Washington University School of Medicine, 660 South Euclid Avenue, Campus Box 8230, St. Louis, MO 63110, USA
3These authors contributed equally to this work
4Present address: Section of Microbial Pathogenesis, Yale University School of Medicine, Boyer Center for Molecular Medicine,

295 Congress Avenue, New Haven, CT 06536-0812, USA


5Present address: Section of Microbial Pathogenesis, Howard Hughes Medical Institute, Yale University School of Medicine,

Boyer Center for Molecular Medicine, 295 Congress Avenue, New Haven, CT 06536-0812, USA
*Correspondence: eduardo.groisman@yale.edu
DOI 10.1016/j.cell.2010.07.046

SUMMARY These mRNA leaders typically control the expression of the


proteins that synthesize and/or transport the metabolites or
Bacterial mRNAs often contain leader sequences ions to which they respond. Whereas leaders known as ribos-
that respond to specific metabolites or ions by witches sense metabolites or ions directly (Henkin, 2008; Winkler
altering expression of the associated downstream and Breaker, 2005), the presence of stretches of certain nucleo-
protein-coding sequences. Here we report that the tides in a leader sequence or particular codons in a leader ORF
leader RNA of the Mg2+ transporter gene mgtA of enables cells to probe the cellular levels of the corresponding
nucleotides and amino acids indirectly. Here we identify a leader
Salmonella enterica, which was previously known
mRNA that controls gene expression by responding to Mg2+
to function as a Mg2+-sensing riboswitch, harbors directly as a riboswitch and to proline levels indirectly, via trans-
an 18 codon proline-rich open reading frame— lation of a proline-codon-rich open reading frame.
termed mgtL—that permits intracellular proline to The mRNA for the Mg2+ transporter gene mgtA from Salmo-
regulate mgtA expression. Interfering with mgtL nella enterica serovar Typhimurium includes a 264 nucleotide
translation by genetic, pharmacological, or environ- long leader sequence that functions as a Mg2+-responding ribos-
mental means was observed to increase the mRNA witch determining whether transcription proceeds into the mgtA
levels from the mgtA coding region. Substitution of CR (Cromie et al., 2006). In low Mg2+, the mgtA leader RNA
the mgtL proline codons by other codons abolished adopts a conformation (i.e., stem loop C) that favors transcrip-
the response to proline and to hyperosmotic stress tion of the full-length mgtA mRNA (Cromie et al., 2006) (Figure 1)
but not to Mg2+. Our findings show that mRNA leader and production of the MgtA protein (Cromie and Groisman,
2010). MgtA-mediated Mg2+ uptake restores cytoplasmic Mg2+
sequences can consist of complex regulatory
to prestress levels, which promotes a different conformation in
elements that utilize different mechanisms to sense
the mgtA leader RNA (i.e., stem loop B) that hinders transcription
separate signals and mediate an appropriate cellular elongation into the mgtA CR (Cromie et al., 2006) (Figure 1).
response. In addition to being regulated by a Mg2+-sensing riboswitch,
mgtA transcription is controlled at the initiation step by two
INTRODUCTION DNA binding proteins: PhoP, which activates transcription
when its cognate sensor PhoQ detects low extracytoplasmic
The leader region (LR) of many bacterial mRNAs has the ability Mg2+ (Garcia Vescovi et al., 1996), acid pH (Prost et al., 2007),
to form mutually exclusive secondary structures that determine or antimicrobial peptides (Bader et al., 2005); and Rob, the over-
whether transcription will continue into the adjacent coding expression of which stimulates mgtA transcription from a site
region (CR). Which secondary structure forms is governed located downstream of the PhoP-dependent start site (Barchiesi
by the binding of specific metabolites, ions, tRNAs, or proteins et al., 2008) (Figure 1). Thus, there is PhoP/PhoQ-dependent
to the LR, by pausing of RNA polymerase at particular sequences production of the mgtA leader mRNA following bacterial growth
within the LR and by translation of short open reading frames in media with <1 mM Mg2+ (Cromie et al., 2006) or pH 5.7 (Choi
(ORFs) located in the leader RNA (Grundy and Henkin, 2006; et al., 2009); however, due to the riboswitch action, the mRNA
Henkin, 2008; Henkin and Yanofsky, 2002; Landick et al., 1996; corresponding to the mgtA CR is observed primarily upon growth
Merino and Yanofsky, 2005; Turnbough and Switzer, 2008). in media containing very low (i.e., 10 mM) Mg2+ (Choi et al., 2009;

Cell 142, 737–748, September 3, 2010 ª2010 Elsevier Inc. 737


Figure 1. Expression of the Salmonella
Mg2+ Transporter Gene mgtA Is Regulated
by the PhoP/PhoQ System, the Rob Protein,
and the mgtA LR
When the sensor PhoQ detects low extracytoplas-
mic Mg2+, acid pH, or antimicrobial peptides,
it promotes phosphorylation of the PhoP protein,
which binds to the mgtA promoter resulting in
transcription initiation. Rob promotes mgtA tran-
scription in response to a yet unidentified signal.
Transcription elongation into the mgtA CR is regu-
lated by the mgtA leader via a Mg2+-sensing
riboswitch and by translation of an 18 codon
proline-rich ORF designated mgtL. The mgtL
ribosome binding site is denoted by RBS adjacent
to a black line. Positions and sequences of stop
codon mutations in the strains used in the experi-
ments presented in Figure 4 are denoted below the
linear mgtL RNA sequence. High-Mg2+ and high-
proline conditions promote formation of stem
loop B, hindering transcription elongation into the
mgtA CR. Low Mg2+ and/or low proline favor
formation of stem loop C, resulting in transcription
of the mgtA CR. See also Figure S1.

formants recovered on X-Gal-containing


Luria-Bertani (LB) ampicillin agar plates
that were of a darker or lighter shade of
blue than those that received a plasmid
control were purified and their plasmid
DNA extracted and used to transform
strain YS809, a derivative of strain
Cromie and Groisman, 2010; Cromie et al., 2006). This raises the YS774 with the same mgtA-lac transcriptional fusion. One of
possibility of additional signals and/or genetic elements acting on the plasmids—designated pSL55—promoted higher levels of
the mgtA leader mRNA synthesized when Salmonella experi- mgtA-lac expression than the plasmid control (Figure 2A), like
ences inducing conditions for PhoP/PhoQ and/or Rob to it did in strain YS774, indicative that the observed phenotype
promote transcription elongation into the mgtA CR. was plasmid linked. Plasmid pSL55 appears to promote mgtA-
Here we demonstrate that the mgtA LR harbors an ORF rich in lac expression specifically because it had no effect on the Lac
proline codons that enables Salmonella to control transcription phenotype of strains harboring lac transcriptional fusions in the
elongation into the mgtA CR in response to changes in cytosolic CRs of the PhoP-dependent mgtC gene or the PhoP-indepen-
proline levels. We establish that high osmolarity promotes tran- dent corA gene (data not shown). The mgtC and corA transcripts
scription of the mgtA CR in a fashion dependent on the proline also include long leaders (Lejona et al., 2003) (T. Latifi and E.A.G.,
codons present in the identified ORF. Also, we show that the unpublished results) and specify proteins participating in Mg2+
low Mg2+ signal and the low proline signal act synergistically to homeostasis (Blanc-Potard and Groisman, 1997; Maguire,
increase the mRNA levels for the mgtA CR. Our findings highlight 2006; Soncini et al., 1996).
the complexity of RNA regulatory sequences and provide Sequencing of the insert in pSL55 revealed the presence
a singular example of a leader RNA that utilizes two distinct of 1579 nucleotides corresponding to position 78347–79925 in
mechanisms to detect two different signals. the chromosome of strain 14028s, which is a portion of the
3228 nucleotide long carB gene (see Table S2 for primers).
RESULTS The insert operates in an orientation-dependent manner
because it did not alter mgtA-lac expression when cloned into
Identification of a Translated ORF in the mgtA LR pUC18 where the opposite strand is transcribed (data not
In a search for new factors that might regulate mgtA expression shown). Although this suggested that pSL55 might promote
by acting upon the mgtA leader mRNA, we introduced a plasmid mgtA-lac expression by functioning as an antisense RNA for
library made in the multicopy number vector pUC19 into strain the carB transcript, we ruled out this possibility because
YS774 (see Table S1 available online), which harbors the pSL55 upregulated mgtA transcription even in a strain deleted
PhoP-independent plac1-6 promoter driving transcription of for the carB gene (Figure 2A). Given that a transcript correspond-
a lac fusion in the mgtA CR from the normal (i.e., PhoP-depen- ing to the strand opposite the carB gene was not detected when
dent) start site (Cromie et al., 2006). Ampicillin-resistant trans- RNA was harvested from wild-type Salmonella grown in LB or in

738 Cell 142, 737–748, September 3, 2010 ª2010 Elsevier Inc.


A ing at the ninth codon, produced high levels of b-galactosidase,
whereas an isogenic strain harboring a modified plasmid with
a stop codon after the 17th mgtL codon did not (Figure S1). (There
is a potential start codon at position 26–28 in the Salmonella mgtA
leader sequence that is in frame with the mgtL ORF and is
preceded by a possible ribosome binding site [Figure 1]. However,
the AUG at this position does not appear to be a true translation
start site because first, the sequence between nucleotides 26 and
70 is not conserved in the other examined species, and second,
wild-type Salmonella harboring plasmid pmgtLD2nt-0 lacZ, a
derivative of plasmid pmgtL-0 lacZ with two nucleotides deleted
at position 29–30, which creates a stop codon at position
79–81 for the ORF starting at position 26, still expressed high
levels of b-galactosidase [Figure S1].) The strains harboring plas-
mids pmgtL-0 lacZ and pmgtLD2nt-0 lacZ displayed similar levels
B of b-galactosidase following growth in low and high Mg2+
(Figure S1). These data indicate that mgtL translation is not regu-
lated by changes in the Mg2+ concentration in the media. More-
over, they reflect that in the pmgtL-0 lacZ and pmgtLD2nt-0 lacZ
plasmids, mgtL-lac transcription is initiated from a Mg2+-blind
promoter and the Mg2+-responding riboswitch is disrupted.
Figure 2. Plasmid pSL55 Promotes mgtA-lac Transcription by Inter-
fering with mgtL Translation Inhibiting mgtL Translation Promotes Expression
(A) b-galactosidase activity (Miller units) from a chromosomal mgtA-lac tran- of the mgtA CR
scriptional fusion driven by the plac1-6 promoter was determined in wild-type Plasmid pSL55 appears to upregulate mgtA-lac expression by
(YS809), carB (SP31), and hfq (SP54) strains harboring plasmids pSL55,
producing a trans-acting RNA that interferes with mgtL transla-
pSL55-Mut, or a control plasmid. Bacteria were grown in LB medium for
4.5 hr. Shown are the mean and standard deviation (SD) from two independent tion because first, the region of complementarity with the mgtA
experiments. leader corresponds to the ribosome binding site and start codon
(B) Nucleotide sequences corresponding to position 61–75 from the mgtA LR of mgtL (Figure 2B). Second, inactivation of the hfq gene, which
including the mgtL ribosome binding sequence (RBS; horizontal black line) and codes for the RNA chaperone usually required for pairing
start codon (bold AUG) (top); the sequence of the pSL55-derived transcript between trans-acting regulatory RNAs and their targets
that is complementary to the mgtA leader mRNA (middle); and the sequence
(Brennan and Link, 2007), eliminated pSL55’s regulatory effect
of the pSL55-Mut-derived transcript that is no longer complementary to
the mgtA leader mRNA (bottom).
(Figure 2A). And third, pSL55-Mut, a pSL55 derivative with nucle-
otide substitutions in the region of complementarity with the
mgtA LR (Figure 2B), could not upregulate mgtA expression
N-minimal media (data not shown), and that mgtA-lac was not (Figure 2A).
derepressed in the DcarB strain (Figure 2A), we concluded that If plasmid pSL55 derepresses mgtA-lac transcription by
pSL55 upregulates mgtA-lac expression by a mechanism that inhibiting mgtL translation, we reasoned that disrupting mgtL
does not involve the carB gene. translation by other means might have the same effect. To test
We then examined the nucleotide sequence of the insert in this hypothesis, we introduced mutations in plasmid pYS1010,
pSL55 and identified a 15 nucleotide segment that is comple- which harbors the plac1-6 derivative of the lac promoter driving
mentary to nucleotides 61–75 in the mgtA leader RNA transcription of the full-length mgtA LR fused to a promoterless
(Figure 2B). Interestingly, this region corresponds to the potential lacZ gene and confers Mg2+-regulated synthesis of b-galactosi-
start codon and ribosome binding site for a previously unidenti- dase (Cromie et al., 2006). Wild-type Salmonella harboring
fied 18 codon ORF—hereafter designated mgtL—that begins at pYS1010 derivatives with stop codons at positions 80–82,
position 71 and ends at position 124 (Figure 1). The presence of 89–91, or 98–100 (Figure 1) produced high levels of b-galactosi-
an ORF and ribosome binding site is conserved in the mgtA LRs dase when grown in high Mg2+ (Figure 4A). These levels were 46-
from Escherichia coli, Shigella flexneri, Klebsiella pneumoniae, to 89-fold higher than those displayed by the strain carrying the
Citrobacter koseri, Enterobacter sp. 638, Dickeya zeae, Dickeya original plasmid pYS1010.
dadantii, and Serratia proteamaculans (Figure 3A). The mgtA The high expression levels exhibited by strains harboring
leader sequences from all these species have the potential ability pYS1010 derivatives with stop codon mutations in mgtL are
to form the alternative stem loop structures B and C identified in probably due to lack of translation of the full-length mgtL
the Salmonella mgtA leader (Figure 1) (Cromie et al., 2006) (data (as opposed to resulting from major alterations in the mgtA
not shown). leader RNA structure that lock the riboswitch in an ON state)
We determined that mgtL is translated in vivo because wild- because first, a plasmid expressing the amber suppressor
type Salmonella carrying the medium-copy-number plasmid supF restored normal levels of mgtA-lac transcription to the
pmgtL-0 lacZ, with the full mgtL coding sequence and its putative strain harboring a pYS1010 derivative with an amber stop codon
ribosome binding site fused in frame to the E. coli lacZ gene start- at position 98–100 whereas the plasmid vector had no effect

Cell 142, 737–748, September 3, 2010 ª2010 Elsevier Inc. 739


A

Figure 3. The Presence of an Open Reading Frame and Ribosome-Binding Site Is Conserved in the mgtA LR from Salmonella enterica,
Escherichia coli, Shigella flexneri, Citrobacter koseri, Klebsiella pneumoniae, Enterobacter sp. 638, Dickeya zeae, Dickeya dadantii, and
Serratia proteamaculans
(A) Alignment of the RNA sequences corresponding to the ribosome binding site and mgtL from the species listed above. Sequences in blue correspond to mgtL.
Asterisks correspond to nucleotides conserved in all species. The number of nucleotides shown in each line is indicated to the right of the nucleotide sequences.
(B) Alignment of the deduced amino acid sequences corresponding to mgtL from the species listed above. Sequences in yellow correspond to proline residues.
Asterisks correspond to positions conserved in all species. The number of sense codons in the mgtL ORFs is indicated to the right of the deduced amino acid
sequences.

(Figure 4C). Furthermore, the supF-promoted suppression was promoted by protein synthesis inhibitors is not limited to a partic-
specific because the supF-carrying plasmid did not reduce ular mechanism of action. Tetracycline exerts its effect on the
expression in a strain carrying a pYS1010 derivative with an mgtA LR (as opposed to the mgtA promoter) because
opal stop codon at the same position (Figure 4C), nor did it increased lacZ mRNA levels in wild-type Salmonella harboring
it modify it in a strain with the original pYS1010 plasmid carrying plasmid pYS1010 (data not shown). Furthermore, tetracycline
the wild-type mgtA leader sequence (Figure 4C). Second, acts by inhibiting mgtL translation because it did not promote
the structures of the wild-type and stop codon mutant mgtA a significant increase in lacZ mRNA levels in wild-type Salmo-
leaders were comparable when examined by in-line probing nella carrying the pYS1010 derivative with a stop codon at posi-
(Figure S2A). And third, in vitro transcription assays demon- tion 98–100 (data not shown). Taken together, our results
strated that Mg2+ regulated transcription elongation beyond indicate that interfering with mgtL translation by genetic or phar-
the mgtA LR similarly in DNA templates corresponding to the macological means increases the mRNA levels for the mgtA CR.
wild-type and stop codon mutant mgtA leaders (Figure S2B). The mgtL-encoded peptide does not appear to act in trans
We next tested whether inhibiting mgtL translation by means because a plasmid carrying mgtL DNA failed to restore normal
other than mutation of the mgtA LR affected the mRNA levels mgtA-lac expression in strains harboring pYS1010 derivatives
for the mgtA CR. The protein synthesis inhibitor tetracycline with stop codons at different positions in mgtL (Figure S2C).
promoted an 28-fold increase in the mgtA CR mRNA 15 min This suggested that mgtL exerts its regulatory effect in cis and
after its addition to wild-type Salmonella (Figure 4D). Thus, tetra- raised the question as to the physiological signal controlling
cycline could overcome the transcriptional silencing of the mgtA mgtL translation.
CR that normally takes place when wild-type Salmonella experi-
ences 500 mM Mg2+ (Cromie and Groisman, 2010; Cromie et al., Proline Limitation Enhances Transcription
2006). Tetracycline appears to affect the mRNA levels produced of the mgtA CR
from the mgtA CR specifically because there was little change in The Salmonella mgtL sequence includes four proline codons
the mRNA levels of the mgtA LR or the phoP CR, which were (Figure 1), which is a disproportionately high frequency for an
examined as controls (Figure 4D). Addition of chloramphenicol 18 codon ORF. The number and location of the proline codons
also increased the mRNA levels of the mgtA CR in wild-type (at the third, fifth, seventh, and ninth positions) are largely
Salmonella (data not shown), implying that the derepression conserved in the nine examined species (Figures 3A and 3B).

740 Cell 142, 737–748, September 3, 2010 ª2010 Elsevier Inc.


A B Figure 4. Translation of mgtL Governs
10000 10000 Transcription Elongation beyond the
mgtA LR
-galactosidase activity

-galactosidase activity
8000 8000
(A) b-galactosidase activity (Miller units) produced
(Miller units)

(Miller units)
6000 by wild-type Salmonella (14028s) harboring 6000
plasmid pYS1010 with the wild-type mgtA leader
4000 4000 (Cromie et al., 2006), or derivatives with stop
2000 2000
codons at different positions in mgtL and/or
mutations that hinder stem loop C formation:
0 0 pstop 80–82; pstop 89–91; pstop 98–100; pstop
2

12
2
-91

0
0

09

12
0C

5G
5G
-82

0C

5G
-91

5G
0C
09

0C
-82
-11

107–109; pstop 110–112; pstop 107–109 D110–


-11
-10
01

01

-1 0

0-1
7-1

0-1
7-1
12

12

14
14

12

14
12
89

14
80

89
80
S1

S1
0
10
98

98
11

-G

-G

11
0-C

0-C

-G
0-C
0-G

0-C
10

11
10
top

112; pYS1010-G120C; pstop 80–82-G120C;


top
pY

top
pY

top
1
top

top
10

-82

-82
top
top

to p
to p
-10
01

01

-10
01
ps
ps

09

ps
ps

09
0

pYS1010-C145G; and pstop 98–100-C145G.


ps

80

ps

80
S1

S1

S1
S1
ps
ps

ps
98
7-1

98
ps

7-1
top
pY

pY

to p

pY
pY
top

top
10

Bacteria were grown in N-minimal medium with

10
ps

ps
top

ps

top

ps
10 mM Mg2+ for 4 hr. Shown are the mean and
ps

ps
SD from at least three independent experiments.
C D (B) b-galactosidase activity produced by the
pstop 98-100 pstop 98-100 pYS1010
strains listed in (A). Bacteria were grown in
40
-galactosidase activity

5000
(amber) (opal) N-minimal medium with 10 mM Mg2+ for 4 hr.
Shown are the mean and SD from at least three
(Miller units)

Fold change

30
(T15 / T0 )

4000
independent experiments.
3000
20 (C) b-galactosidase activity produced by wild-type
2000 Salmonella (14028s) harboring plasmid pUHsupF
1000
10
or the plasmid vector pUH21-2lacIq, and
0
pYS1010 or its derivatives with UAG (pstop
0
98–100; amber) or UGA (pstop 98–100; opal)
or

or

ng
or

er

ng
F
F

F
up
up

up

ad
ct

ct
ct

di

di

stop codons at position 98–100 in the mgtA leader.


ve

ve
ve

co
le

co
Hs
Hs

Hs

oP
pU

pU
pU

gt

gt

Bacteria were grown in N-minimal medium with


ph
m

500 mM Mg2+ for 3 hr and with 1 mM IPTG for


E F 1 hr. Shown are the mean and SD from three inde-
12
2.0 pendent experiments.
10
(no proline / proline)

(no proline / proline)

(D) Fold change in the mRNA levels of the mgtA LR


1.5
Fold change

Fold change

8 and the mgtA and phoP CRs produced by wild-


6 1.0
type Salmonella (14028s) treated with the protein
synthesis inhibitor tetracycline (25 mg/ml). Expres-
4
0.5 sion levels of target genes were normalized to that
2
of the 16S ribosomal RNA rrs gene. Fold change
0 0 was calculated by dividing the mRNA levels from
ng
ng

ng
er

er

ng

samples taken 15 min after treatment with tetracy-


ad

ad
di
di

di

di
co
co

co
le

le

co

cline (T150 ) by that of samples taken before addi-


A

A
A

oP

oP
gt

gt
gt

gt
ph

tion of the antibiotic (T00 ). Shown are the mean


m

ph
m
m

and SD from three independent experiments.


(E) Fold change in the mRNA levels of the mgtA LR and the mgtA and phoP CRs produced by a proline auxotrophic strain (EG19886) grown in modified N-minimal
media with 500 mM Mg2+ in the presence of 1 mM proline for 1 hr, and then grown for 15 min in media containing or lacking proline. Expression levels of target
genes were normalized as described in (D). Fold change was calculated by dividing the mRNA levels of cells grown in the absence of proline by that of cells grown
in the presence of proline. Shown are the mean and SD from three independent experiments.
(F) Fold change in the mRNA levels of the mgtA LR and the mgtA and phoP CRs produced by wild-type Salmonella (14028s) grown and analyzed as described in
(D). Shown are the mean and SD from two independent experiments.
See also Figure S2.

Because the proline codons are located in the mgtL region where The mRNA level for the mgtA CR was 8-fold higher in organisms
introduction of stop codons heightens expression downstream of grown in the absence of proline than in those grown in its pres-
the mgtA leader (Figure 1), we hypothesized that a drop in the ence (Figure 4E). This effect was unique to the mgtA CR, as no
cytosolic proline levels might decrease the availability of proline- differences were detected in the mRNA levels corresponding
charged tRNAs, causing ribosome stalling at mgtL proline to the mgtA LR or the phoP CR (Figure 4E). Furthermore,
codons, leading to formation of stem loop C and an increase in it was specific for proline, as limitation for histidine, which does
the levels of the mgtA CR mRNA. By contrast, when cytosolic not have codons in the mgtL sequence (Figure 1), failed to
proline is abundant, coupling of mgtA leader transcription and increase the mRNA level for the mgtA CR (data not shown).
mgtL translation would be restored, thereby favoring formation Indeed, despite the presence of three arginine codons in mgtL
of stem loop B and reducing the production of mgtA CR mRNA. (Figure 1), arginine limitation did not promote an increase in the
To explore this hypothesis, we grew a proline auxotroph in the mRNA levels of the mgtA CR in an arginine auxotroph (data not
presence of 1 mM proline for 1 hr, washed the cells, split shown). Perhaps this reflects the location of the arginine codons,
the culture into two different media—one containing and one one of which is in stem loop C, and that the remaining arginine
lacking proline—and then harvested the mRNA 15 min later. codons may not be sufficient to mediate mgtA derepression.

Cell 142, 737–748, September 3, 2010 ª2010 Elsevier Inc. 741


A Proline limitation promoted a 2-fold increase in the mRNA
mgtA leader mgtA coding phoP coding
levels of the mgtA CR in wild-type (i.e., prototrophic) Salmonella
14
(Figure 4F). Although this is lower than the increase observed in
12 the proline auxotroph (Figure 4E), it is similar to the gene expres-
(no proline / proline)

10 sion changes promoted by branched-chain amino acids in the


Fold change

8
attenuation-regulated ilvGMEDA operon (Chen et al., 1991).
Moreover, it is higher than what has been reported for the atten-
6
uation-regulated trp operon, where starvation for tryptophan
4 does not alter trp expression in a tryptophan prototroph
2 (Yanofsky and Horn, 1994).
0
L+

L+

L+
Pr o3

7,9

7,9
o3
9

9
7 ,9
Pr o3
P r 5 ,7 ,

Pr 5,7,
Pr 5,7,

The mgtL Proline Codons Are Necessary


gt

gt

gt
Pr

Pr

Pr
,5,

,5 ,

,5,
m

m
o

o
o3

o3

o3
Pr

for the Response to Proline Limitation


We determined that proline limitation failed to promote an
B increase in the mRNA levels for the mgtA CR in a derivative of
80 mgtA leader mgtA coding phoP coding the proline auxotroph with all four mgtL proline codons replaced
by codons specifying other amino acids (Figure 5A). As found
(10 µ M Mg2+ / 500 µM Mg2+)

70

60 with the isogenic mgtL+ strain, proline limitation had little effect
on the mRNA levels for the mgtA leader and phoP CRs
Fold change

50

40
(Figure 5A). Importantly, the mutant still responded to changes
in the Mg2+ concentration (Figure 5B) (albeit not as well as the
30
isogenic strain with a wild-type mgtA leader), indicating that
20
the proline codon substitutions did not lock the mutant into an
10 inactive conformation. These data indicate that the mgtL proline
0 codons are essential for the response to proline limitation, and
L+

L+

L+
Pr o3

7,9

7 ,9
Pr o3
9

o 3 ,9

9
7 ,9
Pr o3

they suggest that this ability is distinct from the mgtA leader’s
Pr 5,7,

Pr 5,7,
,7
gt

gt

gt
Pr

Pr

Pr
,5 ,
,5,

,5,
o5
m

m
o

role as a Mg2+-sensing device.


o3

o3
Pr

We hypothesized that the proline codon at the third position of


mgtL might not be required for regulation of mgtA expression by
C proline because D. dadantii does not have a proline codon at this
80
mgtA leader mgtA coding phoP coding position (Figures 3A and 3B). Indeed, a Salmonella strain in which
70
the mgtL proline codon at the third position was replaced by
Relative mRNA levels

60 a leucine codon derepressed the mgtA CR mRNA when limited


50 for proline, like the strain with the wild-type mgtL (Figure 5A).
40 However, this mutant still responded to changes in the Mg2+
concentration, but to a smaller degree than the mutant
30
substituted in all four mgtL proline codons (Figure 5B).
20
The results presented above suggested that replacement of
10 the mgtL proline codons at the fifth, seventh, and ninth positions
0 by codons specifying other amino acids might be sufficient to
H H L L H H L L H H L L [Mg2+]
+ - + - + - + - + - + - Proline eliminate the response to proline. As predicted, there was little
derepression of the mgtA CR mRNA upon proline limitation in
Figure 5. The mgtA Leader Responds to Proline and Mg2+ Indepen-
a derivative of the proline auxotroph with the last three mgtL
dently and Requires the mgtL Proline Codons for Regulation of the proline codons substituted by the same codons as in the mutant
mgtA CR by Changes in the Proline Concentration with substitutions in all four mgtL proline codons (Figure 5A). As
(A) Fold change in the mRNA levels of the mgtA LR and the mgtA and phoP with the other isogenic strains, proline limitation did not affect the
CRs produced by a proline auxotroph (SP1; mgtL+), or derivatives in which mRNA levels of the mgtA leader and phoP CRs (Figure 5A).
the mgtL proline codon at the third position was substituted (SP2; Pro3), the
The response to Mg2+ of the mgtL derivative with substitutions
last three mgtL proline codons were substituted (SP61; Pro5,7,9), or in which
in the last three proline codons was intermediate to that
all mgtL proline codons were substituted (SP8; Pro3,5,7,9). Bacteria were
grown and mRNA was analyzed as described in Figure 4E. Shown are the
mean and SD from three independent experiments.
(B) Fold change in the mRNA levels of the mgtA LR and the mgtA and phoP (C) Relative mRNA levels of the mgtA LR and the mgtA and phoP CRs
CRs produced by the strains described in (A). Bacteria were grown in modified produced by a proline auxotroph (EG19886) that experienced 500 mM Mg2+
N-minimal medium with 10 mM or 500 mM Mg2+ for 3.5 hr. Expression levels of and 1 mM proline, 500 mM Mg2+ and no proline, 5 mM Mg2+ and 1 mM proline,
target genes were normalized as described in (A). Fold change was calculated or 5 mM Mg2+ and no proline for 15 min. Relative mRNA levels were calculated
by dividing the mRNA levels of cells grown in N-minimal medium with 10 mM by dividing the mRNA levels of cells grown in a given condition by those
Mg2+ by those present in cells grown in 500 mM Mg2+. Shown are the mean present in cells grown in 500 mM Mg2+ and 1 mM proline. Shown are the means
and SD from three independent experiments. and SD from three independent experiments.

742 Cell 142, 737–748, September 3, 2010 ª2010 Elsevier Inc.


exhibited by the one with the third proline codon substituted and A mgtL+
the one with all four proline codons substituted (Figure 5B).
mgtA leader mgtA coding phoP coding
7
Synergism between the Low Mg2+ and the Proline
6

Relative mRNA levels


Limitation Signals
To examine whether direct Mg2+ sensing by the mgtA leader RNA 5

affects indirect proline sensing by the mgtL ORF and vice versa, 4
we grew the mgtL+ proline auxotrophic strain in the presence of 3
500 mM Mg2+ and 1 mM proline for 1 hr, washed the cells, and split
2
the culture into four different media containing either 500 mM
Mg2+ and 1 mM proline, 500 mM Mg2+ and no proline, 5 mM 1
Mg2+ and 1 mM proline, or 5 mM Mg2+ and no proline. Fifteen 0
minutes later, we determined the mRNA levels corresponding - G N NG NP - G N NG NP - G N NG NP
to the mgtA and phoP CRs as well as to the mgtA LR. We found
that the mRNA levels corresponding to the mgtA CR were higher
B
in cells experiencing both low Mg2+ and low proline than in cells Pro3,5,7,9
limited only for Mg2+ or for proline (Figure 5C). As expected, the mgtA leader mgtA coding phoP coding
mRNA levels corresponding to the mgtA LR and the phoP CR 7

were higher in cells experiencing 5 mM Mg2+ than in those Relative mRNA levels
6
exposed to 500 mM Mg2+ (Figure 5C), reflecting that the amount 5
of activated PhoP protein increases as the Mg2+ concentration
4
in the media decreases (Shin et al., 2006). Our results support
the notion that Mg2+ and proline are sensed independently 3
by the mgtA leader RNA. Furthermore, they suggest synergism 2
between the two inducing signals because the mRNA levels for
1
the mgtA CR were higher in cells experiencing both low Mg2+
and low proline than the sum of the mRNA levels produced by 0
- G N NG NP - G N NG NP - G N NG NP
cells experiencing only low Mg2+ or low proline (Figure 5C).
In agreement with this notion, wild-type Salmonella harboring
Figure 6. Hyperosmotic Shock Promotes Transcription of the mgtA
plasmid pYS1010 derivatives with early stop codons in the
CR in a Process that Requires the mgtL Proline Codons
mgtL sequence produced more b-galactosidase when grown in Relative mRNA levels of the mgtA LR and the mgtA and phoP CRs produced
low Mg2+ than the sum of the b-galactosidase activities produced by wild-type Salmonella (YS957) (A), or a derivative in which all mgtL proline
by the same strains grown in high Mg2+ plus the b-galactosidase codons were substituted (EG19870) (B). Bacteria were grown for 1 hr in modi-
activity produced by wild-type Salmonella carrying pYS1010 fied N-minimal medium without casamino acids containing 500 mM Mg2+ and
grown in low Mg2+ (Figures 4A and 4B). either no additional supplements (-), 1 mM glycine betaine (G), 0.3 M NaCl (N),
0.3 M NaCl and 1 mM glycine betaine (NG), or 0.3 M NaCl and 1 mM proline
(NP). Relative mRNA levels were calculated by dividing the mRNA levels of
Hyperosmotic Shock Promotes an Increase in the mgtA
cells grown under the specified condition by the mRNA levels present in cells
CR mRNA Levels grown in 500 mM Mg2+ (i.e., with no additional supplement). Shown are the
Proline plays two major functions in bacterial cells: it is a compo- mean and SD from three independent experiments.
nent of proteins and it can function as an osmoprotectant
(Csonka and Leisinger, 2007). Thus, we hypothesized that
when bacteria experience hyperosmotic shock, the increased the induction of the mgtA CR mRNA promoted by NaCl but
requirement for proline in osmoprotection might decrease its had negligible effects when added in the absence of NaCl
availability to charge proline tRNAs. This could potentially lead (Figure 6A). Proline had a similar (albeit not as strong) effect as
to ribosome stalling at the mgtL proline codons and result in glycine betaine (Figure 6A), presumably because it is not as
derepression of the mgtA CR. We tested this hypothesis by effective as glycine betaine in osmoprotection (Cayley et al.,
comparing the mRNA levels produced by wild-type Salmonella 1992). The increase in the mgtA CR mRNA levels provoked by
experiencing hyperosmotic shock in the presence or absence hyperosmotic shock requires an intact mgtL ORF, because it
of osmoprotectants. was not observed in an isogenic strain substituted in all four
We determined that the mRNA levels corresponding to the mgtL proline codons (Figure 6B). Cumulatively, these data
mgtA CR were 6-fold higher when Salmonella experienced demonstrate that hyperosmotic shock promotes transcription
500 mM Mg2+ + 0.3 M NaCl for 1 hr than in organisms grown in of the mgtA CR in an mgtL-dependent manner.
500 mM Mg2+ (Figure 6A). By contrast, the mRNA levels for the
mgtA LR and the phoP CR were similar under the two growth Formation of Stem Loop C Is Necessary
conditions (Figure 6A), indicative that the induction of the mgtA for Transcription of the mgtA CR
CR mRNA levels promoted by high osmolarity is not mediated Because transcription and translation are coupled in bacteria,
by the PhoP-dependent mgtA promoter. Addition of the osmo- when cytosolic proline is abundant, a ribosome translating the
protectant glycine betaine together with NaCl compromised complete mgtL sequence is likely to occlude the left arm of

Cell 142, 737–748, September 3, 2010 ª2010 Elsevier Inc. 743


A mgtA leader region B trp leader region Figure 7. Regulation of Transcription Elon-
gation into the mgtA and trp CRs by ORFs
Located within Their Respective LRs
Antiterminator
(A) Regulation of transcription elongation by the
Stem-loop C mgtA leader RNA. Top: when proline levels are
limiting in the cytosol, a ribosome translating
Ribosome is stalled at Ribosome Ribosome mgtL stalls at proline codons, which favors forma-
STOP tion of stem loop C and results in transcription of
proline/tryptophan
AUG
G AUG STOP
the mgtA CR. Middle: when proline levels are not
codons RBS RBS UUUUUUU
limiting in the cytosol, a ribosome can translate
the complete mgtL ORF, which favors formation
ON state ON state of stem loop B and hinders transcription elonga-
tion into the mgtA CR by an unknown mechanism.
Stem-loop B Bottom: a ribosome loads but cannot translate
a mutant mgtA leader with nucleotide substitutions
Terminator
in the mgtL start codon, which favors formation of
stem loop C and results in transcription of the
Leader ORF is mgtA CR.
AUG STOP AUG STOP
completely translated RBS RBS
(B) Regulation of transcription elongation by the trp
UUUUUUU
leader RNA. Top: when tryptophan levels are limiting
in the cytosol, a ribosome translating trpL stalls at
OFF OFF state
tryptophan codons, which favors formation of an
state
antiterminator structure and results in transcription
of the trp operon. Middle: when tryptophan levels
Anti-antiterminator
Stem-loop C
are not limiting in the cytosol, a ribosome can trans-
STOP
Terminator
late the complete trpL ORF, which favors formation
of an intrinsic transcription terminator and there is no
Ribosome loads but transcription of the trp operon. Bottom: a ribosome
STOP
there is no mgtL or loads but cannot translate a mutant trp leader with
AUG
AU AU
AUG
trpL translation RBS RBS UUUUUUU nucleotide substitutions in the trpL start codon,
which favors formation of both the anti-antitermina-
ON state OFF state tor and terminator structures and there is no tran-
scription of the trp operon. Note the different posi-
tion of the mgtL and trpL ORFs relative to the
sequences forming stem loop C and the anti-antiter-
minator structures, respectively, which determines
the different phenotypes resulting from mutation of
the mgtL and trpL start codons.

stem loop C (Figure 1 and Figure 7A). This would favor formation tive with a stop codon at position 107–109 no longer conferred
of stem loop B, which has been shown to hamper transcription high levels of b-galactosidase upon wild-type Salmonella when
elongation beyond the mgtA LR (Cromie et al., 2006). nucleotides 110–112 were deleted (Figure 4A), which brought
By contrast, conditions that reduce the levels of free cytosolic the mgtL stop codon only six nucleotides away from the left
proline would promote ribosome stalling at the mgtL proline arm of stem loop C. Cumulatively, these results indicate that
codons, thereby advancing formation of stem loop C and result- the distance between the ribosome translating mgtL and the
ing in transcription elongation into the mgtA CR (Figure 1 and nucleotides forming the left arm of stem loop C (as opposed to
Figure 7A). Therefore, the position that a translating ribosome the size of the translated mgtL product) is critical for mgtL-medi-
reaches in the mgtL ORF should determine whether transcription ated gene control.
continues into the mgtA CR (Figure 1 and Figure 7A). If the lack of translation of the full-length mgtL stimulates tran-
We tested our model by investigating the phenotype of wild- scription elongation beyond the mgtA LR by favoring formation of
type Salmonella harboring pYS1010 derivatives with stop stem loop C (Figure 1 and Figure 7A), hindering formation of stem
codons at different positions within mgtL. A derivative with loop C should abolish this stimulation. As predicted, the G120C
a stop codon at position 110–112, which is only six nucleotides and C145G substitutions in the mgtA leader (Figure 1), which
upstream of the left arm of stem loop C (Figure 1), produced were previously shown to impede formation of stem loop C
very low levels of b-galactosidase when grown in 10 mM Mg2+ (Cromie et al., 2006), thwarted derepression in constructs
(Figure 4A), like the isogenic strain with plasmid pYS1010 harboring stop codons at either of two mgtL positions (Figure 4A).
harboring the wild-type mgtA leader (Figure 4A). By contrast,
there were high levels of b-galactosidase in a pYS1010 derivative DISCUSSION
with a stop codon at position 107–109 (Figure 4A), which is nine
nucleotides from the left arm of stem loop C (Figure 1), similar to The LR of many mRNAs can respond to specific nutritional
the behavior of strains with stop codons at positions 80–82, and/or physical signals by modifying expression of the associ-
89–91, or 98–100 (Figure 4A). Furthermore, the pYS1010 deriva- ated downstream coding sequences. Some of these leader

744 Cell 142, 737–748, September 3, 2010 ª2010 Elsevier Inc.


RNAs rely on the same mechanism to detect more than one some is too close to the sequences that make up stem loop C,
signal whereas others utilize different means of controlling then the alternative stem loop B would form (Figure 1 and
gene expression in response to one signal. For example, two Figure 7A), and transcription would not continue into the mgtA
riboswitches located in tandem, one responding to S-adenosyl- CR. For instance, stop codons at positions 80–82, 89–91,
methionine and the other to coenzyme B12, control transcription 98–100, or 107–109 in the mgtA leader resulted in derepression
of the metE gene in Bacillus clausii (Sudarsan et al., 2006). In whereas a stop codon at position 110–112 did not (Figure 4A).
other cases, a cis-acting riboswitch can also function as Yet, the stop codon at position 107–109 no longer derepressed
a trans-acting regulatory RNA to modulate the expression of expression if nucleotides 110–112 were also deleted (Figure 4A),
genes located somewhere else in the genome (Loh et al., which moved the mgtL stop codon only six nucleotides away
2009). The mgtA leader described here constitutes a singular from the left arm of stem loop C (Figure 1).
example of an mRNA leader that utilizes different mechanisms It was recently reported that a chromosomal C98T mutation in
to sense different signals. the leader RNA causes constitutive high expression of the mgtA
We determined that the mRNA leader corresponding to the CR (O’Connor et al., 2009). In the original description of this
Mg2+ transporter gene mgtA harbors a translated ORF rich in mutation, it was not clear why this nucleotide substitution should
proline codons designated mgtL (Figure 1; Figure S1), which is alter mgtA expression, but we now see that it created a stop
conserved in other bacterial species (Figures 3A and 3B) and codon in mgtL that is identical to the mutation we had previously
enables Salmonella to regulate transcription elongation into the constructed, which resulted in constitutive high expression of
mgtA CR in response to the levels of cytoplasmic proline. mgtA (Figures 4A and 4B).
Whereas the mgtA leader RNA senses Mg2+ directly (Cromie The mgtL proline codons are located at positions 77–79,
et al., 2006), it monitors proline levels in an indirect fashion, via 83–85, 89–91, and 95–97 (Figure 1), which correspond to the
mgtL translation (Figure 4A), using an attenuation-like mecha- region where placing a stop codon results in derepression (Fig-
nism (Landick et al., 1996; Henkin and Yanofsky, 2002). ure 4A). Therefore, we propose that when cytosolic proline levels
The dual sensing ability of the mgtA leader allows Salmonella are low, there would be less proline-charged tRNAs, resulting in
to transcribe the mgtA CR not only when Mg2+ levels drop below ribosome stalling at mgtL proline codons. This would favor
a certain threshold (Cromie and Groisman, 2010; Cromie et al., formation of stem loop C and transcription elongation into the
2006) but also under conditions resulting in a decrease in cyto- mgtA CR (Figure 7A). In contrast, when cytosolic proline levels
solic proline levels (Figures 4E and 4F). are high, the ribosome would translate the complete mgtL
The discovery of a translatable ORF in the mgtA leader sequence, thereby occluding the left arm of stem loop C and,
(Figure 1; Figure S1) suggests a possible explanation for the in this manner, favor formation of stem-loop B (Figure 7A) and
recent observation that the mRNA levels corresponding to the hinder transcription of the mgtA CR. Interestingly, despite
50 -most 127 nucleotides of the mgtA leader are much higher utilizing different mechanisms to sense proline and Mg2+, the
than those for the 30 -most 94 nucleotides of the mgtA leader mgtA leader relies on formation of the same structure—stem
when Salmonella is grown in high-Mg2+ media (Spinelli et al., loop C—to promote transcription elongation into the mgtA CR,
2008). This is because the RNA-processing enzyme RNase E, because mutations that hinder formation of stem loop C pre-
which has been implicated in mgtA mRNA degradation during vented the derepression caused by stop codons in mgtL
growth in high Mg2+ (Spinelli et al., 2008), can target nascent (Figure 4A) and by low Mg2+ (Figure 4B) (Cromie et al., 2006).
transcripts as well as complete messages (Hammarlof and The regulation of mgtA by changes in the cytosolic proline
Hughes, 2008). Therefore, the ribosome translating mgtL, which concentration is reminiscent of class I transcription attenuation,
extends to position 124 in the mgtA leader (Figure 1), could where certain amino acid biosynthetic operons are regulated by
protect the 50 region of the mgtA leader from RNase E action, re- translation of a short ORF in the LR. These ORFs are rich in
sulting in higher levels of this portion of the RNA. codons specifying the amino acid(s) synthesized by the enzymes
encoded in the operon (Landick et al., 1996). Yet, there are signif-
mgtL Translation Regulates Transcription icant differences between the two systems. First, mgtA codes for
of the mgtA CR a transporter rather than for an amino acid biosynthetic enzyme.
We determined that interfering with mgtL translation increased This provides experimental support for the proposal, based on
the mRNA levels for the mgtA CR (Figures 4A and 4D–4F). This genomic analysis, that classical transcriptional attenuators
appears to result from the formation of a particular secondary might direct the expression of operons mediating functions other
structure (i.e., stem loop C) in the mgtA leader (Figure 1 and than nutrient biosynthesis (Merino and Yanofsky, 2005). Second,
Figure 7A) that promotes transcription elongation into the mgtA the position of the leader ORF relative to the potential RNA
CR, as nucleotide substitutions impeding formation of stem secondary structures that can be adopted differs between the
loop C (Cromie et al., 2006) prevented derepression provoked mgtA leader (Figure 7A) and the leaders of attenuation-regulated
by stop codon mutations in mgtL (Figure 4A). Because transcrip- amino acid biosynthetic operons such as the trp operon
tion and translation are coupled in bacteria and because the (Figure 7B). This provides a plausible explanation for the oppo-
ribosome occupies 30 nucleotides, covering 12–15 nucleo- site effects that mutation of the leader ORF start codon has in
tides from the P site (Laursen et al., 2005), the position that these two classes of leaders. It results in superattenuation for
a translating ribosome reaches in mgtL relative to the sequences the trp operon (Landick and Yanofsky, 1987) because there
that make up stem loop C would determine whether transcription would be no ribosomes to stall at Trp codons, which would result
continues into the mgtA CR (Figure 1 and Figure 7A). If the ribo- in formation of a terminator structure (Figure 7B). However,

Cell 142, 737–748, September 3, 2010 ª2010 Elsevier Inc. 745


it gives rise to derepression of the mgtA CR (our unpublished and ribosomes, which could be affected during hyperosmotic
results), because formation of stem loop C would be favored stress. On the other hand, CorA-mediated Mg2+ uptake may
when mgtL is not translated (Figure 7A). Third, an intrinsic be compromised because it is driven by the membrane potential
terminator structure forms in the trp leader when the levels of (Froschauer et al., 2004), which decreases in cells experiencing
charged tRNATrp are high and the ribosome does not pause at high osmolarity (Csonka, 1989). Yet, this stress would not affect
the consecutive Trp codons in the leader ORF (Landick and Mg2+ uptake by the P-type ATPase MgtA protein because it is
Yanofsky, 1987) (Figure 7B). This is in contrast to the absence energized by ATP hydrolysis (Maguire, 1992).
of an intrinsic transcription terminator in the mgtA leader, where That proline limitation promotes expression of the MgtA Mg2+
sequences located downstream of the stem loop B structure transporter suggests that there is a physiological connection
(Figure 1) are necessary to control transcription elongation into between proline and Mg2+. In agreement with this notion, tran-
the mgtA CR (Cromie et al., 2006) by an unknown mechanism. scription of the proline transporter gene proP is promoted during
And fourth, apart from utilizing a classical transcription attenua- growth in low Mg2+ by the PhoP protein (Eguchi et al., 2004),
tion-like mechanism to mediate the response to proline, the which directs transcription initiation of the mgtA gene (Garcia
mgtA leader senses Mg2+ directly functioning as a riboswitch Vescovi et al., 1996).
(Cromie et al., 2006). This raises the intriguing possibility of clas-
sical transcriptional attenuators also sensing physical and/or EXPERIMENTAL PROCEDURES
chemical signals directly.
Bacterial Strains, Plasmids, Primers, and Growth Conditions
2+ Bacterial strains and plasmids used in this study are listed in Table S1. Primers
High Osmolarity Enhances the mRNA Levels of the Mg
used in this study are listed in Table S2. Unless otherwise stated, bacteria were
Transporter Gene mgtA grown at 37 C in LB broth or in N-minimal medium (pH 7.4) (Snavely et al.,
Why does proline limitation promote transcription of the Mg2+ 1991) supplemented with 0.1% casamino acids, 38 mM glycerol, and the indi-
transporter gene mgtA? We contemplated the possibility of the cated concentration of MgCl2.
MgtA protein being a conduit for proline whereby cells experi-
encing low proline would derepress expression of a proline Construction of Strains with Chromosomal Mutations
import system. However, we found that [14C]proline uptake Deletion strains were constructed using the one-step inactivation method
(Datsenko and Wanner, 2000). See Extended Experimental Procedures for
was similar between wild-type and mgtA Salmonella, and
detailed protocols.
between a triple mutant lacking all known proline uptake
systems (ProP, ProU, and PutP) (Csonka and Leisinger, 2007) Treatment with Bacteriostatic Protein Synthesis Inhibitors
and the isogenic mgtA derivative (our unpublished results). We An overnight bacterial culture grown in N-minimal medium with 10 mM MgCl2
also considered that MgtA-mediated Mg2+ uptake might be was used to inoculate 125 ml flasks containing 10 ml of the same medium
necessary for proline biosynthesis, because the activity of the (1:50 dilution) and grown for 3 hr at 37 C with shaking. To promote PhoP-
proline biosynthetic enzyme glutamate 5-kinase is Mg2+ depen- dependent mgtA transcription initiation, bacteria were washed and resus-
dent (Perez-Arellano et al., 2005). Yet, inactivation of the mgtA pended in 10 ml of N-minimal medium with 500 mM MgCl2. Following growth
for 1 hr, a 500 ml aliquot was removed to determine the mRNA levels before
gene did not render Salmonella auxotrophic for proline (our
treatment. Subsequently, tetracycline (to 25 mg/ml final concentration) or
unpublished results). chloramphenicol (to 200 mg/ml final concentration) was added and bacteria
We determined that hyperosmotic shock promoted an were grown for 15 min, when they were harvested and their RNA was isolated
increase in the mRNA levels corresponding to the mgtA CR for analysis.
(Figure 6A). This appears to result from a transient decrease in
the levels of charged-proline tRNAs taking place when proline Effect of Proline Limitation
is used as an osmoprotectant, as there would be less cytosolic Bacteria were grown overnight in modified N-minimal medium containing
0.2% glucose, 10 mM MgCl2, and 1 mM proline. The overnight culture was
proline available to charge the tRNAPros. Indeed, the increase
used to inoculate 125 ml flasks containing 10 ml of the same medium (1:50 dilu-
in mgtA mRNA levels was severely compromised if the osmotic tion) and grown for 3 hr at 37 C with shaking. The harvested bacteria were
shock was alleviated by the osmoprotectant glycine betaine, washed in the modified N-minimal medium containing 500 mM MgCl2, and
which did not influence mgtA expression when added in the grown in 10 ml of the same medium with 1 mM proline for 1 hr. After removing
absence of NaCl (Figure 6A). The osmotic shock-promoted a 250 ml aliquot to determine the mRNA levels before treatment, the harvested
increase in the mRNA levels for the mgtA CR requires the mgtL bacteria were washed with the modified N-minimal medium containing 500 mM
MgCl2, and suspended in 100 ml of the same medium. To see the effect of
proline codons, because a mutant with substitutions in all four
proline limitation, the suspended bacterial cells were split into two flasks con-
proline codons in mgtL lost the ability to derepress the mgtA taining the modified N-minimal media with 500 mM MgCl2 and a mixture of
CR mRNA in response to hyperosmotic shock (Figure 6B). each of the 19 essential amino acids (50 mM final concentration for each of
Why does hyperosmotic shock induce transcription of the them): one flask contained 1 mM proline and the other had no proline added.
Mg2+ transporter gene mgtA when Salmonella also harbors Bacteria were grown for 15 min, when they were harvested and their RNA was
the constitutively expressed Mg2+ transporter CorA? On the isolated for analysis.
one hand, high osmolarity promotes excretion of putrescine
(Schiller et al., 2000), which constitutes the major organic diva- Testing Effect of Growth in High versus Low Mg2+
Bacteria were grown overnight in modified N-minimal medium containing
lent cation in bacterial cells and is normally bound to nucleic
0.2% glucose, 10 mM MgCl2, and 1 mM proline. The harvested bacteria
acids (Wortham et al., 2007). This might create an increased were washed in the modified N-minimal medium containing 500 mM MgCl2,
need for Mg2+ in order to neutralize the negatively charged and this culture was used to inoculate 125 ml flasks containing 10 ml of the
DNA and RNA, and to stabilize structures such as membranes modified N-minimal medium containing either 500 mM MgCl2 or 10 mM

746 Cell 142, 737–748, September 3, 2010 ª2010 Elsevier Inc.


MgCl2 (1:50 dilution) and grown for 3.5 hr at 37 C with shaking. The bacteria REFERENCES
were then harvested and their RNA was isolated for analysis.
Bader, M.W., Sanowar, S., Daley, M.E., Schneider, A.R., Cho, U., Xu, W.,
2+
Klevit, R.E., Le Moual, H., and Miller, S.I. (2005). Recognition of antimicrobial
Testing Effect of Mg and/or Proline Limitation Treatment peptides by a bacterial sensor kinase. Cell 122, 461–472.
Bacteria were grown overnight in modified N-minimal medium containing
Barchiesi, J., Castelli, M.E., Soncini, F.C., and Vescovi, E.G. (2008). mgtA
0.2% glucose, 10 mM MgCl2, and 1 mM proline. The overnight culture
expression is induced by rob overexpression and mediates a Salmonella en-
was used to inoculate 125 ml flasks containing 20 ml of the same medium
terica resistance phenotype. J. Bacteriol. 190, 4951–4958.
(1:50 dilution) and grown for 3 hr at 37 C with shaking. The harvested bacteria
were washed in the modified N-minimal medium containing 500 mM MgCl2, Blanc-Potard, A.B., and Groisman, E.A. (1997). The Salmonella selC locus
and grown in 20 ml of the same medium with 1 mM proline for 1 hr. The contains a pathogenicity island mediating intramacrophage survival. EMBO
harvested bacteria were washed with the modified N-minimal medium J. 16, 5376–5385.
containing 500 mM MgCl2, and suspended in 200 ml of the same medium. Brennan, R.G., and Link, T.M. (2007). Hfq structure, function and ligand
This cell suspension was used to inoculate 125 ml flasks containing 5 ml of binding. Curr. Opin. Microbiol. 10, 125–133.
media containing either 500 mM MgCl2 and 1 mM proline, 500 mM MgCl2
Cayley, S., Lewis, B.A., and Record, M.T., Jr. (1992). Origins of the osmopro-
and no proline, no MgCl2 and 1 mM proline, or no MgCl2 and no proline
tective properties of betaine and proline in Escherichia coli K-12. J. Bacteriol.
(1:100 dilution). Bacteria were grown for 15 min, when they were harvested
174, 1586–1595.
and their RNA was isolated for analysis.
Chen, J.W., Bennett, D.C., and Umbarger, H.E. (1991). Specificity of attenua-
tion control in the ilvGMEDA operon of Escherichia coli K-12. J. Bacteriol. 173,
Testing Effect of Hyperosmotic Shock 2328–2340.
Bacteria were grown overnight in modified N-minimal medium containing Choi, E., Groisman, E.A., and Shin, D. (2009). Activated by different signals, the
0.2% glucose, 0.1% casamino acids, and 10 mM MgCl2. The overnight culture PhoP/PhoQ two-component system differentially regulates metal uptake.
was used to inoculate 125 ml flasks containing 20 ml of the same medium J. Bacteriol. 191, 7174–7181.
(1:50 dilution) and grown for 3.5 hr at 37 C with shaking. The harvested
Cromie, M.J., and Groisman, E.A. (2010). Promoter and riboswitch control of
bacteria were washed in modified N-minimal medium containing 500 mM
the Mg2+ transporter MgtA from Salmonella enterica. J. Bacteriol. 192,
MgCl2 without casamino acids and suspended in 250 ml of the same medium.
604–607.
This cell suspension was used to inoculate 125 ml flasks containing 5 ml of the
modified N-minimal medium without casamino acids containing 500 mM MgCl2 Cromie, M.J., Shi, Y., Latifi, T., and Groisman, E.A. (2006). An RNA sensor for
and either no additional supplements, 1 mM glycine betaine, 0.3 M NaCl, 0.3 M intracellular Mg2+. Cell 125, 71–84.
NaCl and 1 mM glycine betaine, or 0.3 M NaCl and 1 mM proline. Bacteria were Csonka, L.N. (1989). Physiological and genetic responses of bacteria to
grown for 1 hr at 37 C with shaking, when they were harvested and their RNA osmotic stress. Microbiol. Rev. 53, 121–147.
was isolated for analysis. Csonka, L.N., and Leisinger, T. (2007). Biosynthesis of proline. In EcoSal—Es-
cherichia coli and Salmonella: Cellular and Molecular Biology, G. Cohen, ed.
(Washington, DC: ASM Press), http://www.ecosal.org.
RNA Isolation and Determination of Transcript Levels
Total RNA was extracted using an RNeasy Mini Kit (QIAGEN). cDNA was Datsenko, K.A., and Wanner, B.L. (2000). One-step inactivation of chromo-
synthesized using TaqMan reverse-transcription reagents (Applied Biosys- somal genes in Escherichia coli K-12 using PCR products. Proc. Natl. Acad.
tems) following the manufacturer’s instructions. Quantification of transcripts Sci. USA 97, 6640–6645.
was performed by real-time PCR using Fast SYBR Green Master Mix (Applied Eguchi, Y., Okada, T., Minagawa, S., Oshima, T., Mori, H., Yamamoto, K., Ish-
Biosystems) in a 7500 Sequence Detection System (Applied Biosystems). A list ihama, A., and Utsumi, R. (2004). Signal transduction cascade between EvgA/
of primers used for quantitative RT-PCR is presented in Extended Experi- EvgS and PhoP/PhoQ two-component systems of Escherichia coli. J. Bacter-
mental Procedures. See Extended Experimental Procedures for a detailed iol. 186, 3006–3014.
protocol.
Froschauer, E.M., Kolisek, M., Dieterich, F., Schweigel, M., and Schweyen,
R.J. (2004). Fluorescence measurements of free [Mg2+] by use of mag-fura 2
b-Galactosidase Assays in Salmonella enterica. FEMS Microbiol. Lett. 237, 49–55.
b-galactosidase activity was determined as described (Cromie et al., 2006). Garcia Vescovi, E., Soncini, F.C., and Groisman, E.A. (1996). Mg2+ as an extra-
cellular signal: environmental regulation of Salmonella virulence. Cell 84,
165–174.
SUPPLEMENTAL INFORMATION
Grundy, F.J., and Henkin, T.M. (2006). From ribosome to riboswitch: control of
gene expression in bacteria by RNA structural rearrangements. Crit. Rev. Bio-
Supplemental Information includes Extended Experimental Procedures, two
chem. Mol. Biol. 41, 329–338.
figures, and two tables and can be found with this article online at doi:10.
1016/j.cell.2010.07.046. Hammarlof, D.L., and Hughes, D. (2008). Mutants of the RNA-processing
enzyme RNase E reverse the extreme slow-growth phenotype caused by
a mutant translation factor EF-Tu. Mol. Microbiol. 70, 1194–1209.
ACKNOWLEDGMENTS Henkin, T.M. (2008). Riboswitch RNAs: using RNA to sense cellular metabo-
lism. Genes Dev. 22, 3383–3390.
We thank Laszlo Csonka, Kerry Hollands, Robert Landick, and Charles
Henkin, T.M., and Yanofsky, C. (2002). Regulation by transcription attenuation
Yanofsky for comments on the manuscript; Henry Huang for the plasmid
in bacteria: how RNA provides instructions for transcription termination/anti-
harboring supF; and John Roth for strains. This work was supported, in part,
termination decisions. Bioessays 24, 700–707.
by grant AI49561 from the NIH to E.A.G., who is an Investigator of the Howard
Hughes Medical Institute. Landick, R., and Yanofsky, C. (1987). Transcription attenuation. In Escherichia
coli and Salmonella Cellular and Molecular Biology, J.L. Ingraham, ed. (Wash-
Received: March 19, 2010 ington, DC: ASM Press).
Revised: May 24, 2010 Landick, R., Turnbough, C.L., and Yanofsky, C. (1996). Transcription attenua-
Accepted: July 14, 2010 tion. In Escherichia coli and Salmonella Cellular and Molecular Biology, F.C.
Published: September 2, 2010 Neidhardt, ed. (Washington, DC: ASM Press).

Cell 142, 737–748, September 3, 2010 ª2010 Elsevier Inc. 747


Laursen, B.S., Sorensen, H.P., Mortensen, K.K., and Sperling-Petersen, H.U. Shin, D., Lee, E.J., Huang, H., and Groisman, E.A. (2006). A positive feedback
(2005). Initiation of protein synthesis in bacteria. Microbiol. Mol. Biol. Rev. 69, loop promotes transcription surge that jump-starts Salmonella virulence
101–123. circuit. Science 314, 1607–1609.
Lejona, S., Aguirre, A., Cabeza, M.L., Garcia Vescovi, E., and Soncini, F.C. Snavely, M.D., Miller, C.G., and Maguire, M.E. (1991). The mgtB Mg2+ trans-
(2003). Molecular characterization of the Mg2+-responsive PhoP-PhoQ regu- port locus of Salmonella typhimurium encodes a P-type ATPase. J. Biol.
lon in Salmonella enterica. J. Bacteriol. 185, 6287–6294. Chem. 266, 815–823.
Loh, E., Dussurget, O., Gripenland, J., Vaitkevicius, K., Tiensuu, T., Mandin, P.,
Soncini, F.C., Garcia Vescovi, E., Solomon, F., and Groisman, E.A. (1996).
Repoila, F., Buchrieser, C., Cossart, P., and Johansson, J. (2009). A trans-
Molecular basis of the magnesium deprivation response in Salmonella typhi-
acting riboswitch controls expression of the virulence regulator PrfA in Listeria
murium: identification of PhoP-regulated genes. J. Bacteriol. 178, 5092–5099.
monocytogenes. Cell 139, 770–779.
Maguire, M.E. (1992). MgtA and MgtB: prokaryotic P-type ATPases that Spinelli, S.V., Pontel, L.B., Garcia Vescovi, E., and Soncini, F.C. (2008). Regu-
mediate Mg2+ influx. J. Bioenerg. Biomembr. 24, 319–328. lation of magnesium homeostasis in Salmonella: Mg2+ targets the mgtA tran-
script for degradation by RNase E. FEMS Microbiol. Lett. 280, 226–234.
Maguire, M.E. (2006). Magnesium transporters: properties, regulation and
structure. Front. Biosci. 11, 3149–3163. Sudarsan, N., Hammond, M.C., Block, K.F., Welz, R., Barrick, J.E., Roth, A.,
Merino, E., and Yanofsky, C. (2005). Transcription attenuation: a highly and Breaker, R.R. (2006). Tandem riboswitch architectures exhibit complex
conserved regulatory strategy used by bacteria. Trends Genet. 21, 260–264. gene control functions. Science 314, 300–304.
O’Connor, K., Fletcher, S.A., and Csonka, L.N. (2009). Increased expression of Turnbough, C.L., Jr., and Switzer, R.L. (2008). Regulation of pyrimidine biosyn-
Mg2+ transport proteins enhances the survival of Salmonella enterica at high thetic gene expression in bacteria: repression without repressors. Microbiol.
temperature. Proc. Natl. Acad. Sci. USA 106, 17522–17527. Mol. Biol. Rev. 72, 266–300.
Perez-Arellano, I., Rubio, V., and Cervera, J. (2005). Dissection of Escherichia
Winkler, W.C., and Breaker, R.R. (2005). Regulation of bacterial gene expres-
coli glutamate 5-kinase: functional impact of the deletion of the PUA domain.
sion by riboswitches. Annu. Rev. Microbiol. 59, 487–517.
FEBS Lett. 579, 6903–6908.
Prost, L.R., Daley, M.E., Le Sage, V., Bader, M.W., Le Moual, H., Klevit, R.E., Wortham, B.W., Patel, C.N., and Oliveira, M.A. (2007). Polyamines in bacteria:
and Miller, S.I. (2007). Activation of the bacterial sensor kinase PhoQ by acidic pleiotropic effects yet specific mechanisms. Adv. Exp. Med. Biol. 603,
pH. Mol. Cell 26, 165–174. 106–115.

Schiller, D., Kruse, D., Kneifel, H., Kramer, R., and Burkovski, A. (2000). Poly- Yanofsky, C., and Horn, V. (1994). Role of regulatory features of the trp operon
amine transport and role of potE in response to osmotic stress in Escherichia of Escherichia coli in mediating a response to a nutritional shift. J. Bacteriol.
coli. J. Bacteriol. 182, 6247–6249. 176, 6245–6254.

748 Cell 142, 737–748, September 3, 2010 ª2010 Elsevier Inc.


Structural Basis of Semaphorin-Plexin
Recognition and Viral Mimicry from
Sema7A and A39R Complexes with PlexinC1
Heli Liu,1 Z. Sean Juo,2,3,4 Ann Hye-Ryong Shim,1 Pamela J. Focia,1 Xiaoyan Chen,1 K. Christopher Garcia,2,3,4
and Xiaolin He1,*
1Department of Molecular Pharmacology and Biological Chemistry, Northwestern University Feinberg School of Medicine, Searle 8-417,

303 East Chicago Avenue, Chicago, IL 60611, USA


2Howard Hughes Medical Institute
3Department of Molecular and Cellular Physiology
4Department of Structural Biology

Stanford University School of Medicine, Beckman B171B, 279 Campus Drive, Stanford, CA 94305, USA
*Correspondence: x-he@northwestern.edu
DOI 10.1016/j.cell.2010.07.040

SUMMARY organogenesis, and immune responses (Kruger et al., 2005;


Suzuki et al., 2008). Semaphorins are also associated with tumor
Repulsive signaling by Semaphorins and Plexins is progression and other diseases (reviewed in Kruger et al., 2005;
crucial for the development and homeostasis of the Tamagnone and Comoglio, 2000). Within the neural system,
nervous, immune, and cardiovascular systems. Semaphorin-Plexin signaling is implicated well beyond axon
Sema7A acts as both an immune and a neural Sem- guidance, ranging from axon pruning to synaptic formation,
aphorin through PlexinC1, and A39R is a Sema7A specificity, and plasticity (reviewed in He et al., 2002; Waimey
and Cheng, 2006). While Plexins are the predominant receptors
mimic secreted by smallpox virus. We report the
for Semaphorins, the alternative Semaphorin receptors, Neuro-
structures of Sema7A and A39R complexed with pilin-1 and -2, differ from Plexins in structure and function
the Semaphorin-binding module of PlexinC1. Both and appear to serve as obligate coreceptors for either Sema-
structures show two PlexinC1 molecules symmetri- phorin signaling or VEGF signaling in a variety of specific cellular
cally bridged by Semaphorin dimers, in which the (neuronal, vascular, etc.) contexts (Pellet-Many et al., 2008;
Semaphorin and PlexinC1 b propellers interact in Uniewicz and Fernig, 2008).
an edge-on, orthogonal orientation. Both binding Semaphorins have been grouped into eight classes on the
interfaces are dominated by the insertion of the Sem- basis of primary sequence and source, where classes 3–7 are
aphorin’s 4c-4d loop into a deep groove in blade 3 of vertebrate Semaphorins and class V are viral Semaphorins
the PlexinC1 propeller. A39R appears to achieve (Semaphorin Nomenclature Committee, 1999). The Semaphor-
Sema7A mimicry by preserving key Plexin-binding ins are characterized by an N-terminal 500 amino acid (aa)
Sema domain that is essential for signaling through Plexins
determinants seen in the mammalian Sema7A
(Koppel et al., 1997). The crystal structures of the Sema domains
complex that have evolved to achieve higher affinity of Sema3A and Sema4D (also known as CD100) have been
binding to the host-derived PlexinC1. The complex determined, showing that the Sema domain is a seven-bladed
structures support a conserved Semaphorin-Plexin b propeller, and appear to exist as a homodimer by virtue of
recognition mode and suggest that Plexins are acti- conserved structural elements (Antipenko et al., 2003; Love
vated by dimerization. et al., 2003). There are four subfamilies of vertebrate Plexins
(A, B, C, and D), which are all type I transmembrane glycopro-
INTRODUCTION teins featuring an extracellular segment containing an N-terminal
Sema domain, followed by variable numbers of PSI (‘‘found in
Semaphorins and their receptors, Plexins, are two families of Plexins, Semaphorins and Integrins’’) domains, immunoglob-
widely expressed proteins whose respective structures and ulin-like (Ig) domains (Bork et al., 1999), and an intracellular
functions are conserved across the animal kingdom. Originally GTPase-activating (GAP) domain that regulates Rho family
identified as ligand-receptor pairs that control axon guidance GTPases (Oinuma et al., 2004). The structures of the intracellular
by repulsion during central nervous system (CNS) development domains of two Plexins have been solved (He et al., 2009; Tong
(Kolodkin et al., 1993; Luo et al., 1993; Tamagnone et al., et al., 2009), but three-dimensional structural information does
1999; Tessier-Lavigne and Goodman, 1996), Semaphorins and not exist for the Plexin extracellular segment or for the Sema-
Plexins have been shown to serve as path-finding controls for phorin-Plexin interaction.
a diverse array of additional functions in physiology, including In the immune system, a subset of Semaphorins are active
vascularization and angiogenesis (normal and pathological), and are designated as ‘‘immune Semaphorins.’’ These include

Cell 142, 749–761, September 3, 2010 ª2010 Elsevier Inc. 749


Sema4D, Sema4A, and Sema7A, which appear to modulate (Figures 1B–1E). In HEPES-buffered saline (HBS), Sema7A
a variety of immune responses ranging from thymic selection bound PlexinC1-SemaPSI with a KD of 290 nM (Figure 1B).
to B cell homing (Suzuki et al., 2008). Sema7A is a GPI-anchored The ITC measurements for Sema7A/PlexinC1 were complicated
cell-surface glycoprotein expressed on activated lymphocytes by the small enthalpy change ( 2 kcal/mol), which resulted in
and thymocytes (Xu et al., 1998; Yamada et al., 1999), and its shallow titration curves with poor signal-to-noise ratio. There-
receptor is PlexinC1 (also named VESPR or CD232) (Tamagnone fore, we used high concentrations of the samples and injected
et al., 1999). The Sema7A/PlexinC1 interaction was originally large amounts of protein so as to maximize the heat per injection,
shown to induce the activation of monocytes (Holmes et al., and also to fully saturate the titration curve. The Sema7A/Plex-
2002), but recently this receptor-ligand pair has been shown to inC1 KD measured here by calorimetry using soluble proteins,
regulate melanocyte adhesion, and silencing of PlexinC1 is albeit in the nanomolar range, is weaker than the reported KD
seen during the development and progression of melanoma of 2.1nM from Scatchard analysis using intact receptors in the
(Scott et al., 2009). Sema7A also has been suggested to have cell membrane (Tamagnone et al., 1999). Previously reported
neuronal functions, by promoting axon growth through integrins Semaphorin/Plexin affinity measurements are primarily cell
(Pasterkamp et al., 2003). Underscoring the importance of Sem- based and generally in the low nanomolar KD range. We attribute
aphorin/Plexin interactions in the immune system, viruses have the lower-affinity values of the soluble recombinant proteins to
evolved proteins that engage the immune Semaphorin/Plexin the lack of membrane confinement in the ITC format, not to
system, presumably to enhance virus survival in the host. The structural differences in the recombinant proteins. Similar differ-
most well-characterized examples derive from Vaccinia ences between solution-based and cell-based affinities have
(smallpox) virus and Alcelaphine herpesvirus, which encode been seen for ligand binding to other receptors, such as SCF/
secreted Semaphorin homologs A39R and AHVsema (Comeau KIT (55 nM versus 2 nM) (Lemmon et al., 1997; Lev et al.,
et al., 1998; Kolodkin et al., 1993). These viral Semaphorins 1992), CNP/NPRB (2.7 nM versus 0.03 nM) (He et al., 2006; Kol-
show sequence similarity to Sema7A and can also bind to and ler and Goeddel, 1992), and MHC/TCR (micromolar versus nano-
activate PlexinC1 (Comeau et al., 1998). The viral Semaphorins molar) (Huppa et al., 2010; Krogsgaard et al., 2003). We also
probably play an immunomodulatory role by mimicking Sema7A; determined that the affinity of Sema7A for the truncated Plex-
an antibody against PlexinC1 inhibits A39R-induced induction of inC1-SemaPSI and full-length PlexinC1 ECD were nearly iden-
inflammatory cytokines by monocytes (Comeau et al., 1998). tical (Figures 1B and 1C), indicating the Ig and PSI domains C-
We present here the crystal structures of both the Sema7A/ terminal to the PlexinC1-SemaPSI module are not significantly
PlexinC1 complex and the A39R/PlexinC1 complex, revealing involved in Semaphorin recognition. Thus, for structural studies
the basic architecture of a Semaphorin/Plexin recognition com- we proceeded with crystallization of the Sema7A/PlexinC1-
plex. Our data point to a conserved mode of recognition of SemaPSI complex.
Plexins by Semaphorins and provide insights into Plexin activa- The viral-derived A39R bound PlexinC1-SemaPSI with a KD of
tion and viral mimicry. 9.4 nM (Figure 1D). The large enthalpy change ( 14.6 kcal/mol)
facilitated the obtainment of high-quality binding curves. We also
RESULTS characterized A39R for the effect of various PlexinC1 ECD trun-
cations on affinity. A39R affinity and thermodynamic parameters
Biochemical Analysis of PlexinC1 Binding for full-length PlexinC1-ECD (KD 8.9 nM) were nearly identical
by the Mammalian Semaphorin Sema7A to that of A39R binding to PlexinC1-SemaPSI (Figures 1D and
and the Viral Semaphorin A39R 1E). Thus, similar to Sema7A/PlexinC1 binding, this indicated
The extracellular domains of Semaphorins and Plexins are large that the N-terminal Sema-PSI domains of PlexinC1 were suffi-
and highly glycosylated; thus we expressed recombinant forms cient for Semaphorin binding and represented an appropriate
of PlexinC1, Sema7A and A39R using baculovirus-mediated form for cocrystallization of both the Sema7A/PlexinC1 and
mammalian cell gene transduction (BacMam) (Dukkipati et al., A39R/PlexinC1 complexes.
2008) and purified the secreted proteins from HEK293H cells
or N-acetylglucosaminyltransferase I-deficient HEK293 (GnTI- Structures of the Sema7A/PlexinC1 Complex, Free
HEK293) cells (Reeves et al., 2002). Our construct designs A39R, and the A39R/PlexinC1 Complex
were informed by prior structural analysis of Sema4D (Love To obtain diffraction quality crystals of the heavily glycosylated
et al., 2003), which showed that the first membrane-distal PSI Sema7A/PlexinC1-SemaPSI complex (four N-linked glycosy-
domain is integrally associated with the Sema domain. Deletion lated sites on Sema7A and seven on PlexinC1-SemaPSI), we
of the PSI domains in either PlexinC1 or Sema7A destabilized the used Endo-H to trim the N-linked glycans attached to the GnTI
proteins. Ultimately, after testing a variety of constructs, for HEK293-expressed Sema7A and PlexinC1-SemaPSI proteins.
PlexinC1, we expressed the Sema domain plus the first PSI This treatment leaves a single N-acetylglucosamine (GlcNAc)
domain (SemaPSI), and the entire extracellular segment (ECD). residue remaining at each occupied N-linked glycan site,
For Sema7A, we expressed the entire extracellular segment preserving the core glycosylation of the proteins. The glycan-
excluding the GPI anchor; for A39R, we expressed the full-length trimmed proteins exhibited the same solution behavior to wild-
protein, which is a ‘‘minimal’’ secreted Semaphorin that does not type proteins, e.g., the Semaphorins existed exclusively as
contain a PSI domain (Figure 1A). dimers after glycan-trimming, as assessed by gel filtration chro-
To analyze the solution binding of PlexinC1 to its A39R and matography (Figure S1 available online). The Sema7A/PlexinC1-
Sema7A ligands, we used isothermal titration calorimetry (ITC) SemaPSI complex structure was determined to a resolution of

750 Cell 142, 749–761, September 3, 2010 ª2010 Elsevier Inc.


Figure 1. Characterization of the Binding of Sema7A and A39R to PlexinC1
(A) Domain diagram of PlexinC1, Sema7A, and A39R and the constructs used in characterization. The cartoon key for the diagrams is shown to the right.
(B–E) Profiles of the binding of PlexinC1 to Sema7A and A39R as measured by isothermal titration calorimetry. The binding parameters are indicated below the
traces.
See also Figure S1 for gel filtration profiles showing that Sema7A and A39R exist as dimers.

2.4 Å via the method of single isomorphous replacement with propellers are 50 Å for Sema7A-Sema7A, 55 Å for Sema7A-Plex-
anomalous scattering (SIRAS) (Table S1, Figure S2). The asym- inC1, and 80 Å for PlexinC1-PlexinC1.
metric unit of the crystal contains one dimeric complex in a 2:2 Interestingly, the Sema domains of Sema7A and PlexinC1
stoichiometry, comprised of a central Sema7A dimer and two interact ‘‘edge-on’’ using their sides to contact one another,
monomeric PlexinC1-SemaPSI molecules (Figure 2A). rather than the top or bottom faces, as was generally expected.
Overall, the shape of the complex resembles a crab where The planes of the Sema7A and PlexinC1 b propellers are orthog-
Sema7A comprises the body, and the two PlexinC1 molecules onally related. The orientation shown in Figure 2 places the
resemble pincers emanating radially from the body at four and C-termini of Sema7A close to the cell membrane, leading to
eight o’clock. The approximate dimensions of the complex are the GPI anchor. The C terminus of the PSI domain of PlexinC1,
80 Å 3 140 Å 3 160 Å (Figure 2A). The head-to-head docking which is 160 Å from the Semaphorin C terminus, leads to the
architecture of the complex is in an ideal orientation for interac- opposing cell membrane, albeit through one additional PSI and
tion of two cell surface-associated proteins across a cell-cell four Ig domains not present in the current structure. Because
junction. In the complex, both Sema7A and PlexinC1 contain the remaining PSI and Ig domains of PlexinC1 would appear to
large, disk-shaped Sema domains composed of seven-bladed emanate away from the central dyad axis of the complex, they
b propellers, each of which is intimately associated with its are unlikely to contact Sema7A.
respective PSI domain and, in the case of Sema7A, a single Ig We expressed A39R in baculovirus and determined the
domain that would lead to the membrane. In the complex, the unliganded A39R structure, to a resolution of 2.0 Å, by SIRAS
Sema domains mediate the vast majority of the receptor-ligand (Table S1). The structure of A39R shows a dimer of minimal
contact. The center-to-center distances between the Sema Sema domains with no PSI or Ig domains (Figure 2D). The

Cell 142, 749–761, September 3, 2010 ª2010 Elsevier Inc. 751


Figure 2. Structures of the Sema7A/PlexinC1-SemaPSI and A39R/PlexinC1-SemaPSI Complexes
(A) Ribbon models of the Sema7A/PlexinC1-SemaPSI complex in front view (left) and side view (right), with the Sema7A protomers colored in cyan and blue, and
the PlexinC1-SemaPSI protomers in pink and magenta. The N-linked glycans are depicted as sticks with carbon atoms colored in green. A cartoon of a membrane
is drawn above and below the complex to indicate where the respective proteins would be attached to the cell surfaces.
(B) The structure of an individual PlexinC1-SemaPSI molecule from the complex in two orthogonal views, with each of the seven b -propeller blades, the extrusion,
the flap, and the PSI domain individually colored.
(C) Ribbon model of the A39R/PlexinC1-Sema-PSI complex in front view (left) and side view (right), with the A39R protomers colored in yellow and wheat and the
other components colored similarly to (A).
(D) Ribbon model of an A39R protomer from the free A39R dimer, with the structural modules colored in the same format as PlexinC1 shown in (B).
See also Table S1 and Figure S2 for crystallographic statistics and structural comparisons.

Sema domains of A39R and Sema7A are clearly built on the domains mediate all the A39R-PlexinC1 contact. The orienta-
same scaffold, with a root-mean-square deviation (rmsd) of tions of the Sema propellers in the complex are nearly identical
1.5 Å for matching Ca atoms (Figure S3). Major structural differ- to those in Sema7A/PlexinC1, as is the ‘‘edge-on’’ interaction
ences are located only at the N-terminal segment and several mode between the ligand and receptor b propellers.
loops remote from the Plexin-binding site of Sema7A. Sema7A,
like Sema3A and Sema4D, has a long N-terminal segment that Dimerization of Sema7A and A39R Are Mediated by
provides an additional, fifth b strand on the outer edge of blade Conserved Structural Elements with Varied Chemistry
6, whereas A39R lacks this segment (Figure S3). Dimerization appears to be a general and probably important
We followed the same approach as Sema7A/PlexinC1- property of Semaphorins (Antipenko et al., 2003; Love et al.,
SemaPSI to obtain the crystals of the A39R/PlexinC1-SemaPSI 2003) that we can now examine within the context of a receptor
complex, which we solved by molecular replacement (Table complex. The Sema7A molecules in the complex, consisting of
S1). The architecture of the A39R/PlexinC1 complex is similar Sema, PSI, and Ig domains, form a dimer roughly similar to the
to that of Sema7A/PlexinC1, except that as a result of the lack Sema4D dimer (Love et al., 2003) and the Sema3A dimers that
of PSI and Ig domains in A39R, it is shorter in its longest dimen- only contain the Sema domains (Antipenko et al., 2003)
sion (Figure 2C). As in the Sema7A/PlexinC1 complex, the Sema (Figure S2C). This is consistent with our gel filtration analysis

752 Cell 142, 749–761, September 3, 2010 ª2010 Elsevier Inc.


(Figure S1). The Sema7A dimer interface buries 2860 Å2 of Plexins is not a central part of the interface. It appears, then,
solvent-accessible surface area. Two sets of three loops on that Plexins have lost the prominence of this structural element
the top face of the Sema domain, 4b-4c, 4d-5a, and 5b-5c are as the relative functions of the Sema domain in Semaphorins
intertwined at the dimer interface. In comparison, both Sema4D versus Plexins specialized over the course of evolution
and Sema3A use an additional loop for dimerization (Figure S2E) (Antipenko et al., 2003; Love et al., 2003; Stamos
(Figure S2C). The Sema7A Sema-Sema interface is primarily et al., 2004).
hydrophobic. The Ig domain of Sema7A is also involved in the The PSI domain of PlexinC1 is a small cysteine-rich domain
dimer interface but only contributes 15% to the total buried similar to that of Semaphorins and integrins, but its orientation
surface area. The Ig domain of Sema7A, unlike the Sema4D Ig relative to the Sema domain is different from that in Semaphorins
domain, which is canonical, is an unusual variation of the Ig by an 20 rotation (Figure S2E). The PSI domain is intimately
fold, with a 5-on-2 topology in which strand D switches from packed against the Sema domain by a broad array of primarily
the BED b sheet to join the AFGC b sheet (Figure S2D). hydrophobic interactions, which would facilitate the sensitive
The viral Semaphorin A39R also exists as a dimer both in solu- structural transmission of Sema binding from the membrane-
tion and in the crystal (Figure S1). Dimerization of A39R is exclu- distal to the membrane-proximal regions of PlexinC1.
sively mediated by its Sema domain, since this protein does not
have PSI or Ig domains, and the dimer interface is considerably The Sema7A-PlexinC1 Interaction Features
smaller (1990 Å2) than that seen on the canonical Semaphorins a ‘‘Loop-in-Groove’’ Recognition Mode
(Figure S2C). A39R uses the same set of loops as the Sema7A The edge-on, orthogonal stacking of the respective b propellers
Sema for dimerization, but in contrast to the hydrophobic in the Sema7A-PlexinC1 interface (Figure 3A) buries a total of
Sema7A dimer interface, the A39R dimer interface is composed 2100 Å2 solvent-accessible surface area. The extensive inter-
almost exclusively of hydrophilic interactions involving six salt face can be divided into three principal regions: (1) The 4c-4d
bridges. loop of Sema7A inserting into the groove on PlexinC1 that is
The Sema7A and A39R structures, together with the previous bounded by walls composed of the PlexinC1 loop 3b-3c and
Sema3A and Sema4D structures, indicate that although the the bulged strand 3d—this PlexinC1 groove has an open and
dimer interfaces of Semaphorins can vary significantly in a closed (obstructed) end (Figure 3B); (2) the extrusion helix 2
sequence and chemical nature, the general structural mode of of Sema7A contacting the loop 3b-3c of PlexinC1 at one side
domain dimerization is conserved, and this is likely to be impor- of the groove; and (3) a small area of contact between the
tant for the appropriate dimerization geometry of the bound Sema7A blade 3, and the PlexinC1 strand 3d, that forms the
Plexin receptors for signaling. The preservation of the dimeriza- opposing wall of the groove (Figures 3C–3E).
tion geometry despite vastly different interface chemistries The central ‘‘loop-in-groove’’ interaction (Figure 3D) features
suggests that there is evolutionary pressure to achieve this ten hydrogen bonds (all hydrogen bonds discussed are pre-
dimerization mode for Semaphorin function, presumably to dicted from geometry) between the protruding 4c-4d loop of
orient bound Plexins for signaling. Sema7A and the PlexinC1 groove (Table S2). The polar interac-
tions are supplemented by two hydrophobic residues (Leu276
The Structure of PlexinC1-SemaPSI Has Unique and Val278) in the Sema7A 4c-4d loop, which contact several
Features hydrophobic residues in the PlexinC1 groove through van der
Given that the extracellular segments of Plexins have not been Waals interactions. Overall, the PlexinC1/Sema7A loop-in-
structurally characterized, the fold of the PlexinC1 Sema-PSI groove interaction includes a mixture of hydrophobic residues
domains merits some description. The Sema domain of PlexinC1 lining the groove wall, interspersed with hydrophilic residues,
in the complex is generally similar to other seven-bladed to presumably provide ligand specificity through polar interac-
b propeller domains in topology but has unique features that tions. Importantly, at the edge of this groove in PlexinC1,
are distinct from Semaphorins or MET (Stamos et al., 2004) Sema7A Lys280 forms a salt bridge with PlexinC1 Asp200, and
(Figure 2B). After the common nomenclature used by other this interaction appears to be a key component of A39R mimicry
propeller proteins, blade 7 of the PlexinC1 Sema domain is (discussed below).
C-terminally adjacent to blade 1; each of the seven blades is At the obstructed (closed) end of the groove (Figure 3B), the
formed by four antiparallel b strands with strands a-d from the Sema7A extrusion helix 2 interacts in a roughly parallel manner
inside to the outside of the b propeller (Figure 2B). The surface with the PlexinC1 3b-3c loop, also showing several ancillary
bears the loops linking strands b and c (e.g., loop 4b-4c) and link- long-range interactions with the tips of the PlexinC1 2b-2c and
ing strands d and a (e.g., loop 5d-6a) on the top face. Loop 1d-2a 2d-3a loops (Figure 3C). The interaction is intimate only at the
traverses the top of the propeller like a flap, sealing the central PlexinC1 Ala197-Ala198-Ser199 bulge. The obstructed end of
channel of the propeller, which is hollow in Semaphorins, MET, the PlexinC1 groove is largely due to the steric bulk sidechain
and integrins (Figure S2E) (Antipenko et al., 2003; Love et al., of Arg131, which forms a salt bridge with Sema7A Glu376
2003; Stamos et al., 2004; Xiao et al., 2004; Xiong et al., 2002). (Figure 3C).
A long insertion, termed the ‘‘extrusion’’ (Love et al., 2003), is In the third region of the interface, at the open end of the Plex-
located between strands 5c and 5d. A major structural difference inC1 groove (Figure 3B), the outer edge of Sema7A blade 3 forms
is that the extrusion of PlexinC1 is shorter than that of Semaphor- extended interactions with the bulged strand 3d of PlexinC1
ins. The structure of the complex shows that this extrusion in (Figure 3E). There are two spatially separate clusters of salt
Semaphorins is critical for binding Plexins, but the extrusion in bridges in this region of the interface. The first cluster involves

Cell 142, 749–761, September 3, 2010 ª2010 Elsevier Inc. 753


Figure 3. The Interface between Sema7A and PlexinC1-SemaPSI
(A) The overall structure of an interacting pair of Sema7A and PlexinC1-SemaPSI protomers, highlighting the structural elements involved in their binding, colored
cyan for Sema7A and pink for PlexinC1-SemaPSI.
(B) Close-up view of the interface with Sema7A depicted as ribbons and PlexinC1-SemaPSI in a surface representation, showing how the protruding loop 4c-4d
of Sema7A inserts into a groove in PlexinC1 blade 3, and is flanked by the contacts between the Sema7A extrusion helix 2 and the PlexinC1 loop 3b-3c, and the
contacts between the blade 3 of Sema7A and the bulged strand 3d of PlexinC1.
(C) The interactions of residues near the obstructed end of the PlexinC1 groove, with Sema7A in chocolate and PlexinC1 in pink.
(D) The interaction between residues of the 4c-4d loop of Sema7A (cyan) and the PlexinC1 groove (pink).
(E) The interaction between the residues of blade 3 of Sema7A (green) and strand 3d of PlexinC1 (pink). Note that residues 218–220 of PlexinC1 comprise a bulge
from strand 3d.
See also Table S2 and Figure S4 for a list of the interactions and for mutagenesis/binding data on this interface, respectively.

Sema7A residues Arg204 and Arg202 both forming salt bridges binding (Figure S4). In the complex structure, the location of the
with PlexinC1 residue Glu219. Sema7A Tyr213 occupies the ridges flanking the groove in the Semaphorin binding site is
space between this salt bridge and a salt bridge involving the consistent with a previous mutagenesis study implicating
important Sema7A residue Lys280 and PlexinC1 Asp200 (Fig- the region between residues 166–235 of the Sema domain of
ure S2A). The second cluster of charged interactions involve Sema3A in its Plexin-binding specificity (Koppel et al., 1997). In
Sema7A Asp216, which forms bifurcated salt bridges with summary, the Sema7A/PlexinC1 interface is extensive and varied
Arg222 and Lys224 of PlexinC1. Mutagenesis data support that in chemical and structural character and is dominated by the
the interactions at this region are important for Sema7A-PlexinC1 insertion of a long loop in Sema7A into a deep groove in PlexinC1.

754 Cell 142, 749–761, September 3, 2010 ª2010 Elsevier Inc.


Figure 4. The Interface between A39R and PlexinC1-SemaPSI
(A) Structural superposition of the A39R/PlexinC1-SemaPSI and Sema7A/PlexinC1-SemaPSI complexes indicating the closely related docking modes of the viral
and mammalian Semaphorins to the PlexinC1 receptor. The complexes have been aligned on the PlexinC1 component in order to visualize the respective overlap
of the two different Semaphorins. The A39R is in yellow, Sema7A in cyan.
(B) Close-up view of the interface with A39R depicted as ribbons and PlexinC1-SemaPSI in a surface representation.
(C) The interactions of residues near the obstructed end of the PlexinC1 groove, with A39R in chocolate and PlexinC1-SemaPSI in pink.
(D) The interaction between residues of the 4c-4d loop of A39R (blue) and the PlexinC1 groove (pink).
(E) The interaction between the residues of blade 3 of A39R (green) and strand 3d of PlexinC1-SemaPSI (pink).
See also Table S3 for a list of the interactions.

The A39R-PlexinC1 Interaction Globally Resembles accommodations at the periphery of the interface (Figure 4A).
the Sema7A-PlexinC1 Interaction These small movements result in remodeled pairwise interac-
With strikingly similar orientations of the respective b propellers tions by these surrounding structural elements, relative to the
in the two complexes, the A39R-PlexinC1 interface involves Sema7A/PlexinC1 interface, but still preserving several key
the same set of structural elements as the Sema7A-PlexinC1 contacts that presumably are important for the cross-reactivity
interface (Figure 4). The A39R-PlexinC1 interface buries a (discussed below).
total of 1890 Å2 solvent-accessible surface area, slightly smaller The ‘‘loop-in-groove’’ interaction in A39R/PlexinC1 (Fig-
than the Sema7A-PlexinC1 interface. The protruding loop 4c-4d ure 4D) has six hydrogen bonds between the A39R 4c-4d loop
of A39R is in an almost identical conformation as that of Sema7A, and the PlexinC1 groove, four less than in Sema7A/PlexinC1
inserting into the groove of the blade 3 surface of PlexinC1 (Table S3). Only one of these hydrogen bonds is conserved
(Figures 4A, 4B, and 5). The neighboring segments of this loop, between Sema7A/PlexinC1 and A39R/PlexinC1. The diversity
including the extrusion helix 2 of A39R that contacts the loop of amino acid contacts with PlexinC1 formed by the Sema7A
3b-3c of PlexinC1, and the A39R blade 3 that contacts the Plex- versus A39R 4c-4d loop indicates that the PlexinC1 pocket
inC1 strand 3d, have undergone some relatively minor structural has the capacity for highly degenerate interactions, which

Cell 142, 749–761, September 3, 2010 ª2010 Elsevier Inc. 755


Figure 5. The Conformation of the 4c-4d Loop of Semaphorins Is Central for Plexin Recognition
(A–D) Stick models of the isolated 4c-4d loops of Sema7A (cyan), A39R (yellow), Sema4D (green), and Sema3A (pink), showing its conserved conformation at the
bases (top) and the midpoints of the loops, and variable conformation at the tips of the loops (bottom).
(E) Sequence comparison of the 4c-4d loop of Semaphorins, with the key conserved structural determinants highlighted.
See also Figure S5 for the RGD motif at the base of the Sema7A 4c-4d loop.

facilitates cross-reactivity. At the edge of this groove in PlexinC1, PlexinC1 residues as in Sema7A/PlexinC1, but the pattern
A39R presents an arginine (Arg207), as opposed to a lysine of interaction is altered. While there are four salt bridges in
(Lys280) in Sema7A, to form a salt bridge with PlexinC1 two clusters in Sema7A/PlexinC1, there are only two (A39R
Asp200 (discussed below). Arg132/Arg134 to PlexinC1 Glu219) in A39R/PlexinC1 at this
Peripheral to the 4c-4d loop, the structural chemistry of the region (Figure 4E). In A39R/Sema7A, the loss of the other cluster
A39R/PlexinC1 interactions at the obstructed (closed) end of of salt bridges seen in Sema7A/PlexinC1 (PlexinC1 Arg222/
the PlexinC1 groove (Figures 4B and 4C) are quite different Lys224 to Sema7A Asp216) is due to the raised A39R blade 3.
than what is seen for Sema7A/PlexinC1 as a result of a slight While PlexinC1 Lys224 is not directly bonded to A39R in the
rigid-body repositioning (Figure 4A). Compared to Sema7A, the A39R/PlexinC1 complex, PlexinC1 Arg222 manages to form
A39R extrusion helix 2 is shifted outwards relative to the center hydrogen bonds with the hydroxyl of A39R Tyr145. The loss
of the interface and tilted away from PlexinC1 (Figures 4A of these two salt bridges from the mammalian complex may
and 4C). Consequently, the A39R/PlexinC1 interaction is not as be further compensated by the strengthening of the A39R
intimate as the Sema7A/PlexinC1 interaction at this region. Arg132-PlexinC1 Glu219 salt bridge, which is more deeply
However, the shifting of the helix also allows A39R Asp300, buried than in Sema7A/PlexinC1, because of the neighboring
corresponding to Sema7A Gln379 (Figures 3C and 4C), to hydrophobic interaction between A39R Ile125 and PlexinC1
move into the position to form a salt bridge with PlexinC1 Leu220 (Figure 4E). Collectively, The A39R/PlexinC1 interface
Arg131, which is not present in Sema7A/PlexinC1. shows variations from the Sema7A/PlexinC1 interface due to
The A39R/PlexinC1 interactions at the open end of the some intermolecular repositioning around the central 4c-4d
PlexinC1 groove (Figures 4B and 4E) involve the same set of loop. Nevertheless, the general scheme of inserting the long

756 Cell 142, 749–761, September 3, 2010 ª2010 Elsevier Inc.


4c-4d loop into the blade 3 groove in PlexinC1 is identical for both Sema7A and A39R 4c-4d loops (Figure 5) ensures similar
Sema7A and A39R. loop conformations presented to PlexinC1. There is mimicry of
an array of interactions peripheral to the tip of the 4c-4d loop,
as is clear from comparison of the respective binding surfaces.
The Similarities and Variations of the Central
For example, the Lys280 in Sema7A that salt bridges to
Recognition Loops of Semaphorins
Asp200 in PlexinC1 is mimicked by Arg207 in A39R that also
The structural features of the 4c-4d interaction loop of Sema7A
salt bridges to Asp200 on PlexinC1. Also found in both Sema-
and A39R are similar in all Semaphorin structures determined
phorins is a cluster of salt bridges on blade 3 that interact with
to date (Figure 5). The conformation of this loop is stabilized
residues on strand d of PlexinC1. Here, Arg202 of Sema7A salt
by an extensive intraloop hydrogen-bonding network, which
bridges with Glu219 in PlexinC1, and this interaction is mimicked
involves residues highly conserved in most Semaphorins (Fig-
by Arg134 of A39R salt bridging to PlexinC1 Glu219. Arg202
ures 5A–5E). At the base of the loop, Asp269 (Sema7A
and Asp216 in Sema7A are spatially mimicked by Arg132 and
numbering), a buried aspartate conserved in all Semaphorins
Asp148 in A39R, but the hydrogen bonding network with the
(highlighted in Figures 5A–5E), forms a network of four hydrogen
same PlexinC1 residues is remodeled. Key to this cluster is
bonds with main chain amides, which play a central role in orga-
a Tyrosine residue (Tyr213 in Sema7A, Tyr145 in A39R) (Fig-
nizing the loop conformation. At the midpoint, a serine (274 in
ure 6F) that is in a nearly identical position in both complexes,
Sema7A and 201 in A39R) hydrogen bonds with two main-chain
in the center of the cluster of salt bridges.
amides on the opposing strand to narrow the loop. The kinking
In order to ask whether the residues A39R uses to ‘‘mimic’’
of the loop is also facilitated by the presence of two consecutive
Sema7A were energetically important for the viral Semaphorin
glycines (Gly271-Gly272) conserved in all Semaphorins (Fig-
binding, we mutated several of them and tested binding by ITC
ure 5E). At the tip of the loop, the Sema7A Ser274 main-chain
(Figure 6). Mutations of A39R Arg132 or Tyr145 (corresponding
carbonyl forms hydrogen bonds with the main-chain amides
to Sema7A Arg202 and Tyr213) to glutamate or serine abolished
of Ser277 and Val278, a pattern reminiscent of reverse-turns,
PlexinC1 binding (Figures 6C and 6D), whereas mutation of A39R
which may help to maintain a rigid and protruding conformation.
Arg207 (corresponding to Sema7A Lys280) to glutamate
Similar conformation is observed for the tip of this A39R loop
reduced PlexinC1 binding by >60-fold (Figure 6E). The A39R
(Figures 5A and 5B).
Arg207 has a stronger charge and a larger head group than
In Sema3A and Sema4D, however, the tip of this loop has
a Lys residue, and its aliphatic stem is kinked, contacting the
a variation from Sema7A/A39R (Figures 5C and 5D). Class 3–5
neighboring hydrophobic moieties more intimately. Arg207 is a
Semaphorins, including Sema3A and Sema4D, have a one-
more complementary fit in the A39R/PlexinC1 interface com-
residue deletion at the Ser274 position. Sema4D and Sema3A
pared to the Lys280 in the Sema7A-PlexinC1 interface, where
also lack the main-chain hydrogen bonds at the tip seen in
there is more solvent-occupied space adjacent to the side chain
Sema7A and A39R. Consequently, the tips of the 4c-4d loops
(Figures S2A and S2B). Our data (Figure 6E) suggest that the
of these Semaphorins can adopt different main chain conforma-
difference at this position could be an important contributing
tions, probably reflecting the structural requirements of their
factor to the higher-affinity binding to PlexinC1 achieved by
specific Semaphorin-Plexin interactions, yet all appear to
A39R, although additional interactions no doubt also contribute.
contain a polar residue at the loop tips for hydrogen bonding.
We also mutated PlexinC1 Arg222, which is hydrogen bonded to
Because the residues that reside at the apex of the loop are
A39R Tyr145 and serves as shared contact with both Semaphor-
the most deeply embedded in the base of the Plexin groove,
ins. We found a 9-fold affinity loss in A39R-PlexinC1 binding
as exemplified by Sema7A/PlexinC1, it is likely that these are
(Figure 6G). While Sema7A and A39R have exactly the same resi-
major determinants for Semaphorin-Plexin binding specificity
dues at the local region contacting PlexinC1, PlexinC1 Arg222
between classes. Indeed, there is extensive sequence diver-
adopts very different rotamers in the two complexes (Figure 6F).
gence between different classes of Semaphorins at these
The 9-fold affinity loss upon PlexinC1 Arg222Ser mutation
positions, but few within the same class of Semaphorins
suggests that this is an important determinant in A39R/PlexinC1
(Figure 5E).
binding (Figure 6G). Therefore, the residues we mutated, which
are strikingly coincident in the viral and mammalian Sema
The Mimicry of the Mammalian Semaphorin Sema7A complexes, while certainly not giving us a comprehensive ener-
by the Viral Semaphorin A39R getic map of the A39R/PlexinC1 interface landscape, are indeed
The mammalian and viral Semaphorins have arrived at nearly energetically important in ligand-receptor binding.
identical binding modes despite substantial variations in Collectively, our structures suggest that A39R, built on a
sequence (31% identity between Sema7A and A39R). Visualiza- smaller scaffold, has used a limited set of key, highly coincident
tion of the PlexinC1 binding surfaces of Sema7A and A39R amino acid contacts to evolve a more efficient binding interac-
(Figures 6A and 6B) indicates that there are several key corre- tion with PlexinC1 through both the enhancement of particularly
sponding regions of structural mimicry that likely serve as the important polar interactions, such as the substitution of A39R
foundation for their cross-reactivity with a common receptor. Arg207 for the Sema7A Lys280, and slight adjustments of side
There is obvious structural conservation of the centrally located rotamer and main chain positions throughout the binding
4c-4d loop and the presence of identical residues at the tips of surface. These adjustments sum to a substantial optimiza-
the loop (Ser-Leu) that engage the deepest portion of the tion of binding energetics and a concomitant gain in binding
PlexinC1 groove. The preservation of the di-Glycine pair in affinity.

Cell 142, 749–761, September 3, 2010 ª2010 Elsevier Inc. 757


Figure 6. Viral Mimicry of Sema7A by A39R
(A and B) The respective PlexinC1-binding surfaces of Sema7A (cyan, left) and A39R (yellow, right). The residues mapped at the interface are colored red for acidic
residues, blue for basic residues, gray for polar, noncharged residues, and green for apolar residues.
(C–E) Mutations of the A39R residues mapped to the Sema7A-PlexinC1 interface abolished or dramatically reduced A39R-PlexinC1 binding.
(F) The different conformation of PlexinC1 Arg222 in binding A39R and Sema7A.
(G) Calorimetric measurement of the binding between A39R and the Arg222Ser mutant of PlexinC1-SemaPSI.
See also Figure S3 for a structural and sequence comparison of Sema7A and A39R.

758 Cell 142, 749–761, September 3, 2010 ª2010 Elsevier Inc.


DISCUSSION the knowledge that Plexins are active as dimers should help
refine our thinking about possible downstream effectors. The
We present here the structures of two Semaphorin-Plexin quiescent PlexinC1 possibly exists as preformed oligomers,
complexes, establishing the general recognition paradigm given the evidence that some other Plexins, e.g., plexin B, exist
between two protein families with important and wide-ranging as stable homodimers in the absence of ligands (Tong et al.,
physiological functions involving deliverance of repulsive guid- 2007). We have also found that unliganded PlexinC1 ECD exists
ance cues. Unexpectedly, the two b propellers of the Sema- as a mixture of oligomeric states, as assessed by gel filtration
phorin and Plexin Sema domains contact each other edge-on. chromatography and crosslinking, although we have not quanti-
In comparison, MET uses the bottom face of the propeller for tated the exact stochiometries of these states. We propose that
HGF-b chain interaction (Stamos et al., 2004), whereas integrins constitutively dimeric ligands such as Sema7A and A39R orient
bind their ligands with the top face of the propeller (Xiao et al., the inactive PlexinC1 into a dimerized state, in which the
2004; Xiong et al., 2002). Thus, as for other recognition modules distance, geometry and conformation enables the activation of
in biology, such as leucine-rich repeats, immunoglobulin folds, the intracellular GAP activity. This PlexinC1 activation scheme
and even MHC-folds, the b propeller reveals itself to be capable is simpler than a previously proposed model for Sema3A-
of a diverse array of ligand interaction modes. The moderate-to- induced PlexinA1 activation. That model proposed that the
low sequence identity between different classes of Semaphorins Plexin Sema domain associates intramolecularly with its addi-
and Plexins, especially at the binding segments, suggests that tional extracellular domains in an autoinhibited conformation,
there is variability in the specific interatomic contacts in Sema- analogous to the LDL receptor (Rudenko et al., 2002), and that
phorin/Plexin interfaces between families that is buttressed by Sema3A disrupts this intramolecular association (Antipenko
a framework of conserved interactions. This variability may be et al., 2003; Takahashi and Strittmatter, 2001). Although we
necessary for coding the complex pattern of specificity in the have no unequivocal data to support or refute such a model,
growth- or motility-guiding activities. Nevertheless, the orienta- our ITC experiments of A39R binding to full-length versus trun-
tion of each component in the complex, the Sema-Sema dock- cated PlexinC1 ECD show identical binding energetics and
ing geometry, and the global usage of structural elements should monophasic association, indirectly implying that PlexinC1 is in
be generally conserved for Semaphorins and Plexins, as has the same conformation in the full-length ECD and truncated
been found in other ligand-receptor families with similar extents forms (Figures 1D and 1E). An important caveat to our experi-
of sequence identity. ments is that thermodynamic measurements would not detect
The poxvirus-derived A39R has evolved a more efficient a potential biphasic binding kinetics indicative of a two-step
PlexinC1 binding surface, yet it is smaller than that of Sema7A, binding event involving a Plexin conformational change. Never-
with fewer overall contacts. Our structural analysis showed theless, whether Plexin undergoes a conformational change
that the central binding elements of Sema7A are rigorously main- prior to Semaphorin binding, clearly a Semaphorin-induced
tained in the viral complex; therefore, how does A39R achieve arrangement of Plexin into a specifically orientated dimer is the
higher affinity binding? This is very difficult to clearly answer, means of receptor activation. Future structural and mechanistic
as the sources of free energy differences between interacting studies can now address whether the preactivated Plexin is
proteins are not obviously identified by inspection of structures involved in an intramolecular association that contributes to
and are contributed by many subtle effects such as intramolec- the maintenance of an autoinhibited state.
ular side chain cooperativity, solvent restructuring at the binding The Sema7A/PlexinC1 and A39R/PlexinC1 complexes evoke
surface, and enthalpy-entropy compensation. Thus, we suspect several important questions. First, where is the Neuropilin-
that it is due to a collective reduction of energetic penalties docking site in a class 3 Semaphorin-Plexin complex? Although
across the entire Semaphorin/Plexin interface, perhaps in predictions could be made on the basis of our structures
combination with an optimization of protein dynamics, such as and the previous mutagenesis mapping data (Antipenko et al.,
lowering overall temperature factors (Table S1), to gain favor in 2003), accurate insights may require a complex structure
entropic change. The high similarity of Sema7A and A39R encompassing Neuropilin. Second, does Sema7A bind integrins
suggests the likelihood that the poxviruses have acquired the directly? It has been suggested Sema7A promotes axon growth
A39R gene by hijacking and mutating the Sema7A gene, rather and initiates T cell-mediated inflammation responses through
than through convergent evolution where the virus would have integrins, but not PlexinC1, using an RGD-dependent mechanism
evolved a structurally distinct scaffold for PlexinC1 binding (Pasterkamp et al., 2003; Suzuki et al., 2007). The RGD motif of
(Gewurz et al., 2001). Thus, this does not seem to be a case of Sema7A (Arg267-Gly268-Asp269) is buried (Figure S5) and is
molecular ‘‘mimicry’’ as much as simply modifying an existing unlikely to be recognized by integrins. Finally, the complex struc-
binding scaffold with subtle improvements. Given the lack of tures can now be used as a basis to interrogate Sema/Plexin
involvement of the PSI and Ig domains in Plexin interactions, interactions in vivo with high precision. While mutation of the
these domains may have been lost in A39R over the course of key determinants of both Sema7A-PlexinC1 and A39R-PlexinC1
evolution. interfaces clearly compromise binding (Figure 6, Figure S4), fur-
The dimerized Sema7A/PlexinC1 and A39R/PlexinC1 com- ther work will be necessary to demonstrate the consequences
plexes support a model of PlexinC1 activation via dimerization of such perturbation in a cellular or organismal context. Such
akin to many other cell-surface receptors. This, in itself, is an functional validation should determine whether modulation of
important advance since the downstream mechanisms of Sem- Semaphorin-Plexin recognition holds clinical promise for applica-
aphorin/Plexin repulsive signaling are not well understood, and tions involving cell guidance, e.g., directional nerve regeneration.

Cell 142, 749–761, September 3, 2010 ª2010 Elsevier Inc. 759


EXPERIMENTAL PROCEDURES Isothermal Titration Calorimetry
The proteins used for calorimetry were all expressed from HEK293H cells. ITC
Cloning, Cell Culture, and Baculovirus Generation was carried out on a VP-ITC calorimeter (MicroCal, Northhampton, MA) at
Human Sema7A, PlexinC1-SemaPSI, and PlexinC1-ECD, were expressed in 30 C. All samples were thoroughly degassed before titration. The titration
the BacMam system as described previously (Dukkipati et al., 2008), with data were processed with MicroCal Origin software, Version 5.0.
a 7-His tag directly attached to the C terminus of each expression construct.
Full-length Vaccinia virus A39R was expressed in both the BacMam system
and the baculovirus-insect cell system. Sf9 and hi5 cells were maintained in ACCESSION NUMBERS
the HyQ-SFX (HyClone) and InsectXpress (Lonza) media. HEK293H cells
(Invitrogen) and GnTI- HEK293 cells (Reeves et al., 2002) were maintained in The structure factors and coordinates for the Sema7A/PlexinC1-SemaPSI
CDM4HEK293 media (HyClone). For generation of recombinant viruses, the complex, free A39R and the A39R/PlexinC1-SemaPSI complex have been
expression constructs and the BacVector-3000 baculovirus DNA (EMD deposited in the Protein Data Bank (accession codes 3NVQ, 3NVX, and
Chemicals) were used to cotransfect sf9 cells in 6-well plates. After 5 days, 3NVN).
the resulted low titer virus stock was harvested and was used to infect Sf9 cells
at 2 3 106 cell/ml for amplification. SUPPLEMENTAL INFORMATION

Protein Preparation Supplemental Information includes Extended Experimental Procedures, five


The amplified baculoviruses were used to infect 1-6 liters of HEK293 cells figures, and three tables and can be found with this article online at doi:
or hi5 cells at a density of 1.0–1.5 3 106 cells/ml. After 72 hr, the cells were 10.1016/j.cell.2010.07.040.
pelleted and the supernatants were concentrated. The recombinant proteins
in the supernatant were captured by Ni-NTA metal affinity resin (QIAGEN)
and eluted with 300 mM imidazole (pH 7.5). The eluted proteins were further ACKNOWLEDGMENTS
purified with gel filtration.
We thank the staff at the LS-CAT beamlines at APS and the staffs of ALS and
Glycan Trimming, Crystallization, and Data Collection SSRL for the support in X-ray data collection. We also thank L. Colf, and
and Processing N. Goriatcheva for help with the expression of A39R. X.H. is supported by
For the Sema7A-PlexinC1 complex, GnTI HEK293-expressed proteins were the NIH grant 1R01GM07805, and K.C.G. is an Investigator of the Howard
trimmed with carboxypeptidase-A and Endo-H. The digested products were Hughes Medical Institute and is supported by NIH-R01-AI51321. The Struc-
further purified with size exclusion columns and concentrated to 10 mg/ml. tural Biology Facility is supported by the R.H. Lurie Comprehensive Cancer
Crystallization was performed via the sitting-drop vapor-diffusion method. Center of Northwestern University.
The Sema7A and PlexinC1-SemaPSI proteins, at 1:1 molar ratio, were mixed
with an equal volume of reservoir solution and equilibrated against the reser- Received: March 19, 2010
voir solution containing 12% PEG1000, 0.1 M Tris (pH 7.5), and 0.2 M Li2SO4. Revised: June 8, 2010
The A39R used for crystallization was expressed from hi5 cells using the Accepted: July 20, 2010
pAcGP67A vector. A39R was His-tagged at the N-terminus and expressed Published online: August 19, 2010
in the presence of 10 mM kifunensine in the expression media (InsectXpress).
The His-tag was removed from A39R with TEV protease, the glycans were REFERENCES
trimmed with Endo-H, and the protein was further purified by gel filtration.
A39R crystallized in the following condition: 50 mM sodium citrate (pH 5.0), Antipenko, A., Himanen, J.P., van Leyen, K., Nardi-Dei, V., Lesniak, J., Barton,
0.2 M Li2SO4, 15% PEG3350, and 20% glycerol. W.A., Rajashankar, K.R., Lu, M., Hoemme, C., Püschel, A.W., and Nikolov,
For the A39R-PlexinC1-SemaPSI complex, the A39R protein was expressed D.B. (2003). Structure of the semaphorin-3A receptor binding module. Neuron
from the BacMam system following the same procedure for PlexinC1-Sema- 39, 589–598.
PSI. The complex was crystallized in the following condition: 6% PEG8000,
Bork, P., Doerks, T., Springer, T.A., and Snel, B. (1999). Domains in plexins:
0.1 M CaCl2, 0.1 M HEPES (pH 7.0), and 0.2 M NaCl.
links to integrins and transcription factors. Trends Biochem. Sci. 24, 261–263.
The Sema7A/PlexinC1 and A39R/PlexinC1 data were measured at the
Life Sciences Collaborative Access Team (LS-CAT) beamlines 21-ID-D and Bricogne, G., Vonrhein, C., Flensburg, C., Schiltz, M., and Paciorek, W. (2003).
21-ID-G at the Advanced Photon Source (APS) and were processed with Generation, representation and flow of phase information in structure determi-
HKL2000 (Otwinowski and Minor, 1997) (Table S1). The A39R data were nation: recent developments in and around SHARP 2.0. Acta Crystallogr. D
measured at both the Stanford Synchrotron Research Lab (SSRL) and the Biol. Crystallogr. 59, 2023–2030.
Advanced Light Source (ALS) and were processed with Mosflm and scaled Brünger, A.T., Adams, P.D., Clore, G.M., DeLano, W.L., Gros, P.,
with Scala of the CCP4 program suite (Collaborative Computational Project, Grosse-Kunstleve, R.W., Jiang, J.S., Kuszewski, J., Nilges, M., Pannu, N.S.,
Number 4, 1994). The statistics on the data are presented in Table S1. et al. (1998). Crystallography & NMR system: a new software suite for macro-
molecular structure determination. Acta Crystallogr. D Biol. Crystallogr. 54,
Structure Determination and Model Refinement 905–921.
For structure determinations of Sema7A/PlexinC1-SemaPSI and free A39R,
Collaborative Computational Project, Number 4. (1994). The CCP4 suite:
experimental phases were calculated using heavy atom derivatives by the
programs for protein crystallography. Acta Crystallogr. D Biol. Crystallogr.
SIRAS method, as implemented in the program SOLVE (Terwilliger and
50, 760–763.
Berendzen, 1999) and SHARP (Bricogne et al., 2003). For structure determina-
tion of the A39R/PlexinC1-SemaPSI complex, the components from the above Comeau, M.R., Johnson, R., DuBose, R.F., Petersen, M., Gearing, P.,
two structures were used as search models in molecular replacement. The VandenBos, T., Park, L., Farrah, T., Buller, R.M., Cohen, J.I., et al. (1998). A
models were refined with CNS (Brünger et al., 1998). The final models include poxvirus-encoded semaphorin induces cytokine production from monocytes
seven N-linked glycans per PlexinC1-SemaPSI molecule (Asn86, Asn141, and binds to a novel cellular semaphorin receptor, VESPR. Immunity 8,
Asn149, Asn241, Asn252, Asn386, and Asn407), four per Sema7A (Asn105, 473–482.
Asn157, Asn258, and Asn330), and one per A39R (Asn51). Each glycan Dukkipati, A., Park, H.H., Waghray, D., Fischer, S., and Garcia, K.C. (2008).
was modeled with a single GlcNAc residue, as expected, due to the Endo-H BacMam system for high-level expression of recombinant soluble and
treatment of the proteins. A summary of the refinement statistics is given in membrane glycoproteins for structural studies. Protein Expr. Purif. 62,
Table S1. 160–170.

760 Cell 142, 749–761, September 3, 2010 ª2010 Elsevier Inc.


Gewurz, B.E., Gaudet, R., Tortorella, D., Wang, E.W., and Ploegh, H.L. (2001). N-acetylglucosaminyltransferase I-negative HEK293S stable mammalian cell
Virus subversion of immunity: a structural perspective. Curr. Opin. Immunol. line. Proc. Natl. Acad. Sci. USA 99, 13419–13424.
13, 442–450. Rudenko, G., Henry, L., Henderson, K., Ichtchenko, K., Brown, M.S.,
He, Z., Wang, K.C., Koprivica, V., Ming, G., and Song, H.J. (2002). Knowing Goldstein, J.L., and Deisenhofer, J. (2002). Structure of the LDL receptor extra-
how to navigate: mechanisms of semaphorin signaling in the nervous system. cellular domain at endosomal pH. Science 298, 2353–2358.
Sci. STKE 2002, re1. Scott, G.A., McClelland, L.A., Fricke, A.F., and Fender, A. (2009). Plexin C1,
He, X.L., Dukkipati, A., and Garcia, K.C. (2006). Structural determinants of a receptor for semaphorin 7a, inactivates cofilin and is a potential tumor
natriuretic peptide receptor specificity and degeneracy. J. Mol. Biol. 361, suppressor for melanoma progression. J. Invest. Dermatol. 129, 954–963.
698–714. Semaphorin Nomenclature Committee. (1999). Unified nomenclature for the
He, H., Yang, T., Terman, J.R., and Zhang, X. (2009). Crystal structure of the semaphorins/collapsins. Cell 97, 551–552.
plexin A3 intracellular region reveals an autoinhibited conformation through Stamos, J., Lazarus, R.A., Yao, X., Kirchhofer, D., and Wiesmann, C. (2004).
active site sequestration. Proc. Natl. Acad. Sci. USA 106, 15610–15615. Crystal structure of the HGF beta-chain in complex with the Sema domain of
Holmes, S., Downs, A.M., Fosberry, A., Hayes, P.D., Michalovich, D., the Met receptor. EMBO J. 23, 2325–2335.
Murdoch, P., Moores, K., Fox, J., Deen, K., Pettman, G., et al. (2002). Sema7A Suzuki, K., Okuno, T., Yamamoto, M., Pasterkamp, R.J., Takegahara, N.,
is a potent monocyte stimulator. Scand. J. Immunol. 56, 270–275. Takamatsu, H., Kitao, T., Takagi, J., Rennert, P.D., Kolodkin, A.L., et al.
Huppa, J.B., Axmann, M., Mörtelmaier, M.A., Lillemeier, B.F., Newell, E.W., (2007). Semaphorin 7A initiates T-cell-mediated inflammatory responses
Brameshuber, M., Klein, L.O., Schütz, G.J., and Davis, M.M. (2010). TCR- through alpha1beta1 integrin. Nature 446, 680–684.
peptide-MHC interactions in situ show accelerated kinetics and increased Suzuki, K., Kumanogoh, A., and Kikutani, H. (2008). Semaphorins and their
affinity. Nature 463, 963–967. receptors in immune cell interactions. Nat. Immunol. 9, 17–23.
Koller, K.J., and Goeddel, D.V. (1992). Molecular biology of the natriuretic Takahashi, T., and Strittmatter, S.M. (2001). Plexina1 autoinhibition by the
peptides and their receptors. Circulation 86, 1081–1088. plexin sema domain. Neuron 29, 429–439.
Kolodkin, A.L., Matthes, D.J., and Goodman, C.S. (1993). The semaphorin Tamagnone, L., and Comoglio, P.M. (2000). Signalling by semaphorin recep-
genes encode a family of transmembrane and secreted growth cone guidance tors: cell guidance and beyond. Trends Cell Biol. 10, 377–383.
molecules. Cell 75, 1389–1399.
Tamagnone, L., Artigiani, S., Chen, H., He, Z., Ming, G.I., Song, H., Chedotal,
Koppel, A.M., Feiner, L., Kobayashi, H., and Raper, J.A. (1997). A 70 amino A., Winberg, M.L., Goodman, C.S., Poo, M., et al. (1999). Plexins are a large
acid region within the semaphorin domain activates specific cellular response family of receptors for transmembrane, secreted, and GPI-anchored sema-
of semaphorin family members. Neuron 19, 531–537. phorins in vertebrates. Cell 99, 71–80.
Krogsgaard, M., Prado, N., Adams, E.J., He, X.L., Chow, D.C., Wilson, D.B., Terwilliger, T.C., and Berendzen, J. (1999). Automated MAD and MIR structure
Garcia, K.C., and Davis, M.M. (2003). Evidence that structural rearrangements solution. Acta Crystallogr. D Biol. Crystallogr. 55, 849–861.
and/or flexibility during TCR binding can contribute to T cell activation. Mol.
Tessier-Lavigne, M., and Goodman, C.S. (1996). The molecular biology of
Cell 12, 1367–1378.
axon guidance. Science 274, 1123–1133.
Kruger, R.P., Aurandt, J., and Guan, K.L. (2005). Semaphorins command cells
Tong, Y., Chugha, P., Hota, P.K., Alviani, R.S., Li, M., Tempel, W., Shen, L.,
to move. Nat. Rev. Mol. Cell Biol. 6, 789–800.
Park, H.W., and Buck, M. (2007). Binding of Rac1, Rnd1, and RhoD to a novel
Lemmon, M.A., Pinchasi, D., Zhou, M., Lax, I., and Schlessinger, J. (1997). Kit Rho GTPase interaction motif destabilizes dimerization of the plexin-B1
receptor dimerization is driven by bivalent binding of stem cell factor. J. Biol. effector domain. J. Biol. Chem. 282, 37215–37224.
Chem. 272, 6311–6317.
Tong, Y., Hota, P.K., Penachioni, J.Y., Hamaneh, M.B., Kim, S., Alviani, R.S.,
Lev, S., Yarden, Y., and Givol, D. (1992). Dimerization and activation of the kit Shen, L., He, H., Tempel, W., Tamagnone, L., et al. (2009). Structure and
receptor by monovalent and bivalent binding of the stem cell factor. J. Biol. function of the intracellular region of the plexin-b1 transmembrane receptor.
Chem. 267, 15970–15977. J. Biol. Chem. 284, 35962–35972.
Love, C.A., Harlos, K., Mavaddat, N., Davis, S.J., Stuart, D.I., Jones, E.Y., and Uniewicz, K.A., and Fernig, D.G. (2008). Neuropilins: a versatile partner of
Esnouf, R.M. (2003). The ligand-binding face of the semaphorins revealed extracellular molecules that regulate development and disease. Front. Biosci.
by the high-resolution crystal structure of SEMA4D. Nat. Struct. Biol. 10, 13, 4339–4360.
843–848.
Waimey, K.E., and Cheng, H.J. (2006). Axon pruning and synaptic develop-
Luo, Y., Raible, D., and Raper, J.A. (1993). Collapsin: a protein in brain that ment: how are they per-plexin? Neuroscientist 12, 398–409.
induces the collapse and paralysis of neuronal growth cones. Cell 75,
Xiao, T., Takagi, J., Coller, B.S., Wang, J.H., and Springer, T.A. (2004).
217–227.
Structural basis for allostery in integrins and binding to fibrinogen-mimetic
Oinuma, I., Ishikawa, Y., Katoh, H., and Negishi, M. (2004). The Semaphorin 4D therapeutics. Nature 432, 59–67.
receptor Plexin-B1 is a GTPase activating protein for R-Ras. Science 305,
Xiong, J.P., Stehle, T., Zhang, R., Joachimiak, A., Frech, M., Goodman, S.L.,
862–865.
and Arnaout, M.A. (2002). Crystal structure of the extracellular segment of
Otwinowski, Z., and Minor, W. (1997). Processing of X-ray diffraction data integrin alpha Vbeta3 in complex with an Arg-Gly-Asp ligand. Science 296,
collected in oscillation mode. Methods Enzymol. 276, 307–326. 151–155.
Pasterkamp, R.J., Peschon, J.J., Spriggs, M.K., and Kolodkin, A.L. (2003). Xu, X., Ng, S., Wu, Z.L., Nguyen, D., Homburger, S., Seidel-Dugan, C., Ebens,
Semaphorin 7A promotes axon outgrowth through integrins and MAPKs. A., and Luo, Y. (1998). Human semaphorin K1 is glycosylphosphatidylinositol-
Nature 424, 398–405. linked and defines a new subfamily of viral-related semaphorins. J. Biol. Chem.
Pellet-Many, C., Frankel, P., Jia, H., and Zachary, I. (2008). Neuropilins: struc- 273, 22428–22434.
ture, function and role in disease. Biochem. J. 411, 211–226. Yamada, A., Kubo, K., Takeshita, T., Harashima, N., Kawano, K., Mine, T.,
Reeves, P.J., Callewaert, N., Contreras, R., and Khorana, H.G. (2002). Sagawa, K., Sugamura, K., and Itoh, K. (1999). Molecular cloning of a
Structure and function in rhodopsin: high-level expression of rhodopsin with glycosylphosphatidylinositol-anchored molecule CDw108. J. Immunol. 162,
restricted and homogeneous N-glycosylation by a tetracycline-inducible 4094–4100.

Cell 142, 749–761, September 3, 2010 ª2010 Elsevier Inc. 761


Photoadaptation in Neurospora by
Competitive Interaction of Activating
and Inhibitory LOV Domains
Erik Malzahn,1,3 Stilianos Ciprianidis,1,3 Krisztina Káldi,2,3 Tobias Schafmeier,1 and Michael Brunner1,*
1University
of Heidelberg Biochemistry Center, Im Neuenheimer Feld 328, 69120 Heidelberg, Germany
2Department of Physiology, Semmelweis University, POB 259 H-1444 Budapest, Hungary
3These authors contributed equally to this work

*Correspondence: michael.brunner@bzh.uni-heidelberg.de
DOI 10.1016/j.cell.2010.08.010

SUMMARY photoperiodic entrainment is further complicated by light


reflected from the moon since the photoreceptors generally
Light responses and photoadaptation of Neurospora detect even low light levels (Bachleitner et al., 2007; Merrow
depend on the photosensory light-oxygen-voltage et al., 1999).
(LOV) domains of the circadian transcription factor The response of Neurospora to light is mediated by WHITE
White Collar Complex (WCC) and its negative regu- COLLAR-1 (WC-1) and VIVID (VVD). Together with WC-2,
lator VIVID (VVD). We found that light triggers LOV- WC-1 forms the circadian transcription factor White Collar
Complex (WCC), and VVD is expressed under control of light-
mediated dimerization of the WCC. The activated
activated WCC. WC-1 and VVD contain a flavin-binding light-
WCC induces expression of VVD, which then dis- oxygen-voltage (LOV) domain (He et al., 2002; Schwerdtfeger
rupts and inactivates the WCC homodimers by the and Linden, 2003). Flavin-binding LOV domains are blue-light
competitive formation of WCC-VVD heterodimers, sensors in light receptors of plants and fungi (Krauss et al.,
leading to photoadaptation. During the day, expres- 2009). Transduction of the light signal depends on formation of
sion levels of VVD correlate with light intensity, allow- a covalent photoadduct between a conserved cysteinyl residue
ing photoadaptation over several orders of magni- and the bound flavin cofactor (Salomon et al., 2000; Swartz et al.,
tude. At night, previously synthesized VVD serves 2001; Zoltowski et al., 2007, 2009). The photoadduct interferes
as a molecular memory of the brightness of the with the interaction of the LOV core and an adjacent helix (Corch-
preceding day and suppresses responses to light noy et al., 2003; Harper et al., 2003), but it is not clear how the
cues of lower intensity. We show that VVD is essen- signal is further transduced to downstream effectors and tar-
gets. In particular, the molecular basis of light activation of
tial to discriminate between day and night, even in
WCC and adaptation by VVD is not known. VVD has a propensity
naturally ambiguous photoperiods with moonlight. to form rapidly exchanging dimers in response to light (Zoltowski
and Crane, 2008), but the physiological relevance of the dynamic
INTRODUCTION dimerization is obscure.
Once activated by light, LOV domains cannot sense additional
Molecular clocks are circadian timing systems that control light cues. Spontaneous decay of the flavin-cysteinyl photoad-
rhythmic transcription of a large number of genes (Dibner duct resets the LOV domain to the dark state and restores its
et al., 2010; McClung, 2008; McDonald and Rosbash, 2001; light-sensing capacity. The photoadduct of VVD is extremely
Siepka et al., 2007; Vitalini et al., 2006; Woelfle and Johnson, stable (t1/2 4.5 hr), but generally the kinetics of photoadduct
2006). To ensure robust undisturbed circadian oscillation, decay varies from seconds to hours in different LOV domains
clock controlled transcription is in principal independent of light (Corchnoy et al., 2003; Swartz et al., 2001; Zoltowski et al.,
and compensated for temperature and other environmental 2007, 2009).
stimuli. However, exogenous stimuli, such as light, are needed WCC directly controls expression of a large number of genes
as resetting cues to synchronize the clock with the environ- (Smith et al., 2010). The products of two of these genes,
mental cycles (Doyle and Menaker, 2007; Foster et al., 2007; frequency (frq) and vvd, inhibit WCC in negative feedback loops
Helfrich-Förster, 2002; Merrow and Roenneberg, 2007; Nayak (Brunner and Schafmeier, 2006; Loros et al., 2007). The clock
et al., 2007). Hence, circadian systems must be able to sense protein FRQ inhibits WCC in a light-independent fashion and
and properly interpret light intensity signals under a variety of generates a daily rhythm of WCC activity that is essential for
natural conditions. self-sustained circadian rhythms (Schafmeier et al., 2008; Schaf-
Molecular mechanisms that allow circadian systems to meier et al., 2005). In contrast, VVD inhibits WCC in a light-
respond reliably to natural photoperiods with noisy and ambig- dependent fashion, and the oscillating feedback loop dampens
uous light intensity characteristics are not known. In nature, rapidly in constant darkness (Elvin et al., 2005; Heintzen et al.,

762 Cell 142, 762–772, September 3, 2010 ª2010 Elsevier Inc.


Figure 1. Neurospora Responds to Light Intensities over Several Orders of Magnitude
(A) Photoadaptation and reactivation of vvd transcription. Dark-grown Neurospora was exposed for 4 hr to a light intensity of 5 mmol (photons) 3 m2 3 s1 and
then transferred to 130 mmol 3 m2 3 s1 (red line). vvd RNA was measured by qRT-PCR and normalized to actin RNA (black line).
(B) In the photoadapted state, levels of vvd RNA correlate with light intensity. Neurospora was grown for 48 hr in constant light of the indicated intensity, and vvd
RNA was quantified. Average of six RNA measurements of two independent experiments is shown; error bars indicate the standard deviation (SD).
(C) VVD protein levels depend on light intensity. Mycelia were grown for 48 hr at the indicated light conditions and VVD protein levels were measured. A repre-
sentative western blot is shown (n = 3).
(D) Entrainment of the Neurospora clock during cycles of low light intensities. Race tubes were inoculated with WT conidia in light and subsequently entrained to
13/13 hr light-dark (LD) cycles with the indicated light intensities during the light phase. Conidial bands were analyzed densitometrically. Light and dark periods
are indicated by white and black boxes, respectively. Conidial densities of consecutive cycles (26 hr) are plotted below each other. Plots show representative
individual race tubes (n R 9).
(E) Subcellular localization of VVD-GFP. Mycelia of Dvvd strains expressing GFP or VVD-GFP under control of the cpc-1 promoter were analyzed by fluorescence
microscopy. DAPI staining reveals the nuclei.
See also Figure S1.

2001). VVD is not essential for clock function. Deletion of vvd has homodimers that efficiently activate transcription of vvd. VVD
a mild circadian phenotype, resulting in a slight phase delay of accumulates with a delay after light induction and acts as
the circadian clock (Heintzen et al., 2001). This raises a question a competitive inhibitor of WCC homodimerization, leading to
about the physiologically relevant function of VVD. an attenuation of light-induced transcription. Interaction with
When dark-grown Neurospora is abruptly exposed to light, VVD protects light-activated WCC from rapid degradation and
WCC-dependent transcript levels increase rapidly and adapt to thus allows a sizable fraction of WCC to equilibrate with the
a low steady state within 1–2 hr (Schwerdtfeger and Linden, WCC dark form. In the photoadapted state, VVD synthesis trig-
2001). In the photoadapted state, a second transcription spike gered by light-activated WCC is balanced by VVD-dependent
can be evoked by increasing the light intensity (Schwerdtfeger inhibition of WCC. The VVD-mediated desensitization and stabi-
and Linden, 2001). Photoadaptation and light-sensing in the lization of the light activated WCC explain on a molecular level
adapted state depend on VVD. how Neurospora can robustly entrain to artificial and natural
In nature, mycelia growing deeply concealed behind the photoperiods.
bark of a tree may receive the same light intensity level during
the day as openly exposed mycelia receive by moonlight in the RESULTS
course of a clear night. How the photoreceptor system adapts
to different light intensities and facilitates photoperiodic entrain- Neurospora Responds to Light over Several Orders
ment over a broad range of conditions (Tan et al., 2004) is not of Magnitude
known. To characterize the light-sensing system of Neurospora,
In the present study, we elucidated the molecular mechanism we exposed dark-grown mycelia to increasing light intensities
of photoactivation and adaptation of WCC. We show that WCC (5 mmol 3 m2 3 s1 and 130 mmol 3 m2 3 s1) for two consec-
and VVD form highly dynamic homo- and heterodimers via their utive 4 hr periods, and expression of vvd RNA was analyzed by
activated LOV domains. In response to light, WCC initially forms qPCR (Figure 1A). In dark-grown mycelia, vvd RNA could not

Cell 142, 762–772, September 3, 2010 ª2010 Elsevier Inc. 763


be detected. After the mycelia were transferred to the light and Linden, 2003). However, VVD is a small protein (21 kDa) and
(5 mmol 3 m2 3 s1), RNA levels increased rapidly, reached might freely equilibrate between nuclei and cytosol. Hence, it
a peak after 20 min, and subsequently decreased to a lower might readily be lost from nuclei during fractionation. To address
steady-state level. When the light intensity was increased after this issue, we fractionated mycelia that were untreated or treated
4 hr to 130 mmol 3 m2 3 s1, a second spike of vvd RNA induc- with 1% formaldehyde to induce low levels of nonspecific cross-
tion and adaptation was observed. The data confirm that linking (Figure S1B). In untreated mycelia, VVD was found exclu-
Neurospora adapts to constant light but can still sense light in sively in the cytosolic fraction, in agreement with previous
the adapted state (Schwerdtfeger and Linden, 2001). However, reports (Schwerdtfeger and Linden, 2003). In treated mycelia,
we noted that levels of vvd RNA in the adapted state were higher a substantial portion of VVD was recovered in the nuclear frac-
at the higher light intensity. tion, in agreement with the localization of VVD-GFP in living
To determine whether steady-state levels of vvd RNA correlate mycelia. These data suggest that nuclear VVD is readily lost
with the light intensity, we grew mycelia for 48 hr at a number of during subcellular fractionation procedures.
different light intensities, and vvd RNA was quantified. Surpris-
ingly, vvd RNA levels correlated with light intensity over more VVD Competes with Light-Induced Dimerization of WCC
than two orders of magnitude (Figure 1B). Above 130 mmol 3 VVD has been reported to form light-dependent dimers that are,
m2 3 s1, the light-dependent increase in transcript levels however, highly unstable and dynamic (Zoltowski and Crane,
was less pronounced than at lower fluence levels, suggesting 2008). We reasoned that WCC might dimerize in corresponding
that the system approached saturation. VVD protein levels fashion via the LOV domain of WC-1 and that VVD could
increased in a similar fashion (Figure 1C). Transcript levels of compete with this process by interacting with the LOV domain
frq, al-2, and wc-1, which are also controlled by the WCC initially, of WC-1. Attempts to coimmunoprecipitate light or dark states
also rise with increasing light intensity. However, light induction of VVD and WCC from total cell extracts failed in all possible
of these genes saturates readily and therefore does not reflect combinations (data not shown), indicating that VVD and WCC
light intensities over a wide range (Figure S1A available online). do not interact in a stable manner. We constructed a strain
At light intensities below 0.8 mmol 3 m2 3 s1, vvd RNA and expressing a strep-tagged version of VVD (cpc-1-vvdStrep).
VVD protein levels were hardly above background and hence When VVDStrep was purified via a streptavidin affinity resin,
difficult to quantify. To address whether the Neurospora photo- WCC copurified in low but significant amounts (Figure S2A).
receptor system can sense light of lower intensity, we examined To detect interactions of low affinity by independent means,
entrainment to light cues of different intensities in the race-tube we examined the interaction of the LOV domains of WC-1 and
assay (Figure 1D). Entrainment of the conidiation rhythm of VVD in the yeast two-hybrid system. The DNA binding domain
Neurospora to light:dark (LD) cycles depends strictly on func- (BD) of the Gal4 transcription factor was fused to the LOV
tional WCC (Liu, 2003). Neurospora entrained to LD cycles with domain of WC-1, and the transactivation domain of Gal4 (AD)
light intensities as low as 2.8 nmol 3 m2 3 s1, the lowest level was fused to the LOV domain of WC-1 and VVD, respectively.
of light tested. Assuming that the molecular mechanism of light Pairs of LOV domain fusion proteins with BD (‘‘bait’’) and with
sensing and adaptation is the same at high and low light, the the AD (‘‘prey’’) were expressed in a yeast reporter strain that
data suggest that the photoreceptors of Neurospora can allows selection for the interaction of ‘‘bait’’ and ‘‘prey’’ via the
discriminate light intensities over five orders of magnitude. fused LOV domains. Since LOV domains are light receptors,
(2.8 nmol 3 m2 3 s1 to 750 mmol 3 m2 3 s1). the yeast transformants were grown in light as well as in dark-
ness. The LOV domains of WC-1 displayed homodimerization
Subcellular Localization of VVD as well as heterodimeric interaction with VVD when the corre-
The molecular mechanism by which VVD attenuates the activity sponding yeast strains were grown on selective medium in light.
of the WCC is not known, and interaction partners of VVD have However, the strains did not grow in darkness, indicating that the
not been found. VVD might directly inhibit WCC in the nucleus LOV domains did not interact in the absence of light (Figure 2A).
or indirectly affect its activity via signaling molecules, such as A positive control strain grew in light and in darkness while
kinases or phosphatases. To determine whether a nuclear a negative control did not grow at all (Figure 2A). The data
pool of VVD could directly interfere with transcription activation demonstrate that the LOV-domain of WC-1 has the capacity to
by the WCC, we constructed a strain, Dvvd cpc-1-vvd-gfp, interact with itself and with VVD in a light-dependent fashion.
which constitutively expressed GFP-tagged VVD under control We then asked whether the interaction of VVD with WC-1
of the cpc-1 promoter. Expression of VVD-GFP suppressed affects WCC function. WCC is a GATA-type transcription factor
the intense coloring of Dvvd mycelia and restored the wild- that binds to light-responsive elements, such as the proximal
type (WT) phase of conidiation (not shown) suggesting that light regulatory element (LRE) of the frq promoter (pLRE).
VVD-GFP was functional. Analysis of living mycelia by fluores- EMSA analysis can discriminate between light and dark states
cence microscopy revealed that VVD-GFP was located in of the WCC (Froehlich et al., 2002; He and Liu, 2005). Light-acti-
the cytosol and nuclei but displayed slightly greater nuclear vated and dark forms of WCC display predominantly low or high
enrichment than GFP alone, which was uniformly distributed electrophoretic mobility shifts (Figure 2B, lanes 1 and 2). Since
(Figure 1E). we have shown above that the LOV domains of WC-1 dimerize
The observed nuclear localization of VVD-GFP appears to in light-dependent fashion, the light-activated WCC should
contradict previous reports that VVD was not detected in the represent a dimer of the WCC monomer that is the prevalent
nuclear fraction when mycelia were fractionated (Schwerdtfeger form in the dark.

764 Cell 142, 762–772, September 3, 2010 ª2010 Elsevier Inc.


Figure 2. VVD Interferes with Dimerization
of the Light-Activated WCC and Thereby
Mimics the Monomeric Dark State of the
WCC
(A) Light-dependent homo- and heterodimeriza-
tion of the LOV domains of WC-1 and VVD
analyzed by yeast two-hybrid assay. The LOV
domain of WC-1 was fused to the DNA binding
(BD) of the Gal4 transcription factor. The transac-
tivation domain of Gal4 (AD) was fused to the LOV
domains of WC-1 and VVD, respectively as indi-
cated. The yeast strain PJ69-4A expressing HIS3
under control of a Gal4-dependent promoter was
transformed with the indicated combination of
constructs. Positive control, BD-p53/AD-SV40;
negative control, BD and AD vectors without
insert. Corresponding yeast transformants were
plated on either selective (SDC -Leu -Trp -His;
Two Hybrid) or nonselective medium (SDC -Leu
-Trp; Plating) in dilution series as indicated
(1 corresponds to 2.5 OD). Growth of transform-
ants after 3 day incubation is shown.
(B) Light-activated WCC forms dimers. Nuclear
extract of dark-grown mycelia (DD) or light-pulsed
extract (LP) was preincubated without or with
excess of affinity-purified antibody against the
LOV-domain of WC-1. Subsequently samples
were either kept in the dark (DD) or exposed to
a saturating light pulse (LP) when indicated, and
electrophoretic mobility shift assays (EMSA) of
radiolabeled pLRE oligonucleotide were per-
formed. The LOV antibody interferes with dimer-
ization of the light-activated WCC. Monomeric
and dimeric WCC with and without bound LOV
antibody are schematically depicted above the
corresponding lanes. The dark form of the WCC
is shown in gray, and the light-activated WCC is
shown in white.
(C) Concentration dependence of the monomer-
dimer equilibrium of light-activated WCC. EMSA
was performed with 1 and 9 ml light-pulsed nuclear
extract (total volume 20 ml). A long exposure (l.e.)
of the EMSA with low concentration of extract
(1 ml/assay) is shown to compare the ratio of monomeric and dimeric WCC to a high concentration of extract (9 ml/assay).
(D) VVD interferes with dimerization of the light activated WCC functionally mimicking the dark form. Nuclear extract (DD) was light pulsed (LP) and preincubated
with and without recombinant His6-VVD36. When indicated, His antibody was added to neutralize the His6-VVD36, and EMSAs were performed. All EMSAs were
performed at least three times with identical results.
See also Figure S2.

To demonstrate this directly in an EMSA, we generated an binding of the light-activated WCC dimer to its DNA binding
antibody against the LOV domain of WC-1 that should interfere site. Subsequently, LOV antibody was added and the sample
with dimerization of WCC protomers. Nuclear extracts of dark- was then analyzed by EMSA (lane 5). Under these conditions,
grown mycelia were preincubated with an excess of LOV anti- the light-activated WCC was super-shifted, suggesting that the
body over WCC to saturate each WC-1 molecule with one anti- WCC dimer was crosslinked by one antibody bound to two
body. Subsequently, one aliquot was exposed to a saturating LOV domains, a reaction that should be kinetically favored,
light pulse, while the other was kept in the dark. By EMSA anal- when WCC is a dimer. Together, the data demonstrate that
ysis, the dark form of WCC was super-shifted to a single discrete WCC dimerizes in a light-dependent fashion.
band of lower mobility, indicating that one LOV antibody had If WCC dimers are highly dynamic, the monomer-dimer ratio
bound to the WC-1 subunit of the WCC (Figure 2B, lane 3). should be sensitive to the WCC concentration. To vary the
However, a subsequent light pulse did not induce a further concentration of WCC, we performed EMSA in a fixed volume
super-shift (lane 4). This finding indicates that binding of LOV with increasing amounts of light-exposed nuclear extract. As
antibodies to the dark form of WCC interfered with light-depen- expected, the intensity of the EMSA signals increased due to
dent dimerization. In a further assay, nuclear extract was first the increasing amounts of WCC (Figure S2B). At high concentra-
preincubated in light with the pLRE oligonucleotide to allow tions of extract, only the mobility shift corresponding to the

Cell 142, 762–772, September 3, 2010 ª2010 Elsevier Inc. 765


Figure 3. Formation and Decay of the Light Form of
the WCC
(A) Kinetics of formation of the light-activated WCC.
Nuclear extracts of dark grown mycelia (DD, time point
‘‘0’’) were irradiated with the indicated intensities and for
the indicated times and examined by EMSA.
(B) Upper (light complex) and lower (dark complex) bands
were quantified by densitometry and the ratio of upper/
(upper + lower) was calculated for each time point and
the halftime for conversion was determined.
(C) Decay kinetics of the WCC light form. Light-treated
nuclear extracts were incubated in the dark for the indi-
cated times. Left: Samples were analyzed by EMSA.
Open and filled arrowheads indicate the light-activated
and the dark form of the WCC, respectively. EMSAs
were performed at least three times with identical results.
Right: Densitometric analysis of shown EMSA. So that the
functionality of the WCC could be controlled, an aliquot of
each sample was light pulsed to recover the light form
(data not shown).
See also Figure S3.

mobility shift of the light-activated WCC was


fully restored. The data demonstrate that VVD
interacts with WCC and interferes with dimeriza-
tion of the WCC protomers. It thereby function-
ally resets the light-activated WCC to the dark
form.

Photochemical Properties of WCC


and VVD
Activation of WCC protomers by light-induced
dimerization and subsequent competitive inhibi-
tion of dimerization by newly synthesized VVD
readily explains the phenomenon of induction
and adaptation of WCC-dependent transcrip-
tion in response to light on a molecular level.
However, the model does not explain how the
dimeric WCC was detected. However, a long exposure of the system maintains its light-sensing capacity over several orders
X-ray film revealed monomeric and dimeric WCC at low con- of magnitude, since a sizable pool of the WCC must be in the
centrations of light-exposed nuclear extract (Figure 2C and dark state to sense a further increase in light intensity in constant
Figure S2B). This finding indicates that the light-induced dimer- light conditions.
ization of WCC is dynamic and depends on the concentration of To estimate the equilibrium between WCC light and dark
WCC. states, we measured the kinetics of formation and decay of the
For functional characterization of the interaction of VVD and light complex. Nuclear extracts of dark-grown mycelia were
WCC, nuclear extract was first exposed to a saturating light- exposed to light of different intensities, and the kinetics of
pulse, then incubated with recombinant light-activated His6- WCC dimerization was analyzed by EMSA (Figures 3A and 3B).
VVD36 (Zoltowski et al., 2007), and finally, examined by EMSA At high light intensity (420 mmol 3 m2 3 s1), an exposure of
(Figure 2D). In the presence of His6-VVD36, the light-activated 15 s was sufficient for complete light activation of WCC. At inter-
WCC displayed a mobility shift characteristic for the dark form. mediate intensity (80 mmol 3 m2 3 s1), an exposure of about
In its absence, WCC displayed the dimer-specific mobility shift. 5 min was required, and at low light (3.6 and 6 mmol 3 m2 3
These observations indicate that His6-VVD36 disrupted the WCC s1), 20–30 min was needed. When light-exposed nuclear
dimers. In further characterizations, the nuclear extract was extract was transferred to the dark, WCC dimers were stable
exposed to light and preincubated with recombinant light-acti- for several hours (t1/2 4 hr) (Figure 3C).
vated His6-VVD36 as described above. The sample was trans- The ratio of forward versus reverse rates indicates that the
ferred to darkness, and anti-His antibody was added to oligo- fraction of WCC in the dark state depends on light intensity.
merize His6-VVD36 and thereby functionally neutralize the In bright light (420 mmol 3 m2 3 s1), the fraction of WCC in
protein. Although the sample did not receive additional light, the dark form is rather small (6 s/14,400 s = 4 3 104). In constant
subsequent analysis by EMSA revealed that the dimer-specific intermediate light (6 mmol 3 m2 3 s1), about 12.5% of the

766 Cell 142, 762–772, September 3, 2010 ª2010 Elsevier Inc.


mycelia of Dvvd, cpc-1-vvd-gfp were treated with CHX and
exposed to light (Figure 4A), indicating that the previously
synthesized VVD-GFP interfered with the light-induced turnover
of the WCC. The acute induction of transcription by light was
attenuated in the Dvvd, cpc-1-vvd-gfp strain, demonstrating
that VVD-GFP was immediately active as a repressor of the
light-activated WCC (Figure S4A). In a vvd deletion mutant,
light-dependent gene expression is induced to a higher level
and adapts slower (Schwerdtfeger and Linden, 2001) (Fig-
ure S2B). In summary, VVD reduces the transcriptional activity
of the WCC, which results in a stabilization of the transcription
factor, since only the active WCC is subjected to rapid degrada-
tion (Schafmeier et al., 2008).
WCC levels and activity do not depend solely on VVD but are
regulated in a complex manner. On the posttranscriptional level,
FRQ-dependent phosphorylation inactivates and stabilizes
WCC (Schafmeier et al., 2008; Schafmeier et al., 2005). On the
transcriptional level, WCC supports expression of WC-1, the
limiting component of WCC (Káldi et al., 2006).
To assess the contribution of FRQ and VVD to stabilizing the
light-activated WCC, we analyzed WC-1 levels in frq- and vvd-
deficient strains grown in light (Figure 4B). WC-1 levels were
distinctly lower in frq9 (nonfunctional allele of frq) and Dvvd
than in WT cells, demonstrating that FRQ and VVD indepen-
dently stabilize the light-activated WCC to a similar extent.
WC-1 levels were even lower in the double deficient frq9, Dvvd
Figure 4. VVD Stabilizes the Light-Activated WCC strain. This observation indicates that the positive feedback of
(A) Light-induced degradation of WC-1 in the WT and Dvvd, cpc-1-vvd-gfp
WCC on wc-1 transcription cannot compensate its light-induced
strain. Mycelia were grown in constant darkness. Cycloheximide (CHX)
(10 mg/ml) was added, and cultures were transferred to the light. Samples
degradation.
were harvested after the indicated times, and WC-1 was analyzed by western
blotting. VVD Synthesized during the Day Suppresses Moonlight
(B) Positive feedback of FRQ and VVD on levels of light-activated WCC. during the Night
Steady-state expression levels of WC-1 in frq- and vvd-deficient strains grown We reasoned that overexpression of VVD above levels expected
in light are shown in comparison to the WT. A long exposure is shown to illus- by the feedback system on the basis of the prevalent light inten-
trate differences in WC-1 levels in the low-expressing mutant strains. Repre-
sity should result in blindness of Neurospora. To test this predic-
sentative western blots are shown (n = 3).
See also Figure S4. tion, we compared the light-entrainment characteristics of Dvvd,
cpc-1-vvd-gfp, and Dvvd (Figure 5A). After transfer from bright
light to darkness (DD), both strains displayed a free-running con-
WCC should be in the dark state (30 min/240 min). Extrapolating idiation rhythm, indicating that the circadian clock was functional
to low light, we estimate that the fraction of WCC in the dark form and synchronized by the LD transfer. Dvvd entrained to LD
is substantial in dim light (>30% below 1 mmol 3 m2 3 s1). cycles at low and high light intensities. In contrast, Dvvd, cpc-
Thus, the pool of WCC in the dark form is detectable and there- 1-vvd-gfp entrained to LD cycles with a light intensity of
fore of physiological relevance for light-sensing in the photoa- 180 mmol 3 m2 3 s1 but displayed a free-running rhythm at
dapted state. Recombinant His6-VVD36 displayed similar light lower light intensities (2.8 and 180 nmol 3 m2 3 s1). Hence,
sensitivity and photoadduct stability (Figure S3). overexpression of VVD-GFP desensitizes the WCC and renders
the strain blind to the lower levels of light intensity.
VVD Stabilizes the Light-Activated WCC What is the physiological role of desensitization of the WCC
DNA-binding-competent, activated WCC is unstable (Schafme- photoreceptor by VVD? Photoperiods in nature are quite noisy.
ier et al., 2008). Since VVD competes with dimerization of the In particular, nights are not always entirely dark, and thus, abso-
WCC and thereby reduces its activity, we asked whether VVD lute light intensity is an ambiguous cue that does not allow
also affects the degradation kinetics of WCC. WC-1 was rapidly discrimination between day and night. On a clear night with
degraded when dark-grown mycelia were treated with cyclohex- a full moon, the light intensity (20 nmol 3 m2 3 s1) is well
imide (CHX) and then exposed to saturating light (Figure 4A) above the sensitivity of the WCC photoreceptor (<2.8 nmol 3
(Schafmeier et al., 2008). To assess the turnover of light- m2 3 s1), and hence, moonlight should reset the clock.
activated WCC in the presence of VVD under corresponding However, light intensities during the day are more than 103-
conditions, we analyzed the stability of WC-1 in the Dvvd, cpc- fold higher than moonlight. VVD that is expressed at high levels
1-vvd-gfp strain that constitutively expresses VVD-GFP in a during the day is turned over in the dark with a half-time of about
light-independent manner. WC-1 was stable when dark-grown 2.5 hr (Figure 5B). Accordingly, VVD levels should remain

Cell 142, 762–772, September 3, 2010 ª2010 Elsevier Inc. 767


sufficiently high throughout the night to suppress responses
evoked by the considerably dim moonlight and thus allow
entrainment of Neurospora to natural photoperiods.
To test this hypothesis, we exposed race tubes inoculated
with the WT and Dvvd to natural photocycles. Incubation was
started on a dark new moon night and terminated 6 days later
at almost half moon (Figure 5C). The WT entrained to the natural
photoperiod throughout the entire time period. In contrast, Dvvd
entrained for 3 or 4 days, but the rhythm was then obviously
disturbed by the increasing moonlight, and the strain became
arrhythmic. Similarly, the WT entrained to artificial moonlight
(240 nmol 3 m2 3 s1) in a 12 hr high-light:12 hr low-light cycle
while entrainment of Dvvd was disturbed (Figure S5). Both
strains became arrhythmic when the intensity of the artificial
moonlight was raised to 800 nmol 3 m2 3 s1. The data
demonstrate that VVD is required for robust entrainment of
Neurospora to ambiguous natural photoperiods.

DISCUSSION

In this study, we showed that Neurospora has evolved an elabo-


rate molecular mechanism of light activation and adaptation of
transcription that allows discrimination between night and day
on the basis of relative differences in light intensity levels. The
system requires the interaction of two LOV domain photorecep-
tors, the activating photoreceptor WCC, and the inhibitory
photoreceptor VVD that constitute a negative feedback loop
(Elvin et al., 2005; Heintzen et al., 2001). We present here the
molecular mechanism of the feedback and describe properties
that are crucial for the dynamic behavior of this system that
allows light sensing in the photoadapted state over several
orders of light intensity (see model in Figure 6). Light activation
of the LOV domain of the WC-1 facilitates dimerization of proto-
mers of the WCC. Dimers of WCC display a higher activity than
the monomeric WCC protomers, which are prevalent in dark-
ness. Hence, abrupt light exposure of dark grown Neurospora
Figure 5. VVD Is a Critical Element for Controlling Neurospora triggers a burst of transcription of vvd, frq, and other light-
Response to Light
induced genes. Subsequent photoadaptation is a complex pro-
(A) Desensitization of Neurospora to light depends on VVD expression levels.
Densitometric analysis of race-tube assays of Dvvd and Dvvd, cpc-1-vvd-gfp
cess dependent on one hand on rapid degradation of activated
strains as shown in Figure 1D. Strains were grown in high light and then trans- WCC and on the other hand on inactivation and stabilization of
ferred to the dark and exposed to a 13/13 hr LD regime with the indicated light WCC by VVD and FRQ. Rapid degradation of activated WCC
intensity during the light phase. Overexpression of VVD-GFP caused blindness would, in principle, lead to an efficient attenuation of light-
to entraining light cues of lower intensity resulting in a free-running conidiation induced transcription. However, it would be detrimental if light-
rhythm. Plots from representative race tubes are shown (n R 9). induced degradation of WCC were the sole mechanism of adap-
(B) VVD synthesized during the light phase is progressively degraded in the
tation, since this would result in severe depletion of the pool of
dark. Light-grown cultures were transferred to darkness, and VVD levels
were determined after the indicated time periods by western blotting with an WCC. Although transcription of wc-1 is light induced by the
antibody against the N terminus of VVD. Asterisk indicates unspecific reaction WCC (Káldi et al., 2006), this positive feedback loop cannot
of the VVD antibody. Representative western blot is shown (n = 3). compensate for the light-induced degradation. Hence, WCC
(C) VVD is essential for the entrainment of Neurospora to natural photoperiods levels are extremely low in a frq9, Dvvd strain.
with moonlight. Analysis of race-tube assays of the WT and Dvvd. Race tubes VVD, which is rapidly synthesized in light, acts on the light-
were exposed to natural day-night cycles for 6 days beginning at a dark new-
activated WCC by inhibiting, in a competitive manner, the forma-
moon night and ending at almost half moon. Incubation was performed in an
isolated room at 21 C. Conidial density traces of the race-tubes of the WT
tion of highly active WCC dimers. This causes a rapid and effi-
and Dvvd are shown. The moon phases are indicated below the traces. cient attenuation of light-induced transcription. Since inactive
Dvvd became arrhythmic with upcoming moonlight. WCC is not subjected to rapid degradation (Schafmeier et al.,
See also Figure S5. 2008) VVD promotes accumulation of the light-activated but
transcriptionally inactive WCC. Hence, VVD resets the light-acti-
vated WCC to a monomeric, inactive, and stable form that is
functionally equivalent to the dark state (Figure 6).

768 Cell 142, 762–772, September 3, 2010 ª2010 Elsevier Inc.


Figure 6. Model of the Molecular Interactions of WCC
and VVD upon Light Induction and Adaptation
W and V denote the dark forms of the WCC and VVD. Accord-
ingly, W* and V* denote the light-activated forms of the photo-
receptors. Upon light exposure, W is converted to W*, which
dimerizes (W*W*) and efficiently activates transcription of
vvd. The DNA-bound W*W* complex is unstable and rapidly
degraded (Deg.). As V* accumulates, it interacts with W* and
thereby competes with the homodimerization of W*. The
W*V* complex is transcriptionally inactive and stable, leading
to light adaptation of vvd transcription. The stable W*V* accu-
mulates gradually and equilibrates with the dark forms of W
and V. According to law of mass action, the equilibrium is
established by light-dependent conversion of W to W* and
light-independent decay of the W* photoadduct. Stabilization
of W*V* and equilibration with the dark forms lead to a substan-
tial population of W, which allows light sensing in the photoa-
dapted state.

to light cues and thus allows proper interpretation


of ambiguous light signals. Desensitization of
WCC correlates with the expression level of VVD,
which accumulates as a function of duration and
intensity of light exposure. The accumulated VVD
provides a molecular memory of the previous light
history, which is gradually fading in darkness. Since
the extent of desensitization is gradually lost during
the night, the system remains blind for levels of light
that are low in relation to the previous day and can
thus ignore moonlight as a resetting cue. However,
the adapted system remains responsive to stimuli
In addition to VVD, the circadian clock protein FRQ also of higher light intensity. The capacity to respond to light cues
supports inactivation and stabilization of WCC (Schafmeier of higher intensity depends on accumulation of a significant
et al., 2008; Schafmeier et al., 2005). FRQ inactivates WCC in pool of the dark form of WCC. This is facilitated by VVD, which
a light-independent fashion by facilitating its phosphorylation inactivates the light-activated WCC and thereby blocks its
by casein kinase 1a. However, since FRQ expression is sup- degradation. Since the light and dark forms of the WCC are in
ported by WCC, FRQ gradually accumulates to an elevated level equilibrium, stabilization of the light-activated WCC allows accu-
in constant light, resulting in an enhanced feedback of FRQ that mulation of WCC in the dark form. According to law of mass
partially compensates for the increased activity of light-activated action, the equilibrium is established by formation of the photo-
WCC. Thus, vvd-deficient but frq-proficient strains display adduct, which depends on light intensity, and its spontaneous,
a slow and partial attenuation of light-induced transcription light independent decay. When this equilibrium is disturbed by
(Schwerdtfeger and Linden, 2001). a sudden increase in light intensity, the accumulated pool of
Notably, the circadian clock is slightly phase delayed but WCC in the dark state is activated. It transiently outcompetes
otherwise fully functional in vvd-deficient (frq-proficient) strains. the inhibitory capacity of VVD until sufficient additional VVD is
This mild phenotype provokes the question of whether VVD synthesized to establish a new steady state that reflects adapta-
has a physiologically relevant specific function that cannot tion to the higher light intensity. Since the WCC/VVD system can
be provided by FRQ. While FRQ is a general inhibitor of the discriminate light intensities over more than five orders of magni-
WCC, VVD inhibits specifically the light-activated WCC. Under tude, Neurospora readily adapts to habitats that differ in lighting
entrained conditions VVD accumulates to a high level during conditions.
the day and efficiently attenuates transcription induced by the In constant darkness, rhythmic expression of VVD dampens
light-activated WCC. Although VVD is degraded during the night, rapidly and levels of VVD drop below the limit of detection (Elvin
the levels remain sufficiently high to suppress activation of the et al., 2005). The absence of VVD after several days in darkness
WCC by moonlight (>1000-fold lower light intensity than renders the WCC highly sensitive to light cues. This can
daylight). In the absence of VVD (Dvvd), the highly sensitive be viewed as dark adaptation of the light-sensing system of
WCC is activated by moonlight, and the clock cannot entrain Neurospora.
because of the ambiguous light cues. Hence, in natural photope- Finally, the flavin-cysteinyl photoadducts of WCC and VVD are
riods, VVD is an essential component of the circadian clock. stable for several hours while the photoadducts of other LOV
It modulates the sensitivity and responsiveness of the system domains decay in the range of minutes. This extraordinary

Cell 142, 762–772, September 3, 2010 ª2010 Elsevier Inc. 769


photoadduct stability should smoothen the activity profile of the agar. Analysis of race tubes was performed by densitometry with the ChronoII
WCC such that noisy fluctuations in light intensity (e.g., as a result software (T. Roenneberg, Ludwig-Maximilians-Universität Munich).
of moving clouds concealing the sun) are not transduced by the
photoreceptor system. Rather, the photoreceptors integrate the Plasmid Construction
light signals and keep a slowly decaying molecular memory of cpc-1-gfp was constructed by replacement of the ccg-1 promoter with the
cpc-1 promoter in pMF272 (generous gift of Michael Freitag) and insertion of
the maximal light level of the previous time period.
an AscI site between the promoter and the SpeI site. For construction of
We showed that light-sensing and adaptation (desensitization)
cpc-1-vvd-gfp, a genomic PCR product, including the coding region of vvd
in Neurospora depend on homo- and heterodimerization of the and 46 bp upstream of the start codon, was inserted as an AscI-SpeI fragment
activating and inhibitory LOV domains of WC-1 and VVD, into cpc-1-gfp. For construction of cpc-1-vvd-Strep, a PCR fragment contain-
respectively. Flavin-binding LOV domains are also found in plant ing a genomic region of vvd from position 46 to +691, a short linker and nucle-
phototropins (phot1 and phot2). These are membrane-associ- otides coding for a C-terminal Strep-tag (Herzberg et al., 2007), followed by
ated receptor kinases that dimerize in response to blue light a stop codon, was amplified and inserted between the AscI and SpeI sites
of pcpc-1-gfp.
and activate a number of different processes, such as phototro-
pET28a-vvd36 for the expression of a soluble N-terminal truncated version of
pism and chloroplast movement (Christie, 2007). Phototropins VVD fragment (Zoltowski et al., 2007) was kindly provided by Brian R. Crane.
always contain a pair of LOV domains, LOV1 and LOV2. LOV2 Plasmids for the yeast two-hybrid assay were based on pGBD-C1 and
is a dark-state inhibitor of receptor kinase autophosphorylation, pGAD-C1 (James et al., 1996). The VVD36 fragment and the LOV domain of
and its activation by light is essential for signaling (Cho et al., WC-1 were amplified by PCR from Neurospora total complementary DNA
2007; Christie et al., 2002; Kaiserli et al., 2009). The role of (cDNA) and inserted in the EcoRI and BamHI sites of the respective vectors.
LOV1 is less clear. It is not essential for light signaling. Photoac-
tivation of LOV1 appears to have an inhibitory role at high light Neurospora Transformation
intensities (Kaiserli et al., 2009), but the underlying molecular Neurospora conidia (5–7 days old) were transformed by electroporation as
described (Schafmeier et al., 2006). The his-3 locus was used for homologous
mechanism is not understood. The photochemical properties
recombination. Insertion of the target gene was verified by PCR and
of phot1 suggest an interaction between LOV1 and LOV2
sequencing. Homokaryotic strains were generated by purification by several
(Kaiserli et al., 2009). It is tempting to speculate that, like the plating steps and checked by quantitative PCR.
WCC-VVD system, LOV2 might act by facilitating light-depen-
dent dimerization of two phototropins, while heterodimerization
RNA Analysis
of LOV2 with LOV1 could be inhibitory by interfering with Total RNA was extracted with the peqGOLD TriFast reagent (Peqlab). cDNA
receptor dimerization. In principle, heterodimerization of LOV1 was synthesized from 1 mg total RNA with the QuantiTect Reverse Transcrip-
and LOV2 should be kinetically favored since both domains tion Kit (QIAGEN). Transcript levels were quantified by RT-PCR with TaqMan-
occur in the same molecule. However, the quantum yield for Probes in a StepOne system (Applied Biosystems). Triplicate reactions (20 ml)
photoadduct formation (Kasahara et al., 2002) and the rapid containing cDNA equivalent to 0.05 mg RNA were analyzed.
decay of the photoadducts of LOV1 and LOV2 (Salomon et al.,
2000) suggest that light of considerable intensity is required to Protein Analysis
simultaneously activate both LOV domains in one phototropin Extraction of Neurospora protein, subcellular fractionation, and extraction of
nuclear proteins were performed as described (Schafmeier et al., 2005).
molecule. Hence, an intramolecular inhibitory dimerization of
Protein concentration was determined by Bradford assay, and 350 mg (total
LOV1 and LOV2 could desensitize the system at high light inten- extract, cytosol) or 100 mg (nuclei or nuclear extract) were analyzed by SDS-
sities and attenuate the strength of downstream signaling. Thus, PAGE. Western blots were performed as described (Schafmeier et al.,
competitive homo- and heterodimerization of activating and 2005). Enhanced chemiluminescence signals were detected with X-ray films.
inhibitory LOV domains might be a general mechanism for Series of exposures were generated and signals were quantified with Adobe
light-sensing and adaptation. Photoshop as described previously (Schafmeier et al., 2008). The VVD anti-
In summary, we report a molecular mechanism of photo-acti- body used for Western blotting was directed against the N terminus.

vation and adaptation in Neurospora. In response to light, WCC


forms homodimers that induce transcription of vvd. VVD, in turn, Fluorescence Microscopy
competes for binding with WCC, disrupts the WCC dimers, and Neurospora cultures were grown in liquid medium for 1–2 days. Pieces of
mycelia were placed on an object slide and embedded in mounting medium
attenuates light-induced transcription. In the photoadapted
(13% Mowiol [Hoechst], 30% glycerol [pH 8.5]) containing 0.25 mg/ml DAPI.
state, VVD synthesis by light activated WCC is balanced by Microscopy was performed with a Leica DMI 6000B device. GFP and DAPI
VVD-dependent inhibition of WCC. The VVD-mediated desensi- were excited at 470 and 360 nm, respectively.
tization of the light-activated WCC provides a molecular mecha-
nism for how Neurospora robustly entrains to photoperiods. Purification of Recombinant VVD
Affinity purification of VVDStrep was performed with the One-STrEP System
EXPERIMENTAL PROCEDURES (IBA). Protein extracts of light grown Dvvd and Dvvd, cpc-1-vvdStrep corre-
sponding to 150 mg protein were applied to 400 ml Strep-Tactin Superflow
Neurospora Strains and Culture Conditions and incubated for 1 hr at 4 C under constant rotation. After several washing
Neurospora strains used in this study (WT; Dvvd; frq9, Dvvd; Dvvd, cpc-1-gfp; steps, the resin was packed into a column and proteins were eluted with
Dvvd, cpc-1-vvd-gfp; and Dvvd, cpc-1-vvdStrep) carried the ras-1bd mutation desthiobiotin in 250 ml fractions.
(Belden et al., 2007). Standard growth medium contained 2% glucose, 0.5% His6-VVD36 was expressed in Eschericia coli BL21 (DE3) cells under control
L-arginine, 1 3 Vogel’s and 10 ng/ml biotin. Cultures were incubated 48 hr of the T7 promoter. T7 RNA polymerase was induced by addition of 0.1 mM
at 25 C if not indicated otherwise. In the race-tube medium, glucose was IPTG for 18 hr at 16 C. His6-VVD36 was purified by affinity chromatography with
omitted, and arginine was reduced to 0.17%. Race tubes contained 2.2% HisTrap HP columns (GE Healthcare) as recommended by the manufacturer.

770 Cell 142, 762–772, September 3, 2010 ª2010 Elsevier Inc.


Yeast Transformation and Two-Hybrid Assay Belden, W.J., Larrondo, L.F., Froehlich, A.C., Shi, M., Chen, C.H., Loros, J.J.,
The yeast strain PJ69-4A was transformed with 500 ng of plasmid DNA (Gietz and Dunlap, J.C. (2007). The band mutation in Neurospora crassa is a domi-
and Woods, 2002; Ito et al., 1983). Transformed cells were plated on SDC solid nant allele of ras-1 implicating RAS signaling in circadian output. Genes Dev.
medium lacking leucin and tryptophane (SDC -Leu -Trp). Single colonies were 21, 1494–1505.
picked, plated again, and checked for positive transformation by analysis of Brunner, M., and Schafmeier, T. (2006). Transcriptional and post-transcrip-
protein expression. tional regulation of the circadian clock of cyanobacteria and Neurospora.
For the yeast two-hybrid assay, cells were transformed with WC-1LOV- Genes Dev. 20, 1061–1074.
GAL4-BD (bait) and either GAL4-AD-VVD or GAL4-AD- WC-1LOV (prey). Plas-
mids containing GAL4-BD-P53 and GAL4-AD-SV40 were used as positive Cho, H.Y., Tseng, T.S., Kaiserli, E., Sullivan, S., Christie, J.M., and Briggs, W.R.
control, and GAL4-BD and GAL4-AD were used as negative control. Single (2007). Physiological roles of the light, oxygen, or voltage domains of phototro-
clones were inoculated on a sector of plates containing SDC -Leu -Trp and pin 1 and phototropin 2 in Arabidopsis. Plant Physiol. 143, 517–529.
SDC -Leu -Trp -His medium. Dilution series of positive clones were spotted Christie, J.M. (2007). Phototropin blue-light receptors. Annu. Rev. Plant Biol.
on plates for direct comparison of growth rate. 58, 21–45.
Christie, J.M., Swartz, T.E., Bogomolni, R.A., and Briggs, W.R. (2002). Photo-
Electrophoretic Mobility Shift Assay
tropin LOV domains exhibit distinct roles in regulating photoreceptor function.
EMSA with g32P-ATP-labeled double-stranded oligonucleotide corresponding
Plant J. 32, 205–219.
to the proximal LRE (50 -CGCAGAGGACCCTGAACTTTTCGATCCGCTCGA
TCCCCTG GAA-30 ) was performed as described (Froehlich et al., 2002). Puri- Corchnoy, S.B., Swartz, T.E., Lewis, J.W., Szundi, I., Briggs, W.R., and Bogo-
fied oligonucleotide (180 fmol) and 10–20 mg nuclear extract were incubated in molni, R.A. (2003). Intramolecular proton transfers and structural changes
binding buffer (20 mM HEPES, 40 mM KCl, 1 mM EDTA, 10% glycerol, 0.2 mM during the photocycle of the LOV2 domain of phototropin 1. J. Biol. Chem.
DTT, 10 mM MgCl2, 0.1% N-p40) on ice for 30 min. Poly dIdC (1 mg) was used 278, 724–731.
as unspecific competitor. Samples were loaded on a gel containing 4% acryl- Dibner, C., Schibler, U., and Albrecht, U. (2010). The mammalian circadian
amide in TBE buffer. Gels were fixed in 10% acetic acid, dried, and analyzed timing system: organization and coordination of central and peripheral clocks.
by autoradiography. Antibodies used for EMSA specifically were a-His6 Annu. Rev. Physiol. 72, 517–549.
(QIAGEN) and a-LOV, which was generated against a peptide obtained from
Doyle, S., and Menaker, M. (2007). Circadian photoreception in vertebrates.
the WC-1 LOV-domain (RFLQAPDGNVEAGTKREF).
Cold Spring Harb. Symp. Quant. Biol. 72, 499–508.

Analysis of Photoadduct Formation and Decay Elvin, M., Loros, J.J., Dunlap, J.C., and Heintzen, C. (2005). The PAS/LOV
Purified His6-VVD36 or Neurospora nuclear extract were prepared as protein VIVID supports a rapidly dampened daytime oscillator that facilitates
described above. For analysis of photoadduct formation, samples were entrainment of the Neurospora circadian clock. Genes Dev. 19, 2593–2605.
prepared in red light. His6-VVD36 and nuclear extracts were irradiated with Foster, R.G., Hankins, M.W., and Peirson, S.N. (2007). Light, photoreceptors,
the indicated light intensities. Conditions with different light intensities were and circadian clocks. Methods Mol. Biol. 362, 3–28.
established through the use of a variable number of neon tubes in combination
Froehlich, A.C., Liu, Y., Loros, J.J., and Dunlap, J.C. (2002). White Collar-1,
with light absorber foil. Light intensity was measured with a LI-250A Light
a circadian blue light photoreceptor, binding to the frequency promoter.
Meter (LI-COR Biosciences). For decay analysis, His6-VVD36 or nuclear
Science 297, 815–819.
extracts were irradiated with saturating light (10 min, 130 mmol 3 m2 3 s1)
and transferred to darkness. WC-1 photoadduct formation and decay were Gietz, R.D., and Woods, R.A. (2002). Transformation of yeast by lithium
analyzed by EMSA, and the His6-VVD36 photoadduct was analyzed photomet- acetate/single-stranded carrier DNA/polyethylene glycol method. Methods
rically by measurementof the absorbance spectrum of the FAD cofactor as Enzymol. 350, 87–96.
described (Zoltowski et al., 2007). Harper, S.M., Neil, L.C., and Gardner, K.H. (2003). Structural basis of a photo-
tropin light switch. Science 301, 1541–1544.
SUPPLEMENTAL INFORMATION
He, Q., and Liu, Y. (2005). Molecular mechanism of light responses in Neuros-
pora: from light-induced transcription to photoadaptation. Genes Dev. 19,
Supplemental Information includes Extended Experimental Procedures and
2888–2899.
five figures and can be found with this article online at doi:10.1016/j.cell.
2010.08.010. He, Q., Cheng, P., Yang, Y., Wang, L., Gardner, K.H., and Liu, Y. (2002). White
collar-1, a DNA binding transcription factor and a light sensor. Science 297,
ACKNOWLEDGMENTS 840–843.
Heintzen, C., Loros, J.J., and Dunlap, J.C. (2001). The PAS protein VIVID
We thank Johanna Scholz and Krisztina Makara for excellent technical defines a clock-associated feedback loop that represses light input, modu-
assistance. We are grateful to Cornelia Ulbrich for her experienced help with lates gating, and regulates clock resetting. Cell 104, 453–464.
setting up the yeast two-hybrid system. This work was supported by the
Helfrich-Förster, C. (2002). The circadian system of Drosophila melanogaster
DFG grant BR 1375-3-1 to M.B. and T.S., SFB 638 and the FCI to M.B.,
and its light input pathways. Zoology (Jena) 105, 297–312.
GP1188 to E.M. and by an EMBO-HHMI startup grant by the Hungarian
OTKA (K 68960) and ETT (477-05) to K.K., M.B. is a member of CellNetworks Herzberg, C., Weidinger, L.A., Dörrbecker, B., Hübner, S., Stülke, J., and
and S.C. is a member of HBIGS. Commichau, F.M. (2007). SPINE: a method for the rapid detection and analysis
of protein-protein interactions in vivo. Proteomics 7, 4032–4035.
Received: February 16, 2010 Ito, H., Fukuda, Y., Murata, K., and Kimura, A. (1983). Transformation of intact
Revised: June 16, 2010 yeast cells treated with alkali cations. J. Bacteriol. 153, 163–168.
Accepted: August 7, 2010
James, P., Halladay, J., and Craig, E.A. (1996). Genomic libraries and a host
Published: September 2, 2010
strain designed for highly efficient two-hybrid selection in yeast. Genetics
REFERENCES 144, 1425–1436.
Kaiserli, E., Sullivan, S., Jones, M.A., Feeney, K.A., and Christie, J.M. (2009).
Bachleitner, W., Kempinger, L., Wülbeck, C., Rieger, D., and Helfrich-Förster, Domain swapping to assess the mechanistic basis of Arabidopsis phototropin
C. (2007). Moonlight shifts the endogenous clock of Drosophila melanogaster. 1 receptor kinase activation and endocytosis by blue light. Plant Cell 21, 3226–
Proc. Natl. Acad. Sci. USA 104, 3538–3543. 3244.

Cell 142, 762–772, September 3, 2010 ª2010 Elsevier Inc. 771


Káldi, K., González, B.H., and Brunner, M. (2006). Transcriptional regulation of protein from a nuclear repressor toward a cytoplasmic activator. Genes Dev.
the Neurospora circadian clock gene wc-1 affects the phase of circadian 20, 297–306.
output. EMBO Rep. 7, 199–204. Schafmeier, T., Diernfellner, A., Schäfer, A., Dintsis, O., Neiss, A., and Brunner,
Kasahara, M., Swartz, T.E., Olney, M.A., Onodera, A., Mochizuki, N., Fuku- M. (2008). Circadian activity and abundance rhythms of the Neurospora clock
zawa, H., Asamizu, E., Tabata, S., Kanegae, H., Takano, M., et al. (2002). transcription factor WCC associated with rapid nucleo-cytoplasmic shuttling.
Photochemical properties of the flavin mononucleotide-binding domains of Genes Dev. 22, 3397–3402.
the phototropins from Arabidopsis, rice, and Chlamydomonas reinhardtii. Schwerdtfeger, C., and Linden, H. (2001). Blue light adaptation and desensiti-
Plant Physiol. 129, 762–773. zation of light signal transduction in Neurospora crassa. Mol. Microbiol. 39,
Krauss, U., Minh, B.Q., Losi, A., Gärtner, W., Eggert, T., von Haeseler, A., and 1080–1087.
Jaeger, K.E. (2009). Distribution and phylogeny of light-oxygen-voltage-blue- Schwerdtfeger, C., and Linden, H. (2003). VIVID is a flavoprotein and serves as
light-signaling proteins in the three kingdoms of life. J. Bacteriol. 191, 7234– a fungal blue light photoreceptor for photoadaptation. EMBO J. 22, 4846–
7242. 4855.
Liu, Y. (2003). Molecular mechanisms of entrainment in the Neurospora circa- Siepka, S.M., Yoo, S.H., Park, J., Lee, C., and Takahashi, J.S. (2007). Genetics
dian clock. J. Biol. Rhythms 18, 195–205. and neurobiology of circadian clocks in mammals. Cold Spring Harb. Symp.
Loros, J.J., Dunlap, J.C., Larrondo, L.F., Shi, M., Belden, W.J., Gooch, V.D., Quant. Biol. 72, 251–259.
Chen, C.H., Baker, C.L., Mehra, A., Colot, H.V., et al. (2007). Circadian output, Smith, K.M., Sancar, G., Dekhang, R., Sullivan, C.M., Li, S., Tag, A.G., Sancar,
input, and intracellular oscillators: insights into the circadian systems of single C., Bredeweg, E.L., Priest, H.D., McCormick, R.F., et al. (2010). Transcription
cells. Cold Spring Harb. Symp. Quant. Biol. 72, 201–214. factors in light and circadian clock signaling networks revealed by genome-
wide mapping of direct targets for Neurospora WHITE COLLAR COMPLEX.
McClung, C.R. (2008). Comes a time. Curr. Opin. Plant Biol. 11, 514–520.
Eukaryot. Cell, in press. Published online July 30, 2010. 10.1128/EC.00154-10.
McDonald, M.J., and Rosbash, M. (2001). Microarray analysis and organiza-
Swartz, T.E., Corchnoy, S.B., Christie, J.M., Lewis, J.W., Szundi, I., Briggs,
tion of circadian gene expression in Drosophila. Cell 107, 567–578.
W.R., and Bogomolni, R.A. (2001). The photocycle of a flavin-binding domain
Merrow, M., and Roenneberg, T. (2007). Circadian entrainment of Neurospora of the blue light photoreceptor phototropin. J. Biol. Chem. 276, 36493–36500.
crassa. Cold Spring Harb. Symp. Quant. Biol. 72, 279–285.
Tan, Y., Merrow, M., and Roenneberg, T. (2004). Photoperiodism in Neuros-
Merrow, M., Brunner, M., and Roenneberg, T. (1999). Assignment of circadian pora crassa. J. Biol. Rhythms 19, 135–143.
function for the Neurospora clock gene frequency. Nature 399, 584–586. Vitalini, M.W., de Paula, R.M., Park, W.D., and Bell-Pedersen, D. (2006). The
Nayak, S.K., Jegla, T., and Panda, S. (2007). Role of a novel photopigment, rhythms of life: circadian output pathways in Neurospora. J. Biol. Rhythms
melanopsin, in behavioral adaptation to light. Cell. Mol. Life Sci. 64, 144–154. 21, 432–444.
Salomon, M., Christie, J.M., Knieb, E., Lempert, U., and Briggs, W.R. (2000). Woelfle, M.A., and Johnson, C.H. (2006). No promoter left behind: global circa-
Photochemical and mutational analysis of the FMN-binding domains of the dian gene expression in cyanobacteria. J. Biol. Rhythms 21, 419–431.
plant blue light receptor, phototropin. Biochemistry 39, 9401–9410. Zoltowski, B.D., and Crane, B.R. (2008). Light activation of the LOV protein
Schafmeier, T., Haase, A., Káldi, K., Scholz, J., Fuchs, M., and Brunner, M. vivid generates a rapidly exchanging dimer. Biochemistry 47, 7012–7019.
(2005). Transcriptional feedback of Neurospora circadian clock gene by Zoltowski, B.D., Schwerdtfeger, C., Widom, J., Loros, J.J., Bilwes, A.M.,
phosphorylation-dependent inactivation of its transcription factor. Cell 122, Dunlap, J.C., and Crane, B.R. (2007). Conformational switching in the fungal
235–246. light sensor Vivid. Science 316, 1054–1057.
Schafmeier, T., Káldi, K., Diernfellner, A., Mohr, C., and Brunner, M. (2006). Zoltowski, B.D., Vaccaro, B., and Crane, B.R. (2009). Mechanism-based
Phosphorylation-dependent maturation of Neurospora circadian clock tuning of a LOV domain photoreceptor. Nat. Chem. Biol. 5, 827–834.

772 Cell 142, 762–772, September 3, 2010 ª2010 Elsevier Inc.


Cell Flow Reorients the Axis
of Planar Polarity in the
Wing Epithelium of Drosophila
Benoı̂t Aigouy,1 Reza Farhadifar,2 Douglas B. Staple,2 Andreas Sagner,1 Jens-Christian Röper,1 Frank Jülicher,2,*
and Suzanne Eaton1,*
1Max Planck Institute of Molecular Cell Biology and Genetics, Pfotenhauerstrasse 108, Dresden 01307, Germany
2Max Planck Institute for the Physics of Complex Systems, Noethnitzer Strasse 38, Dresden 01187, Germany
*Correspondence: julicher@pks.mpg.de (F.J.), eaton@mpi-cbg.de (S.E.)
DOI 10.1016/j.cell.2010.07.042

SUMMARY Feedback loops arising from interactions of PCP proteins within


and between cells may be sufficient for the local alignment of
Planar cell polarity (PCP) proteins form polarized PCP complexes between small groups of cells (Amonlirdviman
cortical domains that govern polarity of external et al., 2005; Tree et al., 2002a; Tree et al., 2002b). Proposed
structures such as hairs and cilia in both vertebrate cellular mechanisms underlying local feedback loops include
and invertebrate epithelia. The mechanisms that preferential interactions between Fmi/Fz and Fmi/Stbm com-
globally orient planar polarity are not understood, plexes across cell boundaries, and the decreased susceptibility
of these complexes to endocytosis (Chen et al., 2008; Strutt
and are investigated here in the Drosophila wing
and Strutt, 2008; Wu and Mlodzik, 2008). Also, alignment of
using a combination of experiment and theory. microtubules may bias the delivery of Fz to the distal side of
Planar polarity arises during growth and PCP the cell (Shimada et al., 2006).
domains are initially oriented toward the well-charac- While these mechanisms can polarize PCP domains intracellu-
terized organizer regions that control growth and larly and align them locally, less is understood about mecha-
patterning. At pupal stages, the wing hinge con- nisms that specify global alignment of PCP domains with the
tracts, subjecting wing-blade epithelial cells to proximal-distal (PD) axis of the wing. The atypical Cadherins
anisotropic tension in the proximal-distal axis. This Fat (Ft) and Dachsous (Ds), and a regulatory golgi kinase Four-
results in precise patterns of oriented cell elongation, jointed (Fj) influence global orientation of PCP domains by
cell rearrangement and cell division that elongate the a mechanism that is unclear (Adler et al., 1998; Ma et al., 2003;
blade proximo-distally and realign planar polarity Matakatsu and Blair, 2004; Strutt and Strutt, 2002; Zeidler
et al., 2000). The Fat ligand Ds is expressed highly in proximal
with the proximal-distal axis. Mutation of the atypical
regions that give rise to the hinge, but at lower levels in the
Cadherin Dachsous perturbs the global polarity more distal wing blade, during both larval and pupal stages
pattern by altering epithelial dynamics. This mecha- (Cho and Irvine, 2004; Clark et al., 1995; Ma et al., 2003; Mata-
nism utilizes the cellular movements that sculpt katsu and Blair, 2004; Strutt and Strutt, 2002). Fj, which regulates
tissues to align planar polarity with tissue shape. their activity (Ishikawa et al., 2008) is expressed with an opposite
pattern (Villano and Katz, 1995; Zeidler et al., 2000). These
INTRODUCTION opposing patterns have been proposed to cause intracellular
asymmetries that directly bias accumulation of core PCP
The PCP pathway coordinates tissue planar polarity in vertebrate proteins in wing cells (Ma et al., 2008, 2003; Strutt and Strutt,
and invertebrate epithelia (Simons and Mlodzik, 2008; Vladar 2002; Tree et al., 2002a). However this is not the case in the
et al., 2009). PCP proteins localize to apical junctions and form abdomen where Ft/Ds and PCP proteins act independently to
intracellularly polarized domains with different compositions. control trichome polarity (Casal et al., 2006). Even in the wing,
In the Drosophila wing, these domains link proximal and distal Ft and Ds do not act at the time that PCP domains actually align
cell boundaries and orient wing hair outgrowth distally. Distal with the PD axis, but much earlier (during larval or prepupal
complexes containing Flamingo (Fmi), a.k.a. Starry night (Stan), stages), to influence the global PCP pattern (Matakatsu and
Frizzled (Fz), Dishevelled (Dsh) and Diego (Dgo) interact across Blair, 2004; Strutt and Strutt, 2002). Ft and Ds regulate both
cell boundaries with proximal complexes containing Fmi, Stra- the amount and orientation of proliferation in the larval wing
bismus (Stbm), a.k.a. Van Gogh (Vang) and Prickle (Pk). In the epithelium (Baena-Lopez et al., 2005; Bryant et al., 1988; Clark
absence of any single PCP protein, the others do not polarize et al., 1995; Garoia et al., 2005, 2000). It is not yet clear whether
well and hair outgrowth is misoriented (Seifert and Mlodzik, this activity is relevant to the PD alignment of PCP proteins
2007; Strutt and Strutt, 2005; Uemura and Shimada, 2003). during pupal stages.

Cell 142, 773–786, September 3, 2010 ª2010 Elsevier Inc. 773


A B

50 µm
15 hAPF 30 hAPF
C D E F

Time [hAPF]
20 µm 32h20 APF
16 hAPF 20h50 APF PCP order elongation

Figure 1. Time Evolution of Planar Polarity


(A and B) The magnitude and axis of average nematic order (yellow bars) overlaid on Stbm:YFP-expressing wings, at 15 hAPF (A) and 30 hAPF (B). Red dots
indicate Fz:YFP domain polarity. Green ellipses indicate the anterior crossvein.
(C–E) Stbm:YFP at the indicated times between veins L1 and L3. Yellow bars show the PCP nematic for each cell.
(F) Magnitude (arbitrary units) and axis of average nematic order (left) and cell elongation (right) as a function of time (indicated on the y axis and by color) derived
from the wing shown in (C–E).
In all figures, anterior is top and proximal is left.
See also Figure S1.

Our previous experiments suggested that PCP domain retical analysis to extract key mechanisms that couple cell rear-
polarity does not develop de novo during pupal stages; PCP rangements to PCP reorientation.
domains are polarized in both larval wing discs and prepupal
wings, but polarity is not aligned with the PD axis. At early pupal RESULTS
stages, the PCP axis is oriented at an angle to the PD axis
(Classen et al., 2005). Later, by the time wing hairs form, PCP Planar Polarity Points Initially toward the Wing Margin
domains are oriented along the PD axis of the wing. Alignment and Reorients Distally
with the PD axis occurs during a phase in which wing epithelial To investigate how PCP order evolves during pupal develop-
cells are exchanging their cell contacts (Classen et al., 2005). ment, we imaged wings expressing Stbm:YFP between 15 and
One consequence of remodelling is an increase in hexagonal 34 hr after puparium formation (hAPF). We developed a method
order of the wing epithelium. Theoretical analysis suggested to quantify planar polarity based on the cell perimeter intensity of
that different types of fluctuations could guide epithelial cells Stbm:YFP. This method quantifies the axis and magnitude of
initially disordered by proliferation toward a hexagonal state polarity, but not its vector orientation (Supplemental Theoretical
(Farhadifar et al., 2007). Here, we investigate the relationship Procedures, 1.1 and Figures S1A–S1E available online). We
between these cell rearrangements and the temporal evolution started by quantifying the axis of PCP at 15 hAPF (Figure 1A);
of PCP orientation patterns. We quantitatively analyze time- we also determined the polarity vectors in these wings by
lapses of pupal wing epithelia and combine these data with theo- creating Fz:YFP-expressing clones (Figures S1F–S1H). As

774 Cell 142, 773–786, September 3, 2010 ª2010 Elsevier Inc.


suggested previously (Classen et al., 2005), PCP at early stages different velocities (Figures 2D–2F, Movie S3B). Between 15
is correlated over long distances (Figure S1M) and is oriented and 18 hAPF, velocities have both inward and proximal compo-
such that Fz:YFP domains face the wing margin (Figures 1A nents (Figure 2D). Later, flow is proximally oriented and fastest in
and 1C and Figures S1F–S1I). Starting at about 18 hAPF, the the middle of the wing blade (Figure 2E). These inhomogeneous
magnitude of the average alignment of groups of cells (local velocities define local compression, shear and rotation as cells
nematic order, see Supplemental Theoretical Procedures, 1.2) move with respect to each other (Supplemental Theoretical
begins to decrease (Figures 1D and 1F and Figure S1J), and Procedures, 4). The shear axis in a particular region can be
the average axis of alignment begins to rotate toward the PD thought of as the local axis of tissue deformation. In the
axis (Figure 1F and Figures S1J and S1K). Reduced alignment wing, local shear axes are oriented in a fan-like pattern that is
reflects both reduced polarity of individual cells and reduced roughly symmetric about the third wing vein (Figures 2G–2I,
polarity correlation over distance (Figures S1L and S1M). After Figures S2J and S2K, and Movie S3C). Although the wing as
reaching a minimum about 20 hAPF, the magnitude of average a whole does not rotate, local rotation does occur and is mainly
nematic order increases; by 26 hAPF nematic order is again clockwise anterior to the third wing vein and anti-clockwise
maximal and aligned parallel to the PD axis (Figures 1B, 1E, posterior to it (Figures 2J–2L, Figure S2L, and Movie S3D). These
and 1F, Figures S1J and S1K, and Movie S1). Where we have patterns of shear and rotation lengthen the wing blade in the
determined the direction of polarity, we quantify average polar PD axis and narrow it in the anterior-posterior (AP) axis. Strik-
order by a measure ranging from zero to one (one corresponding ingly, the observed antisymmetric rotation pattern would shift
to perfect alignment) (Supplemental Theoretical Procedures, early margin-oriented polarity toward the distal direction.
1.3). At 15 hAPF, polar order averaged over the whole wing blade To investigate whether the tissue flows that reshape the wing
is only 0.60, because polarity is oriented differently in anterior blade were driven by hinge contraction, we completely severed
and posterior wing regions (Figure 1A). By 32 hAPF average polar the hinge from the blade before contraction occurred (Figure 3A).
order reaches 0.98 (Figure 1B). Thus, PCP domains do not After severing, wing-blade tissue flows distally rather than prox-
develop polarity de novo during pupal stages, but rather reorient imally (Figure 3C and Movie S4A). Shear is reduced and is mainly
pre-existing polarity. perpendicular to the PD axis (Figure 3D and Movie S4B), rather
Polarity reorientation is reflected in the influence of fz mutant than forming a fan-like pattern. Rotation is also reduced, and
clones on the polarity of adjacent wild-type tissue. At 15 hAPF, its orientation is reversed compared to unperturbed wings
fz clones perturb polarity of tissue lying between the clone and (compare Figure 3E with Figure 2K, and Movie S4C with Movie
the wing margin (Figures S1N and S1O) consistent with the early S3D). As a result, the wing blade undergoes opposite shape
polarity pattern. After reorientation, polarity is disturbed in wild- changes to those that normally occur. Nevertheless, wing hairs
type tissue lying distal to the clone (Vinson and Adler, 1987). (not shown) and longitudinal veins (Figure 3B) form normally.
We noted that reorientation of PCP correlated with a transient These data suggest that hinge contraction exerts mechanical
cell shape deformation that elongates cells in the PD axis stresses that contribute to the observed cell flow patterns that
(Figure 1F). Quantifying elongation (Supplemental Theoretical change the shape of the wing blade.
Procedures, 2) reveals that average polarity shrinks in magnitude
and reorients toward the PD axis as cells elongate. The magni- Cell Boundary Tension Is Elevated along the PD Axis
tude of average polarity along the PD axis then increases as during Hinge Contraction
cell elongation decreases (Figure 1F). The precise temporal To ask how cell boundary tension changes during hinge contrac-
correlation of these events suggests that they are controlled by tion, we used a pulsed UV laser to sever cell boundaries lying at
the same underlying mechanisms. different angles to the PD axis, and monitored the movements
of adjacent vertices. The initial velocity with which vertices move
Dramatic Morphogenetic Movements Reshape the Wing apart is a measure of cell boundary tension. Before hinge contrac-
during Pupal Development tion, tension is similar on all cell boundaries. When the hinge
To explore global changes occurring in the wing as cells change contracts, tension increases specifically on those cell boundaries
shape and PCP reorients, we imaged ECadherin:GFP-express- lying at angles close to that of the PD axis (Figures S3A and S3B).
ing wings at low magnification from 15 to 32 hAPF. Dramatic This suggests that hinge contraction exerts forces on the blade
morphogenetic movements reshape the wing during this time that direct cellular flows via an anisotropic stress profile.
(Figures 2A–2C and Movie S2). At 15–16 hAPF, the hinge and
blade regions are roughly equal in size and the shape of the hinge Tissue Flow in the Wing Blade Results from Oriented
is not yet recognizable (Figure 2A). Between 15 and 24 hAPF, the Cell Divisions, Cell Elongation, and Neighbor Exchanges
hinge undergoes patterned contractions that halve its area To investigate the cellular events underlying tissue flow,
and shape the allula and costa (Figures 2A–2C, Movie S2, and shear, and rotation, we tracked groups of cells between veins 3
Movie S3A). While wing-blade area remains constant during and 4, starting 15 hAPF. This revealed that local tissue shape
this time, wing shape changes – elongating in the PD axis and change occurs in two distinct phases. The first is dominated by
narrowing in the AP axis (Figures 2A–2C and Movie S2). oriented cell division and PD cell elongation, and the second by
To quantify tissue movements underlying this shape change, oriented cell rearrangements (Figure 4 and Movie S5A).
we tracked different regions and calculated local velocity vectors During phase I (about 15–24 hAPF), cell elongation increases
(Supplemental Theoretical Procedures, 3). As the hinge con- in the PD axis (Figures 4A–4C and 4G). Cell boundary loss
tracts, wing-blade cells flow proximally toward the hinge at exceeds new boundary formation (Figure 4G), as confirmed by

Cell 142, 773–786, September 3, 2010 ª2010 Elsevier Inc. 775


A B
* C
*
15h30' 80 µm
ubi-Ecad:GFP 24h00' 33h30'
D E F
50 µm/h

L3 L3 L3

80 µm
ubi-Ecad:GFP 24h00' 31h40'
G H I -1
0.3 h

J K L -1
0.1 h
-1
0.05 h
-1
0.01 h

anti-clockwise clockwise

Figure 2. Tissue Shape Changes during Hinge Contraction


(A–C) Wing images from a movie spanning roughly 15-33 hAPF. The hinge (blue) contracts, the wing blade (red) elongates. Allula (arrowhead) and costa (asterisk)
are indicated. The area of the wing hinge is 102 3 103 mm2 in (A) and 50 3 103 mm2 in (C). The area of the wing blade is 121 3 103 mm2 in (A) and 127 3 103mm2 in (C).
(D–L) Locally averaged features of the cellular flow field at indicated times.
(D–F) Flow velocity vectors. Speed corresponds to arrow length as indicated.
(G–I) Rate and axis of pure shear. Bar length indicates shear rate.
(J–L) Local rotation rate (indicated by circle size) in radians per hour.
L3 indicates the third longitudinal vein.
Results shown are representative of five separate experiments.
See also Figure S2.

the decrease in average neighbor number (Figure 4F). These new gation (Figure 4G) and stabilize the tissue shape change caused
cell boundaries have no preferred direction during phase I by cell elongation during phase I (Figures 4C–4E). They also
(Figure 4I). Cells undergo between one and two rounds of increase average neighbor number and the fraction of hexago-
oriented cell division that reduce cell size. Between veins 3 nally packed cells (Figure 4F).
and 4, these divisions are on average oriented 20 to the PD
axis (Figure 4H). As phase I ends, cell elongation peaks (Fig- Severing the Hinge from the Wing Blade Blocks PD
ure 4G) and cell division stops (Figure 4F). At the end of this Elongation and Misorients Cell Divisions and Neighbor
phase, tracked tissue patches have deformed in a way that Exchanges
can be accounted for by the combination of cell elongation To investigate the cellular basis of altered shear and rotation in
and oriented cell division. During phase II (about 24–32 hAPF), severed wing blades, we quantified cell elongation and the
cells assemble new contacts parallel to the PD axis (Figures orientation of neighbor exchanges and cell divisions after
4G and 4I). These oriented neighbor exchanges relieve cell elon- severing from the hinge (Figures S3C–S3G and Movie S5B).

776 Cell 142, 773–786, September 3, 2010 ª2010 Elsevier Inc.


A Figure 3. Tissue Shape Changes in Severed Wings
B
(A and B) Images taken at the indicated times from a movie
spanning 15–32 hAPF. The hinge was severed from the blade
along the line indicated.
(C–E) Locally averaged features of the cellular flow field at
22h300 APF.
80 µm (C) Flow velocity vectors. Speed corresponds to arrow length
15h30' Stbm:YFP 32h00'
D as indicated.
C 0.3 h
-1
50 µm/h (D) Rate and axis of pure shear. Shear rate corresponds to bar
length as indicated.
(E) Local rotation rate (indicated by circle size) in radians
L3 per hour.
(F) Magnitude and axis of average nematic order (yellow bars)
overlaid on a severed Stbm:YFP-expressing wing at 32 hAPF.
Green ellipse = anterior crossvein. L3, third longitudinal vein.
Results shown are representative of four separate experi-
80 µm K ments.
22h30' 22h30'
-1 -1 -1
E 0.1 h 0.05 h 0.01 h F

is altered by the changed patterns of cell rearrange-


ments, cell division, and cell elongation. This
implies that cell flows guide reorientation of PCP.

Oriented Cell Boundary Assembly Increases


22h30' anti-clockwise clockwise 32h00' Stbm:YFP the Visibility of Planar Polarity
While the majority of polarity reorientation occurs
during phase I, average PCP order increases in
Cell divisions are strongly oriented in unperturbed wings. When magnitude during phase II, as cells extend new contacts mostly
the hinge is severed from the blade, cell division continues parallel to the PD axis and develop hexagonal packing geometry
(Figure S3H), but the angle of cell division is more disperse (Figure 1F and Figures 4F and 4I). Could the increase in polar
(Figure S3J). In addition, these cells do not elongate further along order be related to these processes? To investigate this, we
the PD axis (Figure S3I), becoming instead more isotropic. While imaged PCP protein localization as new cell boundaries formed
neighbor exchanges still occur, their axis is shifted with respect (Figures 5A–5F and Movie S5C). We noted that Stbm:YFP had
to unwounded wings, lying at ±45 degrees with respect to the PD a clustered rather than a uniform distribution on cell boundaries
axis (compare Figure S3K to Figure 4I). Finally, wing severing throughout pupal development. Even at early stages, these clus-
reduces the final number of hexagonally packed cells ters contain both Stbm:YFP and Fz:CFP, suggesting that they
(62.44% ± 3.5% in wounded wings [n = 3] versus 77% ± 4.8% constitute PCP protein complexes (Figures S4A–S4C). Consis-
in unperturbed wings [n = 5]). Thus, severing the wing dramati- tent with this, Stbm:YFP clustering is reduced in fz mutant tissue
cally changes the observed patterns of cell elongation, cell divi- (Figures S4D and S4E). Individual clusters are persistent and can
sion, and rearrangements. This suggests that anisotropic stress be tracked for several hours. However, proteins within them turn
caused by hinge contraction has a key role in guiding the tissue over more rapidly (Figures S4G–S4I). Clusters do not form rapidly
flow that reshapes the wing blade. It also suggests that cell flow on new cell boundaries derived either from cell division or
increases hexagonal packing, similar to the annealing effect of neighbor exchange (Figure 5 and Movie S5C). During phase II,
cell boundary fluctuations (Farhadifar et al., 2007). new cell boundaries, expanding parallel to the PD axis within
minutes, typically remain devoid of these clusters for hours
(Figures 5A–5H). Consequently, Stbm:YFP clusters become
Severing the Hinge Alters the Global Pattern
restricted mainly to older cell interfaces on the proximal and
of Planar Polarity
distal sides of the cell. Thus, oriented cell boundary expansion
Reorientation of planar polarity in the intact wing occurs at the
and the concomitant increase in hexagonal packing order ensure
same time that the wing blade is reshaped by hinge contraction,
that PCP clusters become well separated within cells and better
suggesting that these events depend on one another. The
aligned between cells; even though PCP has already reoriented
observed pattern of clockwise and anti-clockwise tissue rotation
during phase I, oriented cell boundary expansion during phase II
tends to reorient early margin-directed polarity toward the PD
tends to increase the average nematic order (compare
axis. In addition, the shear pattern presages the final orientation
Figure S4F with Figures S1A—S1E).
of planar polarity (Figure 2H). To ask whether tissue movements
induced by hinge contraction guided polarity reorientation, we
examined the distribution of Stbm:YFP in wing blades that had A Theoretical Analysis of Polarity Reorientation
been severed from the hinge before contraction. In severed While local tissue rotation clearly implies local rotation of the
wings, the global PCP pattern differs from that of the intact polarity axis, it is less simple to understand how tissue shear
wing (compare Figure 1B with Figure 3F), suggesting that polarity affects the axis of planar polarity. Tissue shear is caused by

Cell 142, 773–786, September 3, 2010 ª2010 Elsevier Inc. 777


phase I phase II

A P D B P DC P D D P D E P D

20 µm
15 hAPF 19h30 APF 24 hAPF 28h30 APF 31h30 APF
persistent disappearing new boundary from neighbor exchange new boundary from cell division

F G
1 6 1
phase I phase II 0.32
5.9 cell number (fraction of maximum)
average neighbor number

0.8 0.8 0.28


average neighbor number
5.8

PD elongation
0.24
0.6 0.6

fraction
fraction hexagons
fraction

5.7 0.2
PD cell elongation 0.4
0.4 0.16
5.6 new cell contacts
(fraction of maximum) 0.12
0.2 0.2
5.5 cell contacts that will disappear
phase I phase II (fraction of maximum) 0.08
0 5.4 0
14 16 18 20 22 24 26 28 30 32 14 16 18 20 22 24 26 28 30 32
time (in hAPF) time (in hAPF)

H 90 90 I 90 90

0 180 0 180 0 180 0 180

270 270 270 270


phase I phase II phase I phase II
orientation of division axis orientation of new boundary assembly

Figure 4. Cell Elongation, Division, and Rearrangement


(A–E) A group of cells anterior to the posterior crossvein was tracked between 15h and 31h300 APF. Cell boundaries are color-coded depending on their fate:
red boundaries persist indefinitely, yellow boundaries disappear, green boundaries form during the movie as a result of neighbor exchange, and blue boundaries
form between daughter cells upon division.
(F and G) Quantification of cellular changes in the patch of tissue tracked in (A–E). Fraction of maximal cell number (dark blue), average neighbor number (light
blue, averaged over five frames), fraction of hexagonal cells (brown, averaged over five frames), PD cell elongation (magenta, average maximum value = 0.388 ±
0.053, n = 5), fraction of boundaries that will disappear (yellow), and the fraction of newly formed boundaries from neighbor exchange (green).
(H and I) Angular distribution of cell divisions (blue) (H) and new cell boundaries (green) (I) at the end of phase I and phase II. Yellow and magenta bars indicate
average angle of nematic order of cell division (yellow) and new cell boundary formation (magenta) (see Supplemental Theoretical Procedures). Bar length indi-
cates the degree of focus (magnitude of average nematic order, ranging from 0 to 1, see Supplemental Theoretical Procedures, 1.2). The outer circles in (H) and (I)
correspond to magnitude = 0.4. Average magnitude of phase I cell division order is 0.44+/0.12, n = 4. Average magnitude of new boundary formation order in
phase II = 0.35+/0.04, n = 4.
Data are presented as mean ± SD.
See also Figure S3.

oriented cell divisions, cell elongation, and oriented cell rear- PD axis at this time (Shimada et al., 2006). We turned to theoret-
rangements. Because microtubules have been shown to align ical analysis to explore how each of these processes might affect
with the axis of cell elongation (Cortes et al., 2006; Daga and the axis of planar polarity. Previously, we used a vertex model to
Nurse, 2008; Haase and Lew, 2007; Strauss et al., 2006), cell study the effects of proliferation on cell packing geometry
shape may directly influence the distribution of PCP within the (Farhadifar et al., 2007). Here, we add to this model a simplified
cell. Indeed, microtubules in wing epithelial cells align with the description of the dynamics of PCP order.

778 Cell 142, 773–786, September 3, 2010 ª2010 Elsevier Inc.


A 7 B Stbm:YFP A Physical Description of PCP in the Vertex Model
14
6
Cell shape, as defined by the network of adherens junctions, can
15 11
19 16 5 be described as force-balanced configurations in a vertex
8
20 17
12
3 1 model, in which an arrangement of cells is represented by a
18 13 9
4
set of cell bonds. This model takes into account cell mechanics,
2
21 hAPF 10 cell adhesion and cell division (Farhadifar et al., 2007). Our
C 7a
7b
D description of PCP order is based on attractive interactions
14
6
19 15
11 5 between proximal and distal PCP domains across cell bound-
8b
16 12 8a 3
1 aries and repulsive interactions within cells. This differs from
20 17 9 2 previous approaches (Amonlirdviman et al., 2005; Le Garrec
13 4
18 et al., 2006), in that we do not attempt to describe all details of
25 hAPF 10
14 interactions between PCP proteins, but rather consider effective
E 19 11
6 7b F
16
15 8b 5 interactions between proximal and distal domains. Each cell
8a
20
17
12
9 3
1
bond i, separating cells a and b, is assigned two variables, sai
18
13
10
4 2 and sbi , which describe the level of PCP proteins on either side
8 µm of this bond (Figure 6A). The variables sai can take continuously
29 hAPF varying values; sai = 1 corresponds to a high level of proximal
G 90 H 90 proteins (blue), while sai =  1 corresponds to a high level of
distal proteins (red). We introduce a potential function for interac-
tions of PCP domains:
0 180 0 180
X X X
E = J1 sai sbi  J2 sai saj  J3 3a $Qa ; (1)
i fi;jg a
270 270
I where J1 and J2 are PCP interaction parameters and J3
describes coupling of PCP to cell elongation (for J3 = 0, PCP
does not couple to cell elongation). For J1 >0 the potential favors
J K interactions between proximal and distal proteins across cell
bonds. For J2 >0, accumulation of proximal and distal proteins
on neighboring bonds within a given cell is disfavored. For
J3 >0, alignment of PCP with the long axis of the cell is favored:
L M this is motivated by the idea that aligned microtubules bias the
transport of PCP proteins (Shimada et al., 2006). The first sum
is over all bonds i, the second sum is over all pairs fi; jg of adja-
cent bonds in the network, and the third sum is over all cells a.
N O
The cell shape and PCP nematic order are described by the
tensors 3a and Qa , respectively (Supplemental Theoretical
Procedures, 5). We introduce a time evolution of PCP level on
P Q cell bonds: dsai =dt =  gvE=vsai , where t is time and g is a kinetic
coefficient that determines the PCP relaxation time t = 1=ðgJ1 Þ.
We solve these dynamic equations numerically, imposing two
R S constraints: (1) in each cell, amounts of proximal and distal
proteins are equal and (2) these amounts do not change with
time (Supplemental Theoretical Procedures, 5).
T U

Global Polar Order Generated by Network Growth


To examine how different types of shear reorient polarity, we
must start from a cellular configuration in the vertex model with
Figure 5. Cell Rearrangements Increase the Visibility of PCP
(A–F) Images from a 21–29 hAPF movie of a Stbm:YFP-expressing wing. In (A),
(C), and (E), cell identity is indicated by numbers and cell boundaries are (see Figure 4). Permanent boundaries (red in [N] and [O]) carry PCP clusters
colored according to their fate as in Figure 4. (B), (D), and (F) show PCP that persist for 8 hr. New boundaries (green in [N] and [O]) remain devoid of
nematics for each cell (yellow). Stbm:YFP clusters.
(G and H) Angular distribution of newly formed (green, [G]) and permanent (P-U) Kymographs of a nondividing cell (P), (R), and (T) or a dividing cell (Q), (S),
boundaries (red, [H]). and (U) between 20-21 hAPF (300 frame rate). In (R) and (S), each boundary is
(I) Each kymograph line depicts the linearized perimeter of a selected cell at identified by a random color. Colors in (T) and (U) indicate boundary fate (see
time t. Figure 4). After cell division (Q), (S), and (U) depicts the joint perimeter of the
(J–O) Perimeter intensity kymographs for two cells of the movie (frame rate = two daughter cells and the boundary created by cell division (blue in [U])
60 , movie length = 8 hr) shown in (A–F). In (L) and (M), each boundary is separately.
identified by a random color. Colors in (N) and (O) indicate boundary fate See also Figure S4.

Cell 142, 773–786, September 3, 2010 ª2010 Elsevier Inc. 779


Figure 6. Theoretical Analysis of Shear and
A B
α Rotation on the Reorientation of Planar
Polarity
σ iα (A) Schematic representation of PCP in the vertex
σ iβ
model. The variables, sai and sbi , represent the level
β and type of PCP domains on bond i shared by cells
a and b.
(B) Cell packing with PCP order generated by
simulated proliferation in the vertex model with
C D PCP dynamics, starting from 36 randomly polar-
ized cells with kdt = 0.01, J2/J1 = 0.5 and J3 = 0.
Arrows in (B)–(D) show the direction of polarity.
(C and D) Reorientation of polar order by shear due
to external forces (C) or oriented cell division (D).
Starting from the network (B), and simulating shear
along the horizontal axis, leads to the networks
shown. Parameter values for (C) are kdt = 0.01
and J3/J1 = 0.05. For (D), kdt = 0.01, J3 = 0 and
E F 3 rounds of cell division were simulated. The
shear generated corresponds to an aspect ratio
of 4 in (C) and 1.5 in (D).
(E and F) Calculations of polarity reorientation
based on Equation 2 and measured patterns of
shear and rotation. Starting with the observed
early polarity (E), the time evolution described by
Equation 2 with n =  3 generates a final PD
pattern (F). The wing is the same as that shown
in Figures 2D–2L.
(G and H) same procedure as in (E and F) applied to
the wounded wing shown in Figure 3 with n = 2. We
start with an initial pattern mimicking the early
polarity (G).
See also Figure S5.

G H

ure S5A, and Movie S6A). This is consis-


tent with the observation of global PCP
patterns in the growing wing disc
(Classen et al., 2005), and suggests that
these patterns arise early during
development.

Shear Reorients the Polarity Axis in


the Vertex Model
Starting with a globally polarized
network, we used the vertex model to
examine how different types of shear
could influence the average polarity
globally aligned PCP variables. To generate such a configuration orientation of groups of cells. We first studied the effects of
without an external bias is nontrivial (Burak and Shraiman, 2009). shear caused by cell elongation and oriented cell rearrange-
Surprisingly, a simple and general way to generate large ments. In our simulations, we induced pure shear at a rate ks
networks with long-range polarity is to start from a small group by forcing the network to elongate along one axis at constant
of cells with an initially random PCP configuration (random total area (Supplemental Theoretical Procedures, 6). We
values of sai , Supplemental Theoretical Procedures, 6). This oriented the initial axis of average polarity at an angle of 45
network is then expanded by simulating stochastic cell divisions degrees to this shear axis (Figure 6B). As the network is
at a rate kd . Simultaneously, the dynamic equations for the PCP deformed, cells elongate and undergo T1 transitions that are
variables are solved. Interactions of PCP variables generate oriented along the stretch axis. We find that shear reorients
local order, which aligns over the whole network when it is polarity either parallel or perpendicular to the shear axis, de-
still small. Order is maintained during growth and leads to glob- pending on the value of ks t and on the strength J3 of the
ally aligned polarity in the resulting large networks if the PCP coupling of cell shape to PCP distribution (Figures 6C, Figures
relaxation time t is sufficiently fast (kd t<1, Figures 6B, Fig- S5B–S5D, and Movies S6B—S6C). Reorientation occurs during

780 Cell 142, 773–786, September 3, 2010 ª2010 Elsevier Inc.


a characteristic time 1=j2nks j, where n is a dimensionless coef- to calculate the corresponding time-dependent reorientation of
ficient. We define n to be negative if polarity aligns with the polar order. Local rates of rotation and shear were obtained
shear axis, and positive if it aligns perpendicular to this axis. from the experimentally determined flow field between 15 and
Positive values of J3 promote the alignment of the polarity 32 hAPF. To ask to what extent local rotation alone could repro-
vector with the shear axis, as do faster relaxation times t of duce the observed polarity reorientation, we first considered
PCP (Figures S5B–S5D). Network configurations generated by n = 0, i.e., shear does not influence reorientation. In this case
this process are irregular and cells are stretched. We relaxed polarity realigns toward the PD axis, however, the final average
these networks by introducing fluctuations of cell bond tension polar order achieved is only 0.76 (77% of the experimental value
(Supplemental Theoretical Procedures, 6). This reduced cell at 32 hAPF). Thus, local rotation can account for only part of the
stretch and increased both the fraction of hexagons and the observed reorientation of the polarity axis.
magnitude of average polar order, similar to what is observed If n is nonzero, shear also influences reorientation. Repeating
during phase II in vivo (Figures 4F and 4G, and Figure S5E). this calculation for different values of n shows that the final
We next studied whether shear generated by oriented cell divi- average polar order is largest for nz  3, reaching a value of
sions reorients polarity. We imposed cell divisions in the vertex 0.90 (92% of the experimental value, Figures S5G–S5J). For n
model at rate kd oriented at an angle of about 45 degrees to within the range 10<n<  1 the polarity field reorients robustly
the initial polarity axis (Supplemental Theoretical Procedures, to reach an average polar order of at least 0.85 (Figure S5K,
6). Oriented cell division generates shear parallel to the cell divi- 6E,F and Movie S6E), tolerating small deviations from the exper-
sion axis (defined as the line connecting the centers of the two imentally determined pattern of initial conditions (Figure S5K).
daughter cells) at a rate ks = mkd , where m depends on network Note that the effects of local shear on polarity orientation depend
mechanics. We find again that polarity is reoriented either on PCP domain interactions and dynamics, as captured in the
parallel or perpendicular to the shear axis and that the dynamics vertex model. In the calculations using Equation 2, these effects
of reorientation can be characterized by the coefficient n defined are represented via the value of the parameter n. However, in
above (Figure 6D, Figure S5F, and Movie S6D). Again, faster writing Equation 2 we choose to neglect interactions of average
relaxation times t of PCP promote alignment of the polarity polar order between neighboring patches of tissue to highlight
vector with the shear axis (Figure S5F). the role of shear and rotation. We expect such additional interac-
tions could account for the remaining deviation between the
solution to Equation 2 and the observed final polarity. Note
An Effective Description of Large-Scale Polarity also that positive values of n produce a final polarity pattern
Reorientation by Flow with lower polar order than rotation alone (n = 0) (Figure S5K).
The reorientation of average polarity by shear flow that we This analysis suggests that shear caused by the combination
observe in the vertex model is well known in complex fluids of oriented cell divisions, cell elongation and cell rearrangement
such as liquid crystals. In these systems, polarity reorientation plays an important role in aligning polarity parallel to the shear
is determined by local rotation and shear rates of the flow field axis by a process that is characterized by a negative value of n.
and can be described by the following equation (de Gennes The close agreement between the polarity pattern generated
and Prost, 1993; Joanny et al., 2007) by Equation 2 (Figure 6F) and experimental observations
vq (Figure 1B) provides strong evidence that tissue remodelling is
= nks sin2q + u: (2) responsible for reorienting the axis of planar polarity.
vt
To ask whether Equation 2 could describe the polarity defects
Here, u is the local rotation rate, and ks is the rate of pure shear observed when the hinge and wing blade are severed, we used it
(Supplemental Theoretical Procedures, 7). The orientation of to calculate polarity reorientation given the shear and rotation
polarity is described by the angle q, relative to the axis of pure patterns measured for these wings. For wounded wings, we
shear. The coefficient n describes how pure shear influences find that Equation 2 describes the main features of polarity reor-
reorientation of polarity. In the absence of rotation, u = 0, pure ientation provided that n is positive (Figures 6G and 6H, Figures
shear aligns polarity either with the shear axis ðq = 0Þ or perpen- S5L and S5N, and Movie S6F). This implies that, in wounded
dicular to it ðq = p=2Þ depending on the sign of n. In liquid crystals, wings, shear tends to align polarity perpendicular to the shear
the value of n depends on molecular geometries and intermolec- axis, rather than parallel to it, as it does in unperturbed wings.
ular interactions (de Gennes and Prost, 1993). In the absence of Why should shear affect polarity differently in wounded wings?
pure shear, ks = 0, local tissue rotations reorient polarity as In simulations of the vertex model, we observed that shear gener-
vq=vt = u. ated by oriented cell divisions and by external stresses reorients
Equation 2 for u = 0 describes the main features of polarity polarity with different characteristics (Figures S5B–S5D and S5F).
reorientation by pure shear in the vertex model described above. The value of n thus depends on the types of cell shape changes
The coefficient n that characterizes how shear affects reorienta- and rearrangements occurring during shear. In wounded wings,
tion in the vertex model is the same as the parameter n in Equa- where mechanical boundary conditions are perturbed, the re-
tion 2. We can thus use Equation 2 to determine quantitatively modeling processes that dominate shear differ from those in
how the observed patterns of rotation and shear reorient the unperturbed wings (compare Figures S3C–S3K with Figure 4).
global pattern of planar polarity in the wing. In early pupal wings, fz clones nonautonomously reorient
Starting with the observed polarity pattern at 15 hAPF, we polarity of adjacent wild-type tissue lying between the clone
locally applied Equation 2, with n being the only free parameter, and the wing margin (Figure S1N and S1O). But after remodeling,

Cell 142, 773–786, September 3, 2010 ª2010 Elsevier Inc. 781


the location of affected wild-type tissue is distal to the fz clone sion orientation in both larval and pupal wings. Simulations
(Vinson and Adler, 1987). We wondered whether the shear and suggest that both cell elongation and oriented cell divisions
rotation observed during remodeling could account for reposi- can reorient polarity (Figures 6C and 6D). Interestingly, both
tioning of affected wild-type tissue. To investigate this, we first are perturbed in ds mutants.
simulated growth in the vertex model mimicking the situation Ds influences both larval and pupal epithelial dynamics—so
of a fz mutant clone. Starting with a network of 36 cells, we chose which is most important for development of PD polarity? To
a single cell and set sai = 1 on all its bonds (corresponding to loss investigate this, we altered Ds levels in the posterior compart-
of Fz domains). We then simulated growth of the network while ment of the wing either throughout development or only during
simultaneously solving the dynamic equation of the PCP network larval growth or after pupariation. Overexpressing Ds throughout
(Supplemental Theoretical Procedures, 5), keeping sai = 1 in the wing development causes strong polarity defects in both poste-
simulated clone (indicated in blue). In these simulations, the rior and anterior compartments (Figure S6A). Overexpressing Ds
polarity pattern becomes distorted on one side of the clone as during larval or pupal development alone causes much milder
domains with sai <0 (red) are positioned to face the clone polarity defects (Figures S6A–S6C). We obtain complementary
(compare Figure S5O with S5Q). These simulations generate results by lowering Ds levels. Inducing RNAi against Ds in the
PCP patterns that mimic domineering nonautonomy observed posterior compartment throughout development perturbs
in vivo for fz and stbm mutant clones. Such patterns suggest polarity strongly in both anterior and posterior compartments
the interesting possibility that domineering nonautonomy may (Figure S6D). Ds knock-down during larval or pupal development
arise during growth of the wing and may explain why the polarity alone causes milder polarity defects (Figures S6E and S6F).
of wild-type tissue is already reoriented by fz clones at 15 hAPF. These data suggest that Ds is needed both during growth and
We use these patterns as initial conditions to study reorientation subsequent epithelial remodelling to ensure proper alignment
during shear. of PCP with the PD axis. Ds loss or overexpression throughout
To investigate the effects of shear and rotation on the per- development causes cumulative defects stronger than those
turbed PCP polarity patterns, we solve Equation 2 with n =  3 caused by stage-specific perturbation.
for both networks with and without simulated clones, and an Loss of Ds during pupal stages may perturb PD polarity by
initial average polarity axis lying 45 to the horizontal (mimicking reducing cell elongation or misorienting cell divisions during
margin-oriented polarity). We impose shear and rotation at remodelling. How could loss of Ds during larval growth affect
constant rate, with the shear axis oriented horizontally (corre- evolution of PD polarity? One possibility is that Ds-dependent
sponding to the PD axis of the wing) (Supplemental Theoretical oriented cell divisions in the wing disc guide development of
Procedures, 8). This reorients the global polarity of both net- early margin-oriented polarity; complex global PCP patterns
works and shifts the perturbed region toward the ‘‘distal’’ side are as already seen in the third instar, and prepupal wings
of the clone (compare Figure S5P and R) consistent with exper- show a pattern consistent with margin-oriented polarity (Classen
iments (Vinson and Adler, 1987). et al., 2005). To test this, we quantified the Stbm:YFP pattern in
ds wings at 15 hAPF. The pattern of average nematic order in ds
Ds Guides Global Planar Cell Polarity by Influencing wings deviates from that of wild-type over large regions,
Epithelial Dynamics although PCP domains are well aligned locally, (compare
Perturbing the Ft/Ds pathway misaligns PCP domains, but the Figure 7C and Figure 1A). These observations together with
underlying mechanism is controversial (Axelrod, 2009; Casal simulations suggest that the larval pattern of oriented cell divi-
et al., 2006; Lawrence et al., 2007; Ma et al., 2003; Tree et al., sions contributes to development of margin directed polarity.
2002a). These mutations also cause shorter, broader wings – Loss of Ds may perturb the early polarity pattern indirectly by
at least in part by perturbing oriented cell divisions in larval discs misaligning larval cell divisions.
(Baena-Lopez et al., 2005). To investigate whether Ds also influ- To ask whether abnormal early polarity in combination with the
enced pupal epithelial remodelling, we analyzed time-lapses of altered flow pattern could in principle produce normal PD
ds pupal wings expressing ECadherin:GFP. At 15 hAPF, ds polarity, we started with the altered polarity observed in early
wings are already misproportioned; the hinge is overgrown and ds wings and applied the mutant velocity field using Equation
the wing blade is short and broad (Figure 7A), consistent with 2. For all values of n, polarity fails to orient as it does in wild-
altered larval growth. Nevertheless, the hinge contracts as it type (average polar order < 0.67, compare Figure S5M with
does in wild-type, reducing its area about twofold (Figures 7A S5K). Taken together, these studies indicate that development
and 7B). Thus, Ds is not required for hinge contraction. Calcu- of PD polarity depends both on correct initial polarity and on
lating shear and rotation over the whole blade is difficult in ds tissue rotation and shear produced during hinge contraction.
mutants because the epithelium folds; in regions that can be Although Ds is expressed at high levels in the hinge and at low
quantified, rotation rates are smaller than in wild-type (Figures levels in the wing blade, this discontinuity along the PD axis is not
S2G–S2I0 and S2L and Movie S7A). However, differences in required to direct PD orientation of PCP domains. Uniform Ds
the shear patterns could not be resolved (Figures S2D–S2F0 ). expression rescues polarity in a ds mutant background (Mata-
A clearer picture emerged from analyzing ds mutants at the katsu and Blair, 2004 and Figure S6G-I). Furthermore, lowering
cellular level (Figures 7D–7H and Movie S5D). PD cell elongation Ds levels in the wing blade perturbs polarity more than knock-
is reduced in ds wings (compare Figure 4G and Figure 7J), and down in the hinge (Figure S6K,L). In contrast, discontinuities in
the axis of cell division is less focused than in wild-type (compare Ds levels along the AP axis do perturb polarity at a distance;
Figures 4H and 7K). Thus Ds is required to tightly focus cell divi- overexpression or loss of Ds in the posterior compartment

782 Cell 142, 773–786, September 3, 2010 ª2010 Elsevier Inc.


A 15h30' APF B 30 hAPF C

2 2 2 2
212*103µm 123*103µm 123*103µm 146*103µm 15 hAPF
phase I phase II

D P D
E P D
F P D
G P D
H P D

20 µm
15h45 APF 19h30 APF 24 hAPF 28h30 APF 31h APF
persistent disappearing new boundary from neighbor exchange new boundary from cell division

I 6
J 1
1
phase I phase II 0.32
5.9 cell number (fraction of maximum)
average neighbor number

0.8 0.8 0.28


average neighbor number
5.8

PD elongation
0.24
0.6 0.6
fraction

fraction hexagons
fraction

5.7 0.2
PD cell elongation 0.4
0.4 0.16
5.6 new cell contacts
(fraction of maximum) 0.12
0.2 0.2
5.5 cell contacts that will disappear
phase I phase II (fraction of maximum) 0.08
0 5.4 0
14 16 18 20 22 24 26 28 30 32 14 16 18 20 22 24 26 28 30 32
time (in hAPF) time (in hAPF)

K 90 90 L 90 90

0 180 0 180 0 180 0 180

270 270 270 270


phase I phase II phase I phase II
orientation of division axis orientation of new boundary assembly

Figure 7. Early Polarity, Cell Elongation, Division, and Rearrangement Are Perturbed in ds05142 Wings
(A and B) A ds05142, ubi-ECad:GFP wing at 15h300 and 30 hAPF. The hinge is colored blue and the blade red. Numbers indicate blade and hinge areas in mm2.
(C) Average nematic order in a ds05142,act-stbm:YFP/ ds05142 wing at 15 hAPF (compare to Figure 1A).
(D–H) A group of cells anterior to the posterior crossvein in a ds05142, ubi-ECad:GFP wing was tracked between 15h450 and 31 hAPF. Cell boundaries are color-
coded as in Figure 4A.
(I and J) Quantification of cellular changes in the patch of tissue tracked in (D–H). Fraction of maximal cell number (dark blue), average neighbor number (light blue,
averaged over 8 frames), fraction of hexagonal cells (brown, averaged over 8 frames), PD cell elongation (magenta, average maximum values = 0.243, n = 2
(0.2245, 0.261), fraction of boundaries that will disappear (yellow), and fraction of new boundaries resulting from neighbor exchange (green).
(K and L) Angular distribution of cell divisions (blue) (K) and new boundaries (green) (L) at the end of Phase I and Phase II. Yellow and magenta bars indicate
average angle of nematic order of cell division (yellow) and new cell boundaries (magenta) (see Supplemental Theoretical Procedures and Figure 4). Average
magnitude of phase I cell division order is 0.145 (0.180, 0.109 n = 2). Average magnitude of new boundary formation order in phase II = 0.338 (0.278, 0.398;
n = 2).
See also Figure S6.

Cell 142, 773–786, September 3, 2010 ª2010 Elsevier Inc. 783


nonautonomously perturbs polarity anteriorly (Figures S6A– prepupal wings, and showed that even larval wing discs show
S6F). Altering Ds levels changes the response of wing epithelial a global pattern of PCP (Classen et al., 2005). Thus PCP domains
cells to hinge-dependent pulling forces (Figure 7). We therefore are always polarized, but their global orientation evolves dynam-
considered the possibility that local changes in cell rearrange- ically during development.
ment and elongation could alter large-scale cellular flows and What causes global reorientation of PCP during phase I, and
thus the patterns of shear and rotation in wild-type regions. the subsequent increase in the magnitude of polar order during
Indeed, Ds overexpression in the posterior compartment nonau- phase II of wing-blade remodeling? During phase I, margin-
tonomously reverses local tissue rotation in anterior cells near oriented polarity is reoriented by specific patterns of local rotation
the compartment boundary (compare Figures S6M–S6O, Fig- and shear due to cell flows caused by hinge contraction. Local
ures 2J–2L, and Movie S7B). These observations suggest that, tissue rotation and shear reorient PCP domains by simple phys-
to generate normal flow patterns in response to hinge contrac- ical rules (see Equation [2] and Figure S7) similar to those that re-
tion, cells in the anterior and posterior compartments must react orient molecular order in liquid crystal hydrodynamics. A signifi-
similarly to mechanical stress. cant fraction of PCP reorientation is produced by local tissue
rotation alone. However, tissue shear also helps rotate polarity
DISCUSSION toward the shear axis. Simulations suggest that shear caused
by oriented cell divisions, cell rearrangements and PD cell elonga-
The mechanisms that couple tissue shape to the planar polarity tion can all contribute to polarity reorientation. One mechanism
of constituent cells are not well understood. Here, we describe that may underlie the effect of cell elongation on PCP is the
a novel morphogenetic event that both shapes the wing of tendency of microtubules to align with the long axis of the cell.
Drosophila, and orients global planar polarity of wing cells. We Indeed microtubules are aligned with the PD axis of wing epithelial
show that the hinge region contracts shortly after the prepupal cells and are essential for delivery of PCP proteins to the cortex
to pupal transition. This contraction not only shapes the wing (Shimada et al., 2006). Significantly, under conditions where
hinge, but also exerts anisotropic tension on the adjacent normal PD polarity does not develop (e.g., in ds mutant wings
wing-blade region, causing it to elongate and narrow. Reshaping or in severed wings), oriented cell division, cell rearrangements
of the wing blade occurs as constituent cells flow proximally with and cell elongation are disturbed. During phase II, oriented cell
different velocities, generating reproducible patterns of flow, boundary rearrangements increase hexagonal packing geometry
shear and rotation. These patterns result from a combination of and improve PCP order. Simulations suggest this may occur via
oriented cell division, cell elongation and neighbor exchanges an annealing process that relaxes irregular cell packing to a
that are guided by the anisotropic stresses. more ordered hexagonal lattice with improved PCP order.
This scenario, where externally generated forces have an The atypical Cadherins Ft and Ds are needed to evolve the
important role in epithelial remodeling differs from other con- global PD polarity pattern. Exactly how they influence the global
vergent-extension events where autonomous cell move- pattern has been controversial (Axelrod, 2009; Lawrence et al.,
ments actively drive tissue shape changes (Bertet et al., 2004; 2007). It was proposed that this pathway provides a PD polarity
Blankenship et al., 2006; Keller et al., 2008; Rauzi et al., 2008; cue that depends on higher relative levels of Ds expression in the
Zallen and Wieschaus, 2004). Externally induced epithelial hinge. This inhomogeneity was thought to generate intracellular
stretching has been proposed to provoke stress-relieving rear- asymmetry of Ft/Ds heterodimers throughout the wing, directly
rangements during Drosophila germband extension (Butler generating a small bias in Fz activity within each cell that could
et al., 2009), and in the amphibian ectoderm (Keller et al., then be amplified to produce strong and stable alignment of
1992, 2008). But in these cases movements of the underlying PCP domains with the PD axis (Ma et al., 2003; Tree et al.,
mesoderm, rather than contraction of adjacent epithelial cells, 2002a). But the fact that uniform Ds expression suffices to
provides the external force. The Drosophila pupal wing will rescue polarity in ds wings argues against this view (Matakatsu
be a powerful system for uncovering molecular mechanisms and Blair, 2004) (Figures S6G–S6I). Furthermore, Ft and Ds do
underlying stress-induced epithelial remodelling. not appear to act directly on core PCP proteins in the Drosophila
The mechanisms specifying global PCP domain alignment abdomen (Casal et al., 2006). We propose that the Ft/Ds
have been elusive and controversial (Axelrod, 2009; Casal pathway influences the global PCP pattern, not by generating
et al., 2006; Lawrence et al., 2007; Ma et al., 2003; Tree et al., intracellular asymmetries that directly regulate PCP signaling,
2002a). It had been thought that PCP proteins in the Drosophila but indirectly through its effects on epithelial dynamics during
wing are initially randomly distributed, but become intracellularly both larval and pupal stages. Time-controlled loss of Ds shows
polarized along the PD axis starting approximately 10 hr before that it is required throughout development for evolution of PD
hair formation (Strutt and Strutt, 2005; Vladar et al., 2009; Wu polarity (Figures S6D–S6F) (see also (Matakatsu and Blair,
and Mlodzik, 2009; Zallen, 2007). This led to the search for 2004)). During larval stages, Ds may guide the development of
a global polarizing signal that operated at this time. Here, we early margin-oriented polarity through its influence on oriented
show that PD orientation of PCP complexes does not arise growth; our simulations suggest that oriented cell division
from a random distribution; rather it evolves from a different strongly influences the global axis of planar polarity. Later, during
global polarity pattern present at the prepupal-pupal transition. hinge contraction and wing-blade remodeling, Ds is required
At this stage, PCP domains are organized such that Fz-contain- for the oriented cell division and cell elongation that occur in
ing domains face the wing margin rather than distally. Our response to anisotropic stresses. Both processes influence the
previous work identified a similar global polarity pattern in polarity axis in vertex model simulations.

784 Cell 142, 773–786, September 3, 2010 ª2010 Elsevier Inc.


In future, it will be interesting to examine the roles of aniso- Grace. Images were vectorized using Batik. Figures were composed with
tropic growth and local morphogen signaling in establishing Adobe Illustrator.
the global PCP pattern as wing discs grow. Our theoretical anal-
ysis shows that large scale planar polarity can be stably main- SUPPLEMENTAL INFORMATION
tained during growth even in the absence of global polarizing
Supplemental Information includes Extended Experimental Procedures,
signals. This is possible because polarity easily aligns over
Supplemental Theoretical Procedures, seven figures, and seven movies and
a few cell diameters without orientation defects, and subsequent can be found with this article online at doi:10.1016/j.cell.2010.07.042.
cell rearrangements due to cell division do not destroy this order
during growth. Thus, polarity that was initially established when ACKNOWLEDGMENTS
the tissue was small can be expanded during growth, and any
anterior-posterior or dorsal-ventral boundary-derived signal This work represents a truly collaborative effort. Each author has contributed
would not need to act directly over long distances. significantly to the findings and regular group discussions guided the develop-
ment of the ideas presented here. The manuscript was written jointly by all
The coupling of planar polarity to epithelial dynamics is
authors. S.E., A.S., J.R., R.F., and F.J. were supported by the Max Planck
a robust and flexible mechanism for coordinating tissue shape Gesellschaft. B.A. was funded by the Fondation pour la Recherche Médicale.
with planar polarity of constituent cells and may generalize to D.S. acknowledges the Natural Sciences and Engineering Research Council of
other systems. Indeed, mutations in the Wnt pathway perturb Canada and the German Academic Exchange Service. J.R. was supported by
both convergent extension and the planar orientation of sensory a predoctoral fellowship from the Boehringer Ingelheim Fonds. We thank Tony
hair cells in the mouse cochlea (Wang et al., 2005). Furthermore, Hyman and Stephan Grill for use of the laser cutting microscope. We are grate-
ful to Stephan Grill, Ewa Paluch, Elisabeth Knust, and Carl-Philipp Heisenberg
the planar polarity of sensory hair cells in the lateral line organ of
for critical comments on the manuscript. We thank David Strutt, Seth S.Blair,
zebrafish is oriented by the direction of cell migration rather than VDRC and the Bloomington Stock Center for providing fly stocks, Barry Dick-
long-range secreted cues (Lopez-Schier et al., 2004). Our theo- son for DNA constructs, Julia Gäbel for help with cloning, Sven Ssykor for
retical analysis suggests that shear caused by stretch-induced transgenic injections.
rearrangements or by oriented cell divisions can reorient polarity
either perpendicular or parallel to the shear axis, depending on Received: August 28, 2009
the relative rates of shear and PCP turnover. Thus, this process Revised: May 11, 2010
Accepted: July 23, 2010
is also highly versatile, and could be a fundamental principle of
Published: September 2, 2010
tissue organization.
REFERENCES
EXPERIMENTAL PROCEDURES
Adler, P.N., Charlton, J., and Liu, J. (1998). Mutations in the cadherin super-
Imaging family member gene dachsous cause a tissue polarity phenotype by altering
Pupae were prepared for imaging as described (Classen et al., 2008). Images frizzled signaling. Development 125, 959–968.
were acquired with a Leica TCS-SP2 or an Olympus FV-1000 microscope. Amonlirdviman, K., Khare, N.A., Tree, D.N., Chen, W., Axelrod, J., and Tomlin,
203 and 403 oil immersion objectives were used to follow cell flows and C.J. (2005). Mathematical modeling of planar cell polarity to understand domi-
global morphological changes occurring in the wing. A 633 oil immersion neering non-autonomy. Science 307, 423–426.
objective was used to image pupal wings with cellular resolution. Imaging Axelrod, J.D. (2009). Progress and challenges in understanding planar cell
was performed at 25 ± 2 C, or at 29 C using a Bachhoffer chamber. polarity signaling. Semin. Cell Dev. Biol. 20, 964–971.
Baena-Lopez, L.A., Baonza, A., and Garcia-Bellido, A. (2005). The orientation
Wing Wounding of cell divisions determines the shape of Drosophila organs. Curr. Biol. 15,
Wings were wounded by gently scratching the pupal cuticule in the hinge 1640–1644.
region of the wing using forceps just before imaging (at around 15 hAPF). Bertet, C., Sulak, L., and Lecuit, T. (2004). Myosin-dependent junction remod-
elling controls planar cell intercalation and axis elongation. Nature 429,
667–671.
Image Analysis and Figures
Z projections were created with ImageJ. For image analysis, we developed Blankenship, J.T., Backovic, S.T., Sanny, J.S., Weitz, O., and Zallen, J.A.
‘‘packing analyzer v2.0’’, which measures cell areas, perimeter, packing, elon- (2006). Multicellular rosette formation links planar cell polarity to tissue
gation, polarity, cell divisions and tracks the cells and their boundaries. Briefly, morphogenesis. Dev. Cell 11, 459–470.
images are segmented using the watershed algorithm (Vincent and Soille, Bryant, P.J., Huettner, B., Held, L.I., Jr., Ryerse, J., and Szidonya, J. (1988).
1991). Each watershed catchment’s basin is defined as a cell. Vertices are Mutations at the fat locus interfere with cell proliferation control and epithelial
defined as pixels shared between three or more cells. Cell boundaries are morphogenesis in Drosophila. Dev. Biol. 129, 541–554.
identified as pixels shared by exactly two cells. Cell boundaries smaller than Burak, Y., and Shraiman, B.I. (2009). Order and stochastic dynamics in
three pixels are reclassified as vertices. The polygon class is determined by Drosophila planar cell polarity. PLoS Comput. Biol. 5, e1000628.
counting cell vertices. Cell area is defined as the number of pixels in each
Butler, L.C., Blanchard, G.B., Kabla, A.J., Lawrence, N.J., Welchman, D.P.,
cell. Cell perimeter is defined as the sum of the distance between all the pixels
Mahadevan, L., Adams, R.J., and Sanson, B. (2009). Cell shape changes indi-
surrounding a cell. To track cells, we assign a unique identity to each in the first
cate a role for extrinsic tensile forces in Drosophila germ-band extension. Nat.
movie frame. Cells are then re-identified in the next images based on their
Cell Biol. 11, 859–864.
positions and neighborhoods. Boundaries are defined by the two cells that
share them. Division orientation is defined by a line connecting the centers Casal, J., Lawrence, P.A., and Struhl, G. (2006). Two separate molecular
of the two daughter cells, and the angle it makes with the PD axis (which is systems, Dachsous/Fat and Starry night/Frizzled, act independently to confer
plotted as horizontal). planar cell polarity. Development 133, 4561–4572.
Movies created using our software were compressed using FFmpeg or Chen, W.S., Antic, D., Matis, M., Logan, C.Y., Povelones, M., Anderson, G.A.,
Quicktime Pro. Graphs were created using Microsoft Excel, Gnuplot and Nusse, R., and Axelrod, J.D. (2008). Asymmetric homotypic interactions of the

Cell 142, 773–786, September 3, 2010 ª2010 Elsevier Inc. 785


atypical cadherin flamingo mediate intercellular polarity signaling. Cell 133, Matakatsu, H., and Blair, S.S. (2004). Interactions between Fat and Dachsous
1093–1105. and the regulation of planar cell polarity in the Drosophila wing. Development
Cho, E., and Irvine, K.D. (2004). Action of fat, four-jointed, dachsous and dachs 131, 3785–3794.
in distal-to-proximal wing signaling. Development 131, 4489–4500. Rauzi, M., Verant, P., Lecuit, T., and Lenne, P.F. (2008). Nature and anisotropy
of cortical forces orienting Drosophila tissue morphogenesis. Nat. Cell Biol. 10,
Clark, H.F., Brentrup, D., Schneitz, K., Bieber, A., Goodman, C., and Noll, M.
1401–1410.
(1995). Dachsous encodes a member of the cadherin superfamily that controls
imaginal disc morphogenesis in Drosophila. Genes Dev. 9, 1530–1542. Seifert, J.R., and Mlodzik, M. (2007). Frizzled/PCP signalling: a conserved
mechanism regulating cell polarity and directed motility. Nat. Rev. Genet. 8,
Classen, A., Anderson, K., Marois, E., and Eaton, S. (2005). Hexagonal Packing
126–138.
of Drosophila Wing Epithelial Cells by the Planar Cell Polarity Pathway. Dev.
Cell 9, 805–817. Shimada, Y., Yonemura, S., Ohkura, H., Strutt, D., and Uemura, T. (2006).
Polarized transport of Frizzled along the planar microtubule arrays in
Classen, A.K., Aigouy, B., Giangrande, A., and Eaton, S. (2008). Imaging Drosophila wing epithelium. Dev. Cell 10, 209–222.
Drosophila pupal wing morphogenesis. Methods Mol. Biol. 420, 265–275.
Simons, M., and Mlodzik, M. (2008). Planar cell polarity signaling: from fly
Cortes, S., Glade, N., Chartier, I., and Tabony, J. (2006). Microtubule self-orga- development to human disease. Annu. Rev. Genet. 42, 517–540.
nisation by reaction-diffusion processes in miniature cell-sized containers and
Strauss, B., Adams, R.J., and Papalopulu, N. (2006). A default mechanism of
phospholipid vesicles. Biophys. Chem. 120, 168–177.
spindle orientation based on cell shape is sufficient to generate cell fate diver-
Daga, R.R., and Nurse, P. (2008). Interphase microtubule bundles use global sity in polarised Xenopus blastomeres. Development 133, 3883–3893.
cell shape to guide spindle alignment in fission yeast. J. Cell Sci. 121, 1973– Strutt, H., and Strutt, D. (2002). Nonautonomous planar polarity patterning
1980. in Drosophila: dishevelled-independent functions of frizzled. Dev. Cell 3,
de Gennes, P.G., and Prost, J. (1993). The Physics of Liquid Crystals, Second 851–863.
Edition (Gloucestershire, UK: Clarendon Press). Strutt, H., and Strutt, D. (2005). Long-range coordination of planar polarity in
Farhadifar, R., Roper, J.C., Aigouy, B., Eaton, S., and Julicher, F. (2007). The Drosophila. Bioessays 27, 1218–1227.
Influence of Cell Mechanics, Cell-Cell Interactions, and Proliferation on Epithe- Strutt, H., and Strutt, D. (2008). Differential stability of flamingo protein
lial Packing. Curr. Biol. 17, 2095–2104. complexes underlies the establishment of planar polarity. Curr. Biol. 18,
Garoia, F., Grifoni, D., Trotta, V., Guerra, D., Pezzoli, M.C., and Cavicchi, S. 1555–1564.
(2005). The tumor suppressor gene fat modulates the EGFR-mediated prolifer- Tree, D.R., Ma, D., and Axelrod, J.D. (2002a). A three-tiered mechanism for
ation control in the imaginal tissues of Drosophila melanogaster. Mech. Dev. regulation of planar cell polarity. Semin. Cell Dev. Biol. 13, 217–224.
122, 175–187. Tree, D.R., Shulman, J.M., Rousset, R., Scott, M.P., Gubb, D., and Axelrod,
Garoia, F., Guerra, D., Pezzoli, M.C., Lopez-Varea, A., Cavicchi, S., and Gar- J.D. (2002b). Prickle mediates feedback amplification to generate asymmetric
cia-Bellido, A. (2000). Cell behaviour of Drosophila fat cadherin mutations in planar cell polarity signaling. Cell 109, 371–381.
wing development. Mech. Dev. 94, 95–109. Uemura, T., and Shimada, Y. (2003). Breaking cellular symmetry along planar
Haase, S.B., and Lew, D.J. (2007). Microtubule organization: cell shape is axes in Drosophila and vertebrates. J. Biochem. 134, 625–630.
destiny. Curr. Biol. 17, R249–R251. Villano, J.L., and Katz, F.N. (1995). four-jointed is required for intermediate
Ishikawa, H.O., Takeuchi, H., Haltiwanger, R.S., and Irvine, K.D. (2008). Four- growth in the proximal-distal axis in Drosophila. Development 121, 2767–
jointed is a Golgi kinase that phosphorylates a subset of cadherin domains. 2777.
Science 321, 401–404. Vincent, L., and Soille, P. (1991). Watersheds in digital spaces: an efficient
algorithm based on immersion simulations. IEEE 13, 583–598.
Joanny, J.F., Julicher, F., Kruse, K., and Prost, J. (2007). Hydrodynamic theory
for multi-component active polar gels. N. J. Phys. 9, 422. Vinson, C., and Adler, P.N. (1987). Directional non-cell autonomy and the
transmission of polarity information by the frizzled gene of Drosophila. Nature
Keller, R., Shih, J., and Sater, A. (1992). The cellular basis of the convergence
329, 549–551.
and extension of the Xenopus neural plate. Dev. Dyn. 193, 199–217.
Vladar, E.K., Antic, D., and Axelrod, J.D. (2009). Planar cell polarity signaling:
Keller, R., Shook, D., and Skoglund, P. (2008). The forces that shape embryos: The developing cell’s compass. Cold Spring Harb. Perspect. Biol. 1, a002964.
physical aspects of convergent extension by cell intercalation. Phys. Biol. 5,
Wang, J., Mark, S., Zhang, X., Qian, D., Yoo, S.J., Radde-Gallwitz, K., Zhang,
15007.
Y., Lin, X., Collazo, A., Wynshaw-Boris, A., et al. (2005). Regulation of polarized
Lawrence, P.A., Struhl, G., and Casal, J. (2007). Planar cell polarity: one or two extension and planar cell polarity in the cochlea by the vertebrate PCP
pathways? Nat. Rev. Genet. 8, 555–563. pathway. Nat. Genet. 37, 980–985.
Le Garrec, J.F., Lopez, P., and Kerszberg, M. (2006). Establishment and main- Wu, J., and Mlodzik, M. (2008). The frizzled extracellular domain is a ligand for
tenance of planar epithelial cell polarity by asymmetric cadherin bridges: Van Gogh/Stbm during nonautonomous planar cell polarity signaling. Dev. Cell
a computer model. Dev. Dyn. 235, 235–246. 15, 462–469.
Lopez-Schier, H., Starr, C.J., Kappler, J.A., Kollmar, R., and Hudspeth, A.J. Wu, J., and Mlodzik, M. (2009). A quest for the mechanism regulating global
(2004). Directional cell migration establishes the axes of planar polarity in the planar cell polarity of tissues. Trends Cell Biol. 19, 295–305.
posterior lateral-line organ of the zebrafish. Dev. Cell 7, 401–412. Zallen, J.A. (2007). Planar polarity and tissue morphogenesis. Cell 129, 1051–
Ma, D., Amonlirdviman, K., Raffard, R.L., Abate, A., Tomlin, C.J., and Axelrod, 1063.
J.D. (2008). Cell packing influences planar cell polarity signaling. Proc. Natl. Zallen, J.A., and Wieschaus, E. (2004). Patterned gene expression directs
Acad. Sci. USA 105, 18800–18805. bipolar planar polarity in Drosophila. Dev. Cell 6, 343–355.
Ma, D., Yang, C.H., McNeill, H., Simon, M.A., and Axelrod, J.D. (2003). Fidelity Zeidler, M.P., Perrimon, N., and Strutt, D.I. (2000). Multiple roles for four-
in planar cell polarity signalling. Nature 421, 543–547. jointed in planar polarity and limb patterning. Dev. Biol. 228, 181–196.

786 Cell 142, 773–786, September 3, 2010 ª2010 Elsevier Inc.


Generation of Rat Pancreas in Mouse
by Interspecific Blastocyst Injection
of Pluripotent Stem Cells
Toshihiro Kobayashi,1,2 Tomoyuki Yamaguchi,1,2 Sanae Hamanaka,1,2 Megumi Kato-Itoh,2,3 Yuji Yamazaki,1,2
Makoto Ibata,2 Hideyuki Sato,1,2 Youn-Su Lee,1,2 Jo-ichi Usui,1,6 A.S. Knisely,5 Masumi Hirabayashi,3,4
and Hiromitsu Nakauchi1,2,*
1Division of Stem Cell Therapy, Center for Stem Cell Biology and Regenerative Medicine, Institute of Medical Science, University of Tokyo,

4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan


2Japan Science Technology Agency, ERATO, Nakauchi Stem Cell and Organ Regeneration Project, 4-6-1 Shirokanedai, Minato-ku,

Tokyo 108-8639, Japan


3Center for Genetic Analysis of Behavior, National Institute for Physiological Sciences, Okazaki, Aichi 444-8585, Japan
4School of Life Science, The Graduate University for Advanced Studies, Okazaki, Aichi 444-8585, Japan
5Institute of Liver Studies, King’s College Hospital, London SE5 9RS, UK
6Present address: Department of Nephrology, Graduate School of Comprehensive Human Sciences, University of Tsukuba, 1-1-1 Tennoudai,

Tsukuba, Ibaraki 305-8575, Japan


*Correspondence: nakauchi@ims.u-tokyo.ac.jp
DOI 10.1016/j.cell.2010.07.039

SUMMARY cells and to transplant those organs into the patient. With the
development of induced pluripotent stem cell (iPSC) technology,
The complexity of organogenesis hinders in vitro we are now able to obtain patient-derived PSCs (Takahashi
generation of organs derived from a patient’s pluripo- et al., 2007; Takahashi and Yamanaka, 2006), although actual
tent stem cells (PSCs), an ultimate goal of regenera- developmental potentials remain to be defined, as do risks asso-
tive medicine. Mouse wild-type PSCs injected into ciated with somatic cell reprogramming. The real challenge is to
Pdx1 / (pancreatogenesis-disabled) mouse blasto- create a reproductive system for generation of PSC-derived
organs. The interactions among cells and tissues during devel-
cysts developmentally compensated vacancy of
opment and organogenesis are so complex that the recapitula-
the pancreatic ‘‘developmental niche,’’ generating
tion of these interactions to generate organs in vitro is essentially
almost entirely PSC-derived pancreas. To examine impractical. We have challenged this goal using the biology of
the potential for xenogenic approaches in blastocyst blastocyst complementation.
complementation, we injected mouse or rat PSCs into Blastocyst complementation was first reported by Chen et al.
rat or mouse blastocysts, respectively, generating They demonstrated that deficiency of T and B lymphocyte
interspecific chimeras and thus confirming that lineages in Rag2-deficient (Rag2 / ) mice was complemented
PSCs can contribute to xenogenic development by injecting normal mouse embryonic stem cells (mESCs) into
between mouse and rat. The development of these Rag2 / mouse-derived blastocysts (Chen et al., 1993).
mouse/rat chimeras was primarily influenced by Because Rag2 is an indispensable enzyme for rearrangement
host blastocyst and/or foster mother, evident by of immunoglobulin and T cell receptor genes, the T and B cells
generated in the complemented animals were mESC derived;
body size and species-specific organogenesis. We
there were no host T or B lymphocytes. We assumed that this
further injected rat wild-type PSCs into Pdx1 / complementation was possible because the Rag2 / host, inca-
mouse blastocysts, generating normally functioning pable of generating mature T and B cells, provided a ‘‘develop-
rat pancreas in Pdx1 / mice. These data constitute mental niche’’ for ESC-derived T and B cells.
proof of principle for interspecific blastocyst comple- We hypothesized that with blastocysts derived from a mutant
mentation and for generation in vivo of organs derived mouse strain in which the gene necessary to form a particular
from donor PSCs using a xenogenic environment. organ is deficient, the same principle might apply. To test
this hypothesis, we used, in this study, blastocyst complementa-
tion to generate functional pancreas from donor PSCs. The
INTRODUCTION pancreas, consisting of endocrine and exocrine glands, is formed
by early embryonic interactions of mesenchyme and epithelium
Current stem cell therapy mainly targets diseases that can be (Slack, 1995). Pdx1 (pancreatic and duodenal homeobox1) is
treated by cell replacement, such as Parkinson’s disease or dia- a Hox-type transcription factor that plays a critical role in pancre-
betes mellitus. One of the ultimate goals of regenerative medi- atic development and b cell maturation. Homozygous deficiency
cine, however, is to grow organs using the patient’s own stem of Pdx1 in the mouse results in death soon after birth due to

Cell 142, 787–799, September 3, 2010 ª2010 Elsevier Inc. 787


pancreatic insufficiency (Offield et al., 1996). Targeted disruption atic epithelium and mesenchyme, a key step in pancreatic devel-
of Pdx1 thus should empty a pancreatic ‘‘developmental niche’’ opment. Homozygote Pdx1-LacZ knockin mice (Pdx1 / ) are
in embryos derived from Pdx1 / blastocysts. Therefore, injec- born alive but die within 1 week after birth, presumably due to
tion of mouse PSCs (mPSCs) into Pdx1 / blastocysts should pancreatic insufficiency (Offield et al., 1996).
result in generation of pancreata almost entirely derived from For complementation donor cells, we used GT3.2 mouse
injected mPSCs. iPSCs (miPSCs) that had been generated from tail tip fibroblasts
Although our results verified our initial hypothesis, this system (TTFs) of an adult EGFP-transgenic (TG) C57BL/6 mouse (Okabe
cannot be applied to generate human organs. A xenogenic, but et al., 1997) using three factors (Oct3/4, Sox2, Klf4) in retroviral
not allogenic, blastocyst complementation system must there- vectors (Figure S1 available online). These GT3.2 miPSCs
fore be established. However, little is known about the nature were injected into blastocysts obtained by an intercross of
of the xenogenic barrier, how organogenesis by donor PSCs is heterozygote Pdx1-LacZ knockin mice (Pdx1+/ ), offspring of
influenced, or how intrinsic developmental programs can be C57BL/6(Pdx1+/ ) 3 DBA(Pdx1+/+) or C57BL/6(Pdx1+/ ) 3
regulated in xenogenic environments. To address these issues, BDF1(Pdx+/+) mice. G4.2 mESCs were also injected for compar-
we attempted to generate interspecific chimeras between ison. Neonates were assessed macroscopically and histologi-
mouse and rat using a blastocyst injection technique with mouse cally for pancreatic development and were genotyped for
and rat PSCs. Pdx1. These neonates were highly chimeric: Due to silencing
Generation of interspecific chimeras in livestock animals using of EGFP and to contamination by donor cells, accurate determi-
preimplantation embryos of each species is described; the nation of the genotypes required analysis at the single-cell level.
‘‘geep,’’ or chimera between goat and sheep, is famous as an To identify the genotypes of these neonates accurately, EGFP-
interspecific live chimera (Fehilly et al., 1984). In rodents, negative, c-Kit-positive, and Sca1-positive, lineage marker-
however, methods like those producing the geep succeeded negative (EGFP KSL) bone marrow cells were clone-sorted by
only between mouse subspecies, such as Mus musculus and fluorescence-activated cell sorting (FACS) to obtain single-cell-
Mus caroli, that are closely related but not capable of inter- derived hematopoietic colonies (Figure S2A). After culture for
breeding (Rossant and Frels, 1980). Many groups have sought 2 weeks, cells collected from each colony were subjected to
to generate interspecific chimeras between mouse and rat, PCR analysis (Figure S2B). This ‘‘colony PCR’’ method unambig-
successful with chimeric preimplantation embryos in vitro but uously determined the host embryo genotype and proved that
failing with live chimeric animals (Stern, 1973; Zeilmaker, pancreas had been formed in Pdx1 / mice by blastocyst
1973). Extraembryonic lineage cells like trophectoderm or prim- complementation (Figure S2C).
itive endoderm, derived from xenogenic embryos, might suffer In all postnatal chimeric mice derived from blastocysts
inhibition of implantation on exposure to host uterus; inhibition injected with either miPSCs or mESCs, pancreas was present
of further intrauterine development also is possible (Rossant regardless of host genotype (including Pdx1 / ). As expected,
et al., 1982; Tarkowski, 1962). Only cells of pre-blastocyst origin the pancreas in Pdx1+/ or Pdx1+/+ chimeric mice was a
can contribute to extraembryonic lineage cells, and mESCs/ composite of host-derived cells and EGFP-miPSC- or mESC-
iPSCs do not thus contribute (Rossant, 2007). Therefore, we derived cells, as in whole-body chimerism (Pdx1+/ + miPSCs
rechallenged this old issue with new technology, using iPSCs or mESCs in Figure 1A). In contrast, the pancreatic epithelium
or ESCs that are not capable of contributing to extraembryonic in Pdx1 / chimeric mice was almost entirely composed of
tissues. Using recently established culture conditions with a EGFP-marked miPSC- or mESC-derived cells (Pdx1 / +
combination of signaling inhibitors (Buehr et al., 2008; Li et al., miPSCs or mESCs in Figure 1A). The pancreas of these mice
2008; Ying et al., 2008), we generated rat-ESCs and -iPSCs was grossly and histologically normal. We examined these
(rESCs, riPSCs). Our work has now confirmed the existence of pancreatic tissues further for contributions of donor mPSCs to
interspecific chimeras generated with PSCs, using injection not different pancreatic lineages. Both miPSCs and mESCs supplied
only of mouse PSCs into rat embryos but also of rat PSCs into all pancreatic cell lineages (exocrine and endocrine tissues,
mouse embryos. including ductal epithelia), but pancreatic stromal elements—
Finally, by combining the principle of blastocyst complemen- vessels, nerves, and fibrocytes—were composites of host- and
tation with the production of interspecific chimeras, we suc- mPSC-derived cells in all mice (Figure 1B).
ceeded in generating rat pancreas in Pdx1 / mice. These sets
of experiments provide proof-of-principle data for donor iPSC- miPSCs Rescued Pdx1 / Mice by Blastocyst
derived organ generation in a xenogenic environment. Complementation
We next addressed whether mPSCs can rescue Pdx1 /
RESULTS lethality via blastocyst complementation. As we predicted based
on neonatal analysis (Figure 1), both mESCs and miPSCs
Generation of Donor Mouse iPSC-Derived Pancreas contributed to pancreatic organogenesis; Pdx1 / chimeric
in Pdx1-Deficient Neonates mice survived to adulthood (mESCs injection: n = 4, miPSCs
Our first goal was to generate pancreas from mPSCs. We per- injection: n = 15 in Figure 2A). They even served as Pdx1 /
formed a blastocyst complementation experiment using founders, transmitting their genotype to the next generation.
Pdx1 / blastocysts that would provide a niche for pancreatic Mating Pdx1 / founder male mice with Pdx1+/ female mice
organogenesis. These mice exhibit pancreatic agenesis due to increased to 50% the proportion of pups of homozygously dis-
the absence of Pdx1-specified interactions between prepancre- rupted genotype (Figure 2A). These data indicate that blastocyst

788 Cell 142, 787–799, September 3, 2010 ª2010 Elsevier Inc.


A Figure 1. Mouse PSC-Derived Pancreas Gener-
+ miPSCs + mESCs
ated by Way of Blastocyst Complementation
Pdx1-/- Pdx1+/- Pdx1-/- Pdx1+/- Pdx1-/- (A) mPSC-derived pancreas at neonatal stage. In
Pdx1 / mice complemented with mPSCs, the pan-
creas was almost entirely positive for EGFP, indicating
Phase

mPSC origin (left panels in +miPSCs and +mESCs). On


the other hand, in Pdx1+/ mice complemented with
mPSCs, the pancreas was partially positive for EGFP,
indicating a composite of both mPSC- and host-derived
cells (right panels in +miPSCs and +mESCs). Right panel
shows absence of pancreas in a nonchimeric Pdx1 /
EGFP

mouse at the same developmental stage. See also


Figure S1 for detailed characteristics of miPSCs and
Figure S2 for genotyping of Pdx1 status.
(B) Histological analysis of the distribution of mESC-
derived cells in neonatal mouse pancreas. Sections of
pancreas generated in a Pdx1 / mouse complemented
with mESCs were immunostained for EGFP; for pancre-
B
Pancreatic tissues Blood vessel atic endocrine markers (insulin, glucagon, and somato-
Endocrine Exocrine Ductal cell Endothelial cell statin); for the pancreatic exocrine marker a-amylase;
Insulin Glucagon Somatostatin α-Amylase DBA-Lectin PECAM1 for the ductal marker rhodamine-conjugated DBA-
lectin; and for blood vessels with PECAM-1, with DAPI
nuclear counterstaining.
Sections were observed under confocal laser scanning
microscopy. Scale bars in (B), 20 mm.

were composed of both host-derived and


miPSC-derived cells (Pdx1+/ in Figure 2D),
indicating, as previously reported (Deltour
et al., 1991), nonclonal origin of pancreatic
islets.
To test whether miPSC-derived pancreata
are functional in Pdx1 / chimeric mice, we
performed glucose tolerance testing (GTT).
Whereas streptozotocin (STZ)-induced dia-
betic mice failed to respond to GTT, Pdx1 /
complementation can be used to generate a functional organ chimeric mice responded well, indicating that Pdx1 / mice
and to yield homozygous founder mice even when uncomple- with miPSC-derived pancreas secreted insulin in response to
mented homozygosity is fatal. The method described here thus glucose loading and maintained normal serum glucose levels
can be useful for efficient production of gene-targeted mice (Figure 2E). Histology and function of exogenously derived
that are embryonic lethal without introducing a conditional pancreas were essentially the same for mice derived from
gene targeting system. miPSC- and from mESC-complemented blastocysts (Figure S3).
As expected, in both Pdx1 / and Pdx1+/ mice injected with These data demonstrate that the vacant ‘‘pancreatic niche’’
EGFP-miPSC, miPSC-derived cells contributed to all the non- provided in Pdx1 / mice can be occupied, with developmental
pancreatic tissues of the body, including lung, heart, liver, compensation, by miPSC- or mESC-derived cells after intraspe-
muscle, testis, and brain (data not shown). The extent of contri- cific blastocyst complementation, generating functionally intact
bution varied from tissue to tissue and also varied depending on pancreas.
individual chimera. However, the pancreas in adult Pdx1 / mice
was entirely derived from donor miPSCs (Pdx1 / in Figure 2B). Transplantation of miPSC-Derived Islets Corrected
Detailed analysis of EGFP distributions in adult miPSC-derived Hyperglycemia in Diabetic Mice
pancreas demonstrated that pancreatic islets, exocrine tissues, To assess the functionality of miPSC-derived pancreas, islets
and duct epithelia were entirely derived from donor miPSCs, as from miPSC-derived pancreas were transplanted into mice of
already shown in neonates (Pdx1 / in Figures 2C and 2D). In the original strain (C57BL/6) in which STZ administration had
expected contrast, pancreas from Pdx1+/ mice was a com- induced diabetes. As a control, islets from pancreas in Pdx1+/
posite of host and donor derivatives (Pdx1+/ in Figures 2B chimeric mice injected with miPSCs were transplanted. Blasto-
and 2C). Quantitative analysis of these sections by image J soft- cysts were obtained by an intercross of Pdx1+/ mice (C57BL/
ware revealed percentages of EGFP-positive cells to be 27.4% ± 6 3 DBA2 or C57BL/6 3 BDF1 F1 strains) that were semi-
27.8% in the miPSC + Pdx1+/ setting, 95.6% ± 4.6% in the allogenic to miPSCs used in this study. EGFP-expressing
miPSC + Pdx1 / setting. Of note is that most individual islets miPSC-derived islets (Figure 3A) were isolated conventionally

Cell 142, 787–799, September 3, 2010 ª2010 Elsevier Inc. 789


A Figure 2. Analysis of miPSC-Derived
Injected
Parents
No. of No. of Pdx1 genotype Pancreas in Adult Mice: Derivation of
cell adult mice chimeras +/+ +/- -/- Pancreatic Functional Cells and Their
G4.2 Pdx1+/- 27 18
5 9 4 Response to Glucose Load
+/-
(mESC) x Pdx1 ( 1 : 2 : 1 )
(A) Results of embryo manipulation and genotypes
GT3.2
-/-
(Pdx1 + mESCs) 0 13 15 of adult mice. Note that Pdx1 / mice comple-
29 28
(miPSC) x Pdx1+/- ( 0 : 1 : 1 ) mented with mESCs served as founders and
generated knockout mice more efficiently.
(B) Macroscopic images at adult stage
B Pdx1-/- + miPSCs Pdx1+/- + miPSCs C Pdx1+/- + miPSCs (8–12 weeks) of miPSC-derived pancreas gener-
Pdx1-/- + miPSCs
ated in a Pdx1 / mouse (Pa in left panel) and of
Li
composite pancreas generated in a Pdx1+/
Phase

mouse (right). In left panel, Li = liver, Pa = pancreas,

HE
Pa St
Sp
St = stomach, Sp = spleen, and Ki = kidney.
Ki
(C) Immunohistological studies of sections ob-
tained from pancreas revealed clear differences
in the distributions of miPSC-derived cells.
DAPI / EGFP
EGFP

Sections were stained with HE or for EGFP with


DAPI nuclear counterstaining.
(D) Immunohistological analysis of a generated
islet, showing the distribution of miPSC-derived
cells. Sections were stained for EGFP, with anti-
insulin antibodies, and with DAPI nuclear counter-
D E (mg/dl)
staining. Yellow lines show borders of endocrine
DAPI / EGFP / Insulin DAPI EGFP Insulin
600 and exocrine tissues. See also Figure S3 for anal-
+ miPSCs
Pdx1-/-

ysis of mESC-derived pancreas in adult mice.


400
(E) Results of GTT in Pdx1 / (-) and Pdx1+/ (:)
STZ-diabetic model
Pdx1-/-+ miPSCs mice (n = 6 each) complemented with miPSCs.
200
Pdx1+/-+ miPSCs Mice with STZ-induced diabetes (d and A) served
+ miPSCs

as controls. Fasting after 20 hr, blood was sampled


Pdx1+/-

0
via tail vein at intraperitoneal glucose administra-
0 30 60 90 120 tion (1 g/kg; 0 min) and 15, 30, 60, and 120 min
Minutes after glucose administration thereafter.
Sections in (C) were observed under fluorescence
microscopy and in (D) were observed under
confocal laser scanning microscopy. Scale bars
in (C), 100 mm; in (D), 50 mm. Error bars in (E) indi-
cate ± SD.

based on a method developed previously (Gotoh et al., 1985) Blastocyst complementation thus permits demonstration of
and were transplanted beneath the renal capsule of recipient proof-of-principle both for pancreas generation from PSCs and
mice. Nonfasting blood glucose levels were then monitored. As for diabetic therapy using donor iPSC-derived syngenic islets.
these islets were of donor origin (i.e., C57BL/6 strain), an immu-
nosuppressive regimen was not used. Generation of Interspecific Chimeras between Mouse
To prevent nonspecific loss of islets due to inflammation, and Rat
anti-inflammatory cytokine monoclonal antibody (mAb) cocktails Our second goal was to generate interspecific chimeras
were given at transplantation and 2 and 4 days thereafter between mouse and rat. To achieve this goal, we generated
(arrows, Figure 3E), as described (Satoh et al., 2007). Two not only miPSCs but also riPSCs and rESCs using an established
months after transplant, EGFP-expressing miPSC-derived islets protocol (Hirabayashi et al., 2009; Ying et al., 2008). These
were still detectable at the graft site (Figures 3B and 3C). Produc- mouse and rat PSCs enabled us bidirectionally to generate inter-
tion of insulin by the transplanted islets was confirmed immuno- specific chimeras.
histologically (Figure 3D). The induced-diabetic recipients no We injected EGFP-marked GT3.2 miPSCs into rat blastocysts
longer exhibited hyperglycemia; they maintained normal blood (r-blastocysts) or EGFP-marked riPSCs (Figures S4A and S4B)
glucose levels and responded normally to GTT (Figures 3E into mouse blastocysts (m-blastocysts). Because post-implan-
and 3F). This is in contrast to the therapeutic effect conferred tation development reportedly is severely hampered after intra-
by islets, composites of blastocyst- and miPSC-derived cells, uterine transfer of xenogenic blastocysts (Rossant et al., 1982;
obtained from pancreas of Pdx1+/ chimeric mice (C57BL/6 3 Tarkowski, 1962), injected r-blastocysts or m-blastocysts were
DBA2 or C57BL/6 3 BDF1 F1 origin). This effect lasted only for transferred, respectively, into the uteri of pseudopregnant rats
a short time, presumably due to immune rejection by the host or mice. After intrauterine transfer of injected r-blastocysts or
C57BL/6 diabetic mice (Figure 3E). These data strongly indicate m-blastocysts with development to the fetal stage, we evaluated
that the miPSC-derived pancreas, with islets, formed in an allo- EGFP expression by fluorescence microscopy for each trans-
genic host is functional and that the ‘‘autologous’’ islets thus ferred embryo. EGFP-expressing cells were found in the body
formed can be used to treat diabetes, without rejection. of each injected conceptus, but never in placenta (Figure 4A).

790 Cell 142, 787–799, September 3, 2010 ª2010 Elsevier Inc.


A B C D
DAPI / EGFP / Insulin
Phase Phase HE

EGFP EGFP EGFP


DAPI

E F
(mg/dl) (mg/dl)
600 600
Blood glucose

400 Blood glucose 400

200 200

0 0
0 10 20 30 40 50 60 (days) 0 30 60 90 120 (min)
Days after islet transplantation Minutes after glucose administration

Pdx1-/- + miPSCs derived islet transplantation Syngenic islet transplantation (PC)


Pdx1+/- + miPSC derived islet transplantation Sham transplantation (NC)

Figure 3. Transplantation of ‘‘Autologous’’ Islets from iPSC-Derived Pancreas: A Therapeutic Model


(A) EGFP-positive islets isolated from miPSC-derived pancreas. Via the common bile duct and pancreatic duct, miPSC-derived pancreas was perfused by colla-
genase. Density gradient centrifugation isolated a fraction enriched in EGFP-expressing islets.
(B) Kidney capsule 2 months after islet transplantation. At the site of transplantation (arrow), miPSC-derived islets still expressed EGFP.
(C) Renal subcapsular transplantation site (B, arrow); sections were stained with HE or immunostained for EGFP with DAPI nuclear counterstaining.
(D) Transplanted miPSC-derived islets in (C), immunostained for EGFP and insulin with DAPI nuclear counterstaining.
(E) Transplantation of miPSC-derived islets into mice with STZ-induced diabetes. Each mouse received 150 islets. Arrows indicate time points at which an mAb
cocktail (anti-INF-g, anti-TNF-a, anti-IL-1b) was administered. Nonfasting blood glucose levels were measured weekly for 2 months after transplantation.
Glucose levels are shown for STZ-induced diabetic mice transplanted with miPSC-derived islets (green; n = 3), with islets derived from Pdx1+/ chimeric
mice (orange; n = 2), with islets derived from syngenic strain of host strain (C57BL/6) as positive control (blue; n = 2), and with sham transplantation control
mice as negative control (purple; n = 3).
(F) Results of GTT 2 months after islet transplantation. Donors were Pdx1 / chimeric mice and Pdx1+/ chimeric mice, with blood sampling at the same time
points as in Figure 2E.
Sections in (C) were observed under fluorescence microscopy and in (D) were observed under confocal laser scanning microscopy. Scale bar in (D), 50 mm. Error
bars in (E) and (F) indicate ± SD.

This finding indicates that injected mouse or rat iPSCs can 26.5%, respectively, were detected in mouse and rat inter-
contribute to xenogenic development, with generation of inter- specific chimeras (representative FACS data shown in Fig-
specific chimeras. ure 4B). We also examined chimerism in hematopoietic cells
Next, we tried to quantitate contribution of mouse or rat iPSCs by staining cells from livers of interspecific chimera fetuses
to these interspecific chimeras. Chimerism in interspecific with antibodies specific for mouse and rat CD45 antigens.
embryos appeared to vary individual-to-individual and organ- Cells that expressed mouse or rat CD45 represented distinct
to-organ. Since quantitation of PSC-derived cells was difficult populations in interspecific chimeras, with only cells derived
in organs, we analyzed embryonic fibroblasts and hematopoietic from injected iPSCs expressing EGFP (Figure 4B). Whereas
cells. FACS analysis of embryonic fibroblasts revealed that a high proportion (28.3%) of mouse blood cells was detected
donor-derived EGFP+ cell percentages of about 28.0% and in r-blastocyst-derived chimeric fetal liver, rat blood cells were

Cell 142, 787–799, September 3, 2010 ª2010 Elsevier Inc. 791


A B Figure 4. Generation of Interspecific
Embryonic fIbroblast Fetal liver
rBL + miPSCs mBL + riPSCs Chimeras between Mouse and Rat
104
(A) Interspecific chimera fetuses generated by
19.3 1.7

rBL + miPSCs

mCD45-APC
103
Phase

injection of miPSCs into r-blastocyst (rBL: left


30.0 49.0
28.0 102
98.8
panels) and by injection of riPSCs into m-blasto-
101
cyst (mBL: right panels). With rat embryo manipu-
100
100 101 102 103 10
4
lation, fetuses were analyzed 12 days after embryo
100 101 102 103 104 100 101 102 103 104

EGFP rCD45-PE EGFP


transfer into uteri of 3.5 dpc pseudopregnant rats
4
10
EGFP

6.68 0.02 (embryonic day (E) 15.5). With mouse embryo

mBL + riPSCs

mCD45-APC
103
93.1 0.23
26.5 87.0 manipulation, fetuses were analyzed 11 days after
102

embryo transfer into uteri of 2.5 dpc pseudopreg-


101

nant mice (E13.5). See also Figure S4 for charac-


100
4
100 101 102 103 10
teristics of riPSCs and interspecific embryos after
100 101 102 103 104 100 101 102 103 104

EGFP rCD45-PE EGFP


injection of riPSCs into mouse 8-cell/morula stage
embryos.
C D (B) Representative data of FACS analysis on cells
rBL mBL
+ miPSCs +riPSCs Control
from fetal liver and on embryonic fibroblasts
Exon 2 Exon 3 Exon 4
5+

5+
derived from interspecific chimeras. Right panels

in D45 +
5+

5+

te
in 45 +

tem No
r

pla
B
D4

D4
rke

B
D4

D4

mP

rP
D
show cells from fetal liver immunostained with

mC
mC

mC
Ma

rC
rC

rC
rOct3/4 locus = 966bp
mOct3/4 locus = 861bp
rOct3/4 locus
mOct3/4 locus
anti-mCD45 and -rCD45 antibodies. Note that
anti-mouse or -rat monoclonal antibodies (mAb)
against CD45 can distinguish CD45-expressing
E F G hematopoietic cells in a species-specific manner.
No. of
Almost all CD45-expressing cells derived from
(total transferred embryos = 100%)

n = 16 n = 19 24 26 109 30 transferred n=6 n = 12 n = 23 n = 19


(%) (%) (%)
embryoa
100 100 100 injected cells express EGFP, indicating iPSC
Relative frequency

75 75 75 origin.
(C) Schema of mouse and rat Oct3/4 loci (mOct3/4
50 50 50
and rOct3/4). Of note is that in the rat Oct3/4 locus
Aborted
25 25 25
Non-chimera
Chimera
the 2 introns flanking exon 3 are longer than in the
0 0 0 mouse Oct3/4 locus. This difference in length can
+ +
EF CD45 EF CD45 Embryo Rat Mouse Mouse Rat Embryo Rat Mouse Mouse Rat
distinguish origins of otherwise similar cells.
in FL in FL
Cell miPSC riPSC Cell miPSC riPSC
mBL+riPSCs rBL+riPSCs Arrowheads indicate common primers of each
species for PCR. PCR product sizes for both
species are shown below.
(D) Results of genotyping to identify origin of hematopoietic cells expressing mouse or rat CD45 in fetal liver of interspecific chimeras. Peripheral-blood CD45-
expressing cells from each species served as positive controls.
(E) Correlation of chimerism between embryonic fibroblasts and CD45+ hematopoietic cells in fetal liver. Cells were prepared from interspecific chimeras gener-
ated by injection of riPSCs into m-blastocysts or from intraspecific chimeras generated by injection of riPSCs into r-blastocysts.
(F) Relative frequencies of aborted, nonchimeras, and chimeras embryonic development (E13.5 and 15.5).
(G) Chimerism analysis of embryonic fibroblasts from chimeras generated by injection of miPSCs or riPSCs into mouse or rat blastocysts. Fibroblasts were
obtained from chimeras and analyzed for EGFP intensity by FACS. Plotted dots show degrees of chimerism for individual embryos. See also Figure S5 for inter-
specific chimeras using ESCs.
Scale bars in (A), 5 mm.

only rarely present (less than 3.3%) in m-blastocyst-derived chimeras (Figures 4F and 4G). In addition, high contributions
chimeric fetal liver (Figure 4B). This tendency was specific to by xenogenic cells appeared to be associated with morpholog-
interspecific chimeras and not observed in intraspecific ical abnormalities and embryonic lethality (data not shown). To
chimeras (Figure 4E). It is not clear why the difference in contri- exclude the possibility that these abnormalities were caused by
bution of iPSCs to hematopoietic cells between mouse and rat donor iPSCs, we also attempted to generate interspecific
interspecific chimeras was so marked. chimeras using mouse or rat ESCs. DsRed-marked EB3DR
To further confirm interspecific chimerism, genomic DNA mESCs could also generate interspecific rat chimeras (top
extracted from FACS-sorted cells expressing CD45 was PCR- panels in Figure S5A), with embryonic development rate and
amplified using primers common to the mouse and rat Oct3/4 degree of chimerism similar to those generated by miPSC
loci (Figure 4C). PCR products of different lengths, indicating injection (Figure S5B). The Venus-marked WIv3i-1 and -5
origin in each species, were clearly present (Figure 4D). These rESC lines, with high contribution to rat embryo development
results strongly indicated that the animals harboring these cells and germline competency (Hirabayashi et al., 2009), could
were mouse/rat interspecific chimeras. also generate interspecific chimeras after injection into m-blas-
To investigate the influence of iPSC contribution to xenogenic tocyst (middle and bottom panels in Figure S5A), but embry-
development at the fetal stage, we assessed embryonic devel- onic development rate and degree of chimerism were lower
opment rate of interspecific chimeras, and the extent of chime- than reported for intraspecific chimeras (Figure S5C). These
rism, by FACS analysis using established embryonic fibroblasts. results suggest that generation of interspecific chimeras
Both embryonic development rate and degree of chimerism between mouse and rat is less efficient than generation of intra-
were lower in interspecific chimeras than in intraspecific specific chimeras.

792 Cell 142, 787–799, September 3, 2010 ª2010 Elsevier Inc.


A B Figure 5. Postnatal Analysis of Interspecific
rBL + miPSCs mBL + riPSCs
(g) Chimeras
7.5 n=4 n=4 n=8 n=7 (A) Photograph of newborn interspecific chimeras
Phase
derived from r-blastocyst injected with miPSCs
Phase

and m-blastocyst injected with riPSCs.

Body weight
5.0 (B) Neonatal body weights of interspecific
Non-chimera chimeras were measured and plotted. One
Non-chimera
EGFP chimera obtained after injection of miPSCs into
2.5
r-blastocyst showed a high contribution of mouse
cells as shown in the insert, with body weight and
EGFP

0 size equivalent to those of newborn mouse (right


rBL+ mBL+ Non-chimera
miPSCs Rat riPSCs Mouse
panel).
(C) Photograph of interspecific chimera at 8 weeks
of age.
(D) Analysis of chimerism in neonates by organ or
rBL + miPSCs mBL + riPSCs tissue. Interspecific chimeras generated by miPSC
C
injection into r-blastocyst (left panels) and by
riPSC injection into m-blastocyst (right panels)
are shown. Sections of representative organs
(heart, liver, pancreas, and kidney) after immunos-
taining for EGFP antibody with DAPI nuclear coun-
terstaining are shown.
(E) Neonatal pancreas and adult liver of inter-
specific chimera generated from r-blastocyst
injected with miPSC, immunostained for EGFP
and insulin or for EGFP and albumin or CK19
with DAPI nuclear counterstaining. In pancreas,
an islet-like cell cluster contains cells producing
D rBL + miPSCs mBL + riPSCs insulin composed of both miPSC-derived cells
that express EGFP and r-blastocyst-derived cells
Heart Pancreas Heart Pancreas that do not. In liver, albumin-positive hepatocytes
and CK19-positive cholangiocytes (arrowhead)
also express EGFP, indicating miPSC origin.
DAPI / EGFP

(F) Distribution of xenogenic cells in neonatal testis


and in developing gonad. MVH-positive germ cells
Liver Kidney Liver Kidney did not express EGFP, indicating nonxenogenic
cell origin.
Sections in (D) were observed under fluorescence
microscopy and in (E) and (F) were observed under
confocal laser scanning microscopy. See also
Figure S6 for macroscopic images of neonates
E F rBL + miPSCs mBL + rESCs and results of peripheral blood analysis of adults.
rBL + miPSCs Scale bars in (A) and (B), 10 mm; in (D), 100 mm;
Pancreas (P1) Liver (Adult) Testis (P1) Gonad (♀ E13.5) (E) and (F), 50 mm.
DAPI / EGFP / Insulin DAPI / EGFP / Albumin DAPI / EGFP / CK19 DAPI / EGFP / MVH DAPI / EGFP / MVH

Interspecific Chimeras Were Live-born; Some Grew black coat) (Figure 5C, r-blastocyst + miPSCs: n = 8, m-blasto-
into Adulthood cyst + riPSCs: n = 4). The full-term development rate of inter-
To investigate the developmental potential of generated specific chimeras, either with miPSCs into r-blastocyst or with
chimeras and to assess the functionality of the cells, tissues, or riPSCs into m-blastocyst, was 20%. In both settings it was
organs derived from injected cells, we analyzed interspecific lower than that for intraspecific chimeras (50%).
chimeras at neonatal and adult stages. Mouse- or rat-iPSC-
injected interspecific chimeras survived after birth and expressed Determination of Body Size in Interspecific Chimeras
EGFP ubiquitously (as did intraspecific chimeras; Figure 5A, Adult rats typically are ten times bigger than adult mice, whereas
r-blastocyst + miPSCs: n = 5, m-blastocyst + riPSCs: n = 10). newborn rats are three times bigger than newborn mice.
As these chimeras developed into adulthood, chimerism could Because mouse and rat gestations are of similar length (19 and
be judged by coat color because miPSCs (C57BL/6, black 21 days, respectively), organogenesis requires more cell prolifer-
coat) were injected into r-blastocyst (Wistar, white coat) or riPSCs ation and differentiation during rat development than during
(Wistar) were injected into m-blastocyst (BDF11 3 C57BL/6, mouse development. What determines the size of interspecific

Cell 142, 787–799, September 3, 2010 ª2010 Elsevier Inc. 793


A Figure 6. Correlation between Body Weight
Chimera #1 Chimera #2 Chimera #3 Chimera #4
and Chimerism in Interspecific Chimeras
(A) Photographs of interspecific chimeras derived
from r-blastocyst injected with miPSCs, with FACS
analysis of their chimerisms in peripheral blood
mononuclear cells.
(B) Body weight change after weaning for inter-
specific chimeras (green) and their nonchimeric
105 105 105 105 littermates (orange).
mCD45-APC

104 104 104 104

103 103 103 103

5.35 0.29 15.6 2.06 17.8 2.62 55.5 1.3


102 102 102 102
0
0.24 94.1 0
0.05 82.3 0
1.28 78.3
positive cells (Figure S6B). In sections
0
0.27 42.9
0 10 10 10 10 2 3
0 10 4
10 5
10 10 2 3
0 10 104
10 5
10 0 10 10 102 10 3 4 immunostained with anti-EGFP antibody,
5 2 3 4 5

rCD45-PE representative images demonstrated that


EGFP-positive cells become various
types of tissues (Figure 5D). Of particular
B note was a pancreatic islet generated by
(g) (g)
injection of miPSCs into r-blastocyst.

600 600
Cells in this islet marked immuno-
Body weight

Body weight

histochemically for insulin; the islet was


400 Littermate #1 400 Littermate #3
Littermate #2 Littermate #4
a composite of EGFP-positive and -nega-
Chimera #2 Chimera #1 tive cells, indicating that the islet con-
Chimera #4 Chimera #3
200 200 sisted of host rat cells and exogenous
donor mouse cells (Figure 5E). Islet poly-
0 0 clonality also was seen with intraspecific
4 9 14 (wk) 4 9 14 (wk)
chimeras (Figure 2E). The expression of
Weeks after birth Weeks after birth
other functional molecules in xenogenic
cells (i.e., hepatocytes or cholangiocytes
chimeras is an intriguing biological question. Interestingly, body in the liver or leukocytes in the peripheral blood) was also
size and weight of interspecific chimeras born from rat foster detected (Figure 5E and Figure S6C).
mothers were similar to those of normal newborn rats, whereas It is known that rats do not have gallbladders, whereas mice
for those born from mouse foster mothers they were similar to do. To date, when mPSCs were injected into r-blastocysts, the
those of newborn mice (Figures 5A and 5C). However, high interspecific chimeras produced (generally the size of rats)
contributions by xenogenic cells may also affect interspecific have not had gallbladders (n = 8). In contrast, interspecific
chimera size. One chimera obtained after injection of miPSCs chimeras generated from m-blastocysts complemented with
into r-blastocysts showed body weight and size equivalent to rPSCs (more like mice in size) have had gallbladders (n = 4).
those of a newborn mouse, although its birth mother was a rat To see the xenogenic contribution to germ cells, testis and
(Figure 5B). This particular chimera’s donor miPSC-derived cell developing gonad of the interspecific chimeras were examined.
contribution was extremely high (Figure 5B, inlet). However, Cells were not found that coexpressed EGFP or Venus and the
chimeras with high contributions of xenogenic iPSC-derived germ cell marker mouse vasa homolog (MVH) (Figure 5F). As
cells generally were not identified, suggesting an association the injected miPSCs or rESCs were both confirmed as germ-
with embryonic lethality. In most chimeras, the size of newborns line-competent pluripotent stem cells in intraspecific settings
seemed to conform with that in the species from which the blas- (data not shown), germ cell development may be impaired or
tocyst originated. A correlation between body weight and contri- more limited in the xenogenic environment.
bution of donor iPSC-derived cells as estimated from hair color
and peripheral blood cells is shown in Figure 6. The origins of Generation of Rat Pancreas in Mouse via Interspecific
placenta and uterus may have a key role in size determination. Blastocyst Complementation
Injected xenogenic PSCs never developed into placenta, which Our last goal was to generate xenogenic rat pancreas in Pdx1 /
was always of host blastocyst origin (Figure 4A, Figure S5A). mice by interspecific blastocyst complementation. For efficient
Therefore it is difficult to determine whether it is placenta or production of Pdx1 / mice, embryos were generated by inter-
uterine environment that influences the size of embryos. cross of Pdx1 / founder male mice (in which pancreas arose
principally from exogenous miPSCs) with Pdx1+/ female mice.
Distribution of Donor iPSC-Derived Cells With this founder system, half the mice born were Pdx1 /
in the Xenogenic Environment (in contrast to intercross of Pdx1+/ heterozygotes in which
We determined the distribution of mouse- or rat-iPSC-derived only 25% of offspring were Pdx1 / ). For donor riPSCs, we
EGFP-positive cells in neonatal interspecific chimeras. With used the riPSC#3 line (Figure S4A). This line was selected among
miPSC injection into r-blastocyst (Figure S6A) and with riPSC 11 established riPSC clones for embryonic developmental rate
injection into m-blastocyst, almost all organs contained EGFP- and degree of chimerism after injection into mouse embryos

794 Cell 142, 787–799, September 3, 2010 ª2010 Elsevier Inc.


A Phase EGFP B Figure 7. Generation of Rat Pancreas in
EGFP α-Amylase Pdx1-Deficient Mouse by Interspecific Blas-
Pdx1-/- + riPSCs

DAPI DAPI
tocyst Complementation
(A) riPSC-derived pancreas in neonatal Pdx1 /
mouse. In Pdx1 / mice complemented with
riPSCs, almost all pancreata expressed EGFP,
Glucagon Somatostatin Insulin indicating riPSC origin (top panel). On the other
DAPI DAPI DAPI
hand, in Pdx1+/ mice complemented with riPSCs,
Pdx1+/- + riPSCs

pancreata only partially expressed EGFP, indi-


cating a composite of both riPSC- and mouse-
derived cells (bottom panel).
(B) Immunohistological studies of sections ob-
tained from riPSC-derived pancreas. Serial frozen
sections were immunostained for EGFP, a-
amylase, insulin, glucagon, and somatostatin
C D (mg/dl)
Pdx1-/- + riPSCs 600 with DAPI nuclear counterstaining. See also
Pdx1-/-+ riPSCs
Figure S7 for genotyping to identify Pdx1 status
400 Pdx1+/-+ riPSCs and origin of each species in interspecific chimera.
(C) Photograph of adult Pdx1 / mouse comple-
200
mented with riPSCs.
(D) Results of GTT in Pdx1 / (-) and Pdx1+/
(:, A) mice complemented with riPSCs.
0
0 30 60 90 120 (min) (E) riPSC-derived pancreas in adult Pdx1 /
Minutes after glucose administration mouse. Compare similar results of neonatal anal-
ysis (Figure 5D).
E Phase EGFP F (F) Histological studies of sections obtained from
HE EGFP riPSC-derived adult pancreas revealed clear
Li
Pdx1-/- + riPSCs

St DAPI
Du
differences in the distributions of riPSC-derived
Sp
cells after staining by anti-EGFP antibody with
Pa
Ki
DAPI nuclear counterstaining (right panel).
Sections were also stained with HE.
Sections in (F) were observed under light or fluo-
rescence microscopy and in (B) were observed
Pdx1+/- + riPSCs

under confocal laser scanning microscopy. Scale


bars in (B) and (F), 100 mm.

(data not shown). After injection, 139 embryos were transferred (an exocrine tissue marker) and insulin, glucagon, and somato-
into uteri of pseudopregnant mice and 34 mice were born. statin (endocrine tissue markers; Figure 7B).
They were analyzed at neonatal and adult stages. As in wild-type experiments, successful maturation into adult-
The contribution of EGFP-marked riPSC-derived cells in the hood (8 weeks) was uncommon in Pdx1 / mice complemented
pancreas of neonatal Pdx1+/ interspecific chimeras was small with riPSCs; however, adult mice with riPSC-derived pancreas
relative to that of host-derived cells, as seen in whole-body (Figure 7C; n = 2) had intact pancreas expressing EGFP (Figures
chimerism (Pdx1+/ + riPSCs in Figure 7A bottom panel; n = 5). 7E and 7F). Quantitative analysis of the sections by image J soft-
In contrast, the pancreatic epithelia in Pdx1 / interspecific ware revealed percentages of EGFP-positive cells to be 81.9% ±
chimeras were entirely composed of EGFP-marked riPSC- 3.4%. Additionally, on GTT in adulthood, insulin was secreted in
derived cells (Pdx1 / + riPSCs in Figure 7A top panel; n = 10). response to glucose loading and normal serum glucose levels
Each genotype was confirmed by PCR using genomic DNA were maintained (Figure 7D). These results indicate that genera-
extracted from FACS-sorted mouse CD45 (mCD45)-positive tion of a xenogenic iPSC-derived organ is possible via interspe-
splenocytes (Figure S7A). Neonates with entirely EGFP-positive cific blastocyst complementation.
pancreata thus were clearly identified as of Pdx1 / genotype
(Figure S7B). The existence of interspecific chimeras between DISCUSSION
mouse and rat was also confirmed by FACS patterning, which
demonstrated distinct populations of mCD45- and rat CD45 We report three innovative observations, using proof-of-prin-
(rCD45)-positive cells, with only rCD45-positive cells expressing ciple approaches. (1) If an empty developmental niche for an
EGFP after riPSC injection (Figure S7A). FACS-sorted mCD45- organ is provided (as with the Pdx1 / mouse and the pancreatic
and rCD45-positive cells were also confirmed as, respectively, niche), PSC-derived cellular progeny can occupy that niche
mouse or rat in origin by genomic PCR testing using Oct3/4 and developmentally compensate for the missing contents of
locus primers (Figure S7C), which clearly identified cell origin. the niche, forming an organ almost entirely composed of
On immunostaining, riPSC-derived pancreas expressed EGFP cells derived from donor PSCs. (2) Generation of interspecific
almost universally (Figure 7B) and also expressed a-amylase chimeras between mouse and rat is possible with injection of

Cell 142, 787–799, September 3, 2010 ª2010 Elsevier Inc. 795


mouse or rat PSCs into embryos from the other species; injected among extraembryonic lineage cells, with exposure of xeno-
PSC-derived cells are distributed throughout the body and genic cells to the uterine environment, is inhibitory to implanta-
appear to function normally. (3) The combination of (1) and (2) tion and/or to further intrauterine development of interspecific
successfully generates rat pancreas in mouse with injection of chimeras.
riPSCs into Pdx1 / mouse embryos, a technique that we term It is of prime interest that body size and weight of interspecific
‘‘interspecific blastocyst complementation.’’ chimeras conformed with those of the species of the foster
We demonstrated that the Pdx1 / mice derived from blasto- mother (Figures 5A–5C). What xenogenic components contribute
cysts complemented with mPSCs were born with functional to the phenotypic determination of interspecific chimeras?
pancreas almost entirely derived from donor PSCs and grew It seems that placenta and/or uterine environment are respon-
into adulthood without showing any evidence of pancreatic sible for size determination of adult interspecific chimeras
insufficiency. Several groups have used the same technique to both as embryos and as adults. Given that placenta must be of
study the development of thymic epithelium (Muller et al., the same origin as the foster mother for successful generation
2005), to compensate for cardiac defects (Fraidenraich et al., of interspecific chimeras, it is not clear which is primarily respon-
2004), or to determine if yolk sac hematopoiesis and germ cell sible for this determination. Degree of chimerism may also
development are of clonal or nonclonal origin (Ueno et al., influence the phenotype. As observed in intraspecific chimeras,
2009; Ueno and Weissman, 2006). Although Stanger et al. tested contribution and distribution of xenogenic cells in interspecific
development of pancreas and liver in embryos to define organ chimeras vary organ-to-organ. There is a negative correlation
size determinants (Stanger et al., 2007), no study has exploited between contribution of donor (mouse) PSC-derived cells and
this technique to produce donor-derived functional organs and body weight (Figure 6). Although we could obtain interspecific
rescued a lethal phenotype to adulthood. chimeras consistently (Figure 4 and Figure 5), embryonic lethality
Direct in vitro differentiation of insulin-producing cells from was high and postnatal development was poor. Some interac-
PSCs has been a major focus of stem cell therapy, as recently tions between mouse- and rat-derived cells indispensable
demonstrated by a sophisticated protocol to generate pancre- for organism survival may not work across species, resulting in
atic endoderm efficiently via stepwise endodermal differentia- death during embryonic development or in retarded postnatal
tion (Kroon et al., 2008). However, the in vitro generation development.
of insulin-producing cells still needs further improvement in Another example is the formation of gallbladders in inter-
differentiation efficiency, in insulin production levels, or in speed specific chimeras. Those derived from m-blastocysts have
of insulin response to glucose changes. In addition, the risk gallbladders but those from r-blastocysts do not. It is conceiv-
of tumor development due to contamination with undifferenti- able that the temporo-spatial development of donor PSC-
ated PSCs must be rigorously assessed before clinical use. derived cells is regulated by the xenogenic host microenviron-
Compared with those generated in vitro, insulin-producing cells ment, which governs morphogenesis and organogenesis.
obtained from pancreas that is formed in vivo by blastocyst However, the data also indicate that the intrinsic develop-
complementation must have gone through near-normal differ- mental program imprinted in PSCs may create competition
entiation processes with proper epigenetic changes. The between host blastocyst-derived and donor PSC-derived cells
tissues obtained, such as insulin-producing cells, thus are to form chimeric organs. The balance between host and donor
presumed to be fully functional and the risk of teratoma devel- cells at certain critical points during embryonic development
opment due to contamination of undifferentiated PSCs to be thus may be important. Xenogenic developmental systems
negligible. Nonetheless, the oncogenicity of iPSC-derived cells may be useful to elucidate these and other developmental
due to reactivation of introduced genes (Miura et al., 2009; questions and to help in generation of chimeras between
Nakagawa et al., 2008) or to genome abnormality due to long- species evolutionarily more distant from one another than
term culture remains to be assessed. To establish iPSCs without are mouse and rat.
genomic integration of a retroviral sequence should further Most importantly, we succeeded in generating functional rat
reduce the risk associated with the use of iPSCs (Okita et al., pancreas in Pdx1 / mice via interspecific blastocyst comple-
2008). mentation. In all interspecific neonates derived from Pdx1 /
We assumed that aggregation of early embryos would lead blastocysts injected with riPSCs, pancreas was present.
to the presence in trophectoderm of xenogenic cells that are Although full maturation into adulthood was not common, once
reported to be harmful to embryonic development after uterine the mice matured into adulthood, the generated riPSC-derived
implantation. We therefore injected mESCs/iPSCs, but not blas- pancreas was morphologically and histologically normal and
tomere cells, into r-blastocysts. As predicted, EGFP-positive was not associated with any sign of diabetes or other abnormal-
PSC-derived cells were not detected in placentas (Figure 4A ities; GTT results strongly indicated normal function. Generation
and Figure S5A), and we succeeded in generating interspecific of functional cells (sperm, hepatocytes) in xenogenic environ-
chimeras. To confirm this further, we then attempted to generate ments has been reported (Mercer et al., 2001; Shinohara et al.,
interspecific chimeras by injecting rESCs/iPSCs into m-blasto- 2006). In addition, hematopoietic xeno-chimeras have been
cysts. After injection into 8-cell/morula stage mouse embryos, used commonly as a method to study functionality of hematopoi-
the riPSCs were eventually enclosed within the inner cell mass etic stem cells (Kamel-Reid and Dick, 1988; McCune et al.,
of the m-blastocyst and were never detected in the m-blastocyst 1988). No study, however, has demonstrated generation in
trophectoderm (Figures S4C and S4D). Our study, consistent a xenogenic environment of a PSC-derived functional organ
with others, indicates that the presence of xenogenic cells that can rescue embryonic lethality to adulthood.

796 Cell 142, 787–799, September 3, 2010 ª2010 Elsevier Inc.


The organ generation system described may be applied to EXPERIMENTAL PROCEDURES
treat organ failure in humans if pigs or other large animals are
Animals
used. There are, however, several issues that need to be
C57BL/6NCrSlc, BDF1, DBA/2CrSlc, ICR mice, and Wistar rats were
addressed to bring this principle into the clinic. For example, purchased from SLC Japan (Shizuoka, Japan). Pdx1-LacZ heterozygous
though we were able to generate interspecific chimeras between mice (Offield et al., 1996), kindly provided by Dr. Y. Kawaguchi (Kyoto
mouse and rat, their embryonic lethality is high and maturation University) and Dr. C.V. Wright (Vanderbilt University), were crossed with
into adulthood is uncommon. The nature of this xenogenic barrier C57BL/6, DBA2, or BDF1 strain mice. C57BL/6 mice were given STZ (Sigma,
is not clear, but it is evident that the evolutionary distance St. Louis, MO, USA) to induce diabetes. Mice with nonfasting blood glucose
levels > 400 mg/dL 1 week after STZ administration (200 mg/kg) were regarded
accounts for this, as we do not see these problems in intraspecific
as hyperglycemic and thus as diabetic mice. All experiments were performed
chimeras. Livestock animals such as pigs or sheep may be too
in accordance with the animal care and use committee guidelines of the Insti-
distant evolutionarily for successful complementation. In addi- tute of Medical Science, University of Tokyo.
tion, as described in the allogenic system, vessels, nerves, and
some interstitial elements that are not under the influence of Culture of ESCs/iPSCs
Pdx1 expression were composites of host- and miPSC- or Undifferentiated mESCs were maintained on gelatin-coated dishes without
mESC-derived cells. Although we showed in this study that islets feeder cells in Glasgow’s modified Eagle’s medium (Sigma) supplemented
with 10% fetal bovine serum (FBS; Nichirei Bioscience, Tokyo, Japan),
prepared from miPSC-derived pancreas generated in allogenic
0.1 mM 2-mercaptoethanol (Invitrogen, San Diego, CA, USA), 0.1 mM nones-
hosts indeed were successfully transplanted into ‘‘autologous’’ sential amino acids (Invitrogen), 1 mM sodium pyruvate (Invitrogen), 1%
mice with STZ-induced diabetes without rejection, whether the L-glutamine penicillin streptomycin (Sigma), and 1000 U/ml of mouse leukemia
same principle applies to transplantation of islets obtained from inhibitory factor (LIF; Millipore, Bedford, MA, USA). The G4.2 mESCs and
pancreas generated in xenogenic animals remains to be seen. EB3DR mESCs, kindly provided by Dr. H. Niwa (Center for Developmental
Transplantation of islets from rat pancreas generated in Pdx1 / Biology, RIKEN), were derived from EB3 mESCs (Niwa et al., 2000) and carried
mice into diabetic rats should answer this question. However, the CAG promoter-driven EGFP or DsRed gene. Undifferentiated miPSCs
were maintained on mitomycin-c treated mouse embryonic fibroblasts
due to the size difference between mouse and rat, and to high
(MEFs) in Dulbecco’s modified Eagle’s medium (Invitrogen) supplemented
embryonic lethality and poor postnatal maturation of interspecific with 15% knockout serum replacement (Invitrogen), 0.1 mM 2-mercaptoetha-
chimeras, it is not possible to obtain sufficient numbers of islets to nol, 0.1 mM nonessential amino acids, 1 mM HEPES buffer solution (Invitro-
treat diabetic rats. Generation of mouse pancreas in Pdx1 / rats gen), 1% L-glutamine penicillin streptomycin, and 1000 U/ml of mouse LIF.
will be necessary to do such experiments. The GT3.2 miPSCs were generated from TTFs of a male EGFP Tg mouse
Production of organ-deficient livestock animals and genera- (kindly provided by Dr. M. Okabe, Osaka University) by introducing three
factors (Klf4, Sox2, Oct3/4) in retroviral vectors (Okabe et al., 2009). The
tion of chimeras is another issue, but nuclear transfer technology
GT3.2 miPSCs ubiquitously express EGFP under the control of the CAG
available for livestock animals may permit establishment of promoter.
organ-deficient pig lines, for example (Lai et al., 2002). The Undifferentiated rESCs/iPSCs were maintained on mitomycin-c treated
successful generation of pig chimeras using blastocyst injection MEFs in N2B27 medium (Ying et al., 2003) containing 1 mM MEK inhibitor
has also been reported (Nagashima et al., 2004). The major diffi- PD0325901 (Axon Groeningen, The Netherlands), 3 mM GSK3 inhibitor
culty seems to be that primate and rodent PSCs differ (Nichols CHIR99021 (Axon), with or without FGF receptor inhibitor SU5402 (Calbio-
chem, La Jolla, CA, USA), and 1000 U/ml of rat LIF (Millipore). The riPSCs
and Smith, 2009); limits of primate PSCs in contributing to
were generated from Wistar rat embryonic fibroblast by introducing three
embryo development have been suspected. Poor contribution mouse factors (Oct3/4, Klf4, Sox2) in lentiviral vectors as a doxycycline-induc-
of human ESCs to embryo development after injection into ible expression unit using N2B27 medium containing the three inhibitors
mouse blastocysts or chick embryo has been demonstrated described above. Expression of representative ESC marker genes has been
(Goldstein et al., 2002; James et al., 2006). Generation of inter- confirmed and teratoma formation by these riPSCs after injection into immu-
specific chimera technology may prove a tool useful in assess- nodeficient mice has also been confirmed (our unpublished data). The riPSCs
ment of pluripotent stem cell potential, thereby addressing this ubiquitously express EGFP under the control of the Ubiquitin-C promoter.
The WIv3i-1 or WIv3i-5 rESCs were generated from Venus Tg rat blastocyst
issue.
(Hirabayashi et al., 2009). These rESCs ubiquitously express Venus under
Another issue of concern is the fact that PSC-derived cells the control of the CAG promoter.
are found not only in pancreas but in all organs and tissues,
including brain and gonads. Therefore, without proper control Embryo Culture and Manipulation
of the differentiation potential of PSCs, generation of human Preparation of wild-type or Pdx1 heterozygous intercrossing embryos was
organs in livestock animals will face an ethical issue. There are carried out according to published protocols (Nagy et al., 2003). In brief,
mouse 8-cell/morula stage embryos were collected in Medium 2 (Millipore)
several approaches to address this. One is use of committed
from oviduct and uterus of mice 2.5 days postcoitum (dpc). These embryos
stem or progenitor cells in place of PSCs. If they are introduced were transferred into potassium simplex optimized medium with amino acids
into an appropriate microenvironment at an appropriate devel- (Millipore) and were cultured for 24 hr for blastocyst injection.
opmental time point, to restrict differentiation toward a particular Rat blastocysts were collected in a bicarbonate-buffered medium
organ may be possible. An alternative is to use genetically modi- composed of Roswell Park Memorial Institute medium (RPMI) 1640, Eagle’s
fied PSCs whose differentiation potential is restricted to certain solution, and Ham’s F12 containing 18% FBS medium (Ogawa et al., 1971)
tissues or organs. from oviduct and uterus of rats 4.5 dpc. These embryos were transferred
into modified rat 1-cell embryo culture medium (Oh et al., 1998) containing
In conclusion, the approach described here will be of use not
80 mM NaCl (Wako Pure Chemical Industries, Osaka, Japan) and 0.1% poly-
only for better understanding of the mechanism of organogen- vinyl alcohol (Sigma) and were cultured for about 1 hr until injection.
esis but also as an initial step toward the ultimate regenerative For micromanipulation, ESCs or iPSCs were trypsinized and suspended in
medicine of the future. ESC or iPSC culture medium. A piezo-driven micromanipulator (Prime Tech,

Cell 142, 787–799, September 3, 2010 ª2010 Elsevier Inc. 797


Tokyo, Japan) was used to drill zona pellucida and trophectoderm under the alpha-1,3-galactosyltransferase knockout pigs by nuclear transfer cloning.
microscope and 10–15 ESCs or iPSCs were introduced into blastocyst cavities Science 295, 1089–1092.
near the inner cell mass. After blastocyst injection, embryos underwent follow- Li, P., Tong, C., Mehrian-Shai, R., Jia, L., Wu, N., Yan, Y., Maxson, R.E.,
up culture for 1–2 hr. Mouse blastocysts then were transferred into the uteri Schulze, E.N., Song, H., Hsieh, C.L., et al. (2008). Germline competent embry-
of pseudopregnant recipient ICR female mice (2.5 dpc) and rat blastocysts onic stem cells derived from rat blastocysts. Cell 135, 1299–1310.
were transferred into the uteri of pseudopregnant recipient Wistar female
McCune, J.M., Namikawa, R., Kaneshima, H., Shultz, L.D., Lieberman, M., and
rats (3.5 dpc).
Weissman, I.L. (1988). The SCID-hu mouse: murine model for the analysis of
human hematolymphoid differentiation and function. Science 241, 1632–1639.
SUPPLEMENTAL INFORMATION
Mercer, D.F., Schiller, D.E., Elliott, J.F., Douglas, D.N., Hao, C., Rinfret, A.,
Addison, W.R., Fischer, K.P., Churchill, T.A., Lakey, J.R., et al. (2001). Hepatitis
Supplemental Information includes Extended Experimental Procedures and
C virus replication in mice with chimeric human livers. Nat. Med. 7, 927–933.
seven figures and can be found with this article online at doi:10.1016/j.cell.
2010.07.039. Miura, K., Okada, Y., Aoi, T., Okada, A., Takahashi, K., Okita, K., Nakagawa,
M., Koyanagi, M., Tanabe, K., Ohnuki, M., et al. (2009). Variation in the safety
of induced pluripotent stem cell lines. Nat. Biotechnol. 27, 743–745.
ACKNOWLEDGMENTS
Muller, S.M., Terszowski, G., Blum, C., Haller, C., Anquez, V., Kuschert, S.,
We thank Ryo Sumazaki for critical advice on this work, Kazuya Ise and Carmeliet, P., Augustin, H.G., and Rodewald, H.R. (2005). Gene targeting of
Hirofumi Noguchi for technical advice on islet isolation and islet transplanta- VEGF-A in thymus epithelium disrupts thymus blood vessel architecture.
tion, and Naoki Iwamori for technical advice on embryo manipulation. This Proc. Natl. Acad. Sci. USA 102, 10587–10592.
work was supported by grants from JST, the Ministry of Education, Culture, Nagashima, H., Giannakis, C., Ashman, R.J., and Nottle, M.B. (2004). Sex
Sport, Science, and Technology, Japan. H.N. is a founder and shareholder differentiation and germ cell production in chimeric pigs produced by inner
of ReproCELL Inc. cell mass injection into blastocysts. Biol. Reprod. 70, 702–707.
Nagy, A., Gertsenstein, M., Vintersten, K., and Behringer, R. (2003). Manipu-
Received: January 25, 2010 lating the Mouse Embryo: A Laboratory Manual, Third Edition (Cold Spring
Revised: May 5, 2010 Harbor, NY: Cold Spring Harbor Laboratory Press).
Accepted: June 30, 2010
Nakagawa, M., Koyanagi, M., Tanabe, K., Takahashi, K., Ichisaka, T., Aoi, T.,
Published: September 2, 2010
Okita, K., Mochiduki, Y., Takizawa, N., and Yamanaka, S. (2008). Generation of
induced pluripotent stem cells without Myc from mouse and human fibro-
REFERENCES blasts. Nat. Biotechnol. 26, 101–106.
Nichols, J., and Smith, A. (2009). Naive and primed pluripotent states. Cell
Buehr, M., Meek, S., Blair, K., Yang, J., Ure, J., Silva, J., McLay, R., Hall, J.,
Stem Cell 4, 487–492.
Ying, Q.L., and Smith, A. (2008). Capture of authentic embryonic stem cells
from rat blastocysts. Cell 135, 1287–1298. Niwa, H., Miyazaki, J., and Smith, A.G. (2000). Quantitative expression of Oct-
Chen, J., Lansford, R., Stewart, V., Young, F., and Alt, F.W. (1993). RAG-2- 3/4 defines differentiation, dedifferentiation or self-renewal of ES cells. Nat.
deficient blastocyst complementation: an assay of gene function in lympho- Genet. 24, 372–376.
cyte development. Proc. Natl. Acad. Sci. USA 90, 4528–4532. Offield, M.F., Jetton, T.L., Labosky, P.A., Ray, M., Stein, R.W., Magnuson,
Deltour, L., Leduque, P., Paldi, A., Ripoche, M.A., Dubois, P., and Jami, J. M.A., Hogan, B.L., and Wright, C.V. (1996). PDX-1 is required for pancreatic
(1991). Polyclonal origin of pancreatic islets in aggregation mouse chimaeras. outgrowth and differentiation of the rostral duodenum. Development 122,
Development 112, 1115–1121. 983–995.

Fehilly, C.B., Willadsen, S.M., and Tucker, E.M. (1984). Interspecific chimae- Ogawa, S., Sato, K., and Hashimoto, H. (1971). In vitro culture of rabbit ova
rism between sheep and goat. Nature 307, 634–636. from the single cell to the blastocyst stage. Nature 233, 422–424.

Fraidenraich, D., Stillwell, E., Romero, E., Wilkes, D., Manova, K., Basson, Oh, S.H., Miyoshi, K., and Funahashi, H. (1998). Rat oocytes fertilized in modi-
C.T., and Benezra, R. (2004). Rescue of cardiac defects in id knockout fied rat 1-cell embryo culture medium containing a high sodium chloride
embryos by injection of embryonic stem cells. Science 306, 247–252. concentration and bovine serum albumin maintain developmental ability to
the blastocyst stage. Biol. Reprod. 59, 884–889.
Goldstein, R.S., Drukker, M., Reubinoff, B.E., and Benvenisty, N. (2002). Inte-
gration and differentiation of human embryonic stem cells transplanted to the Okabe, M., Ikawa, M., Kominami, K., Nakanishi, T., and Nishimune, Y. (1997).
chick embryo. Dev. Dyn. 225, 80–86. ‘Green mice’ as a source of ubiquitous green cells. FEBS Lett. 407, 313–319.

Gotoh, M., Maki, T., Kiyoizumi, T., Satomi, S., and Monaco, A.P. (1985). An Okabe, M., Otsu, M., Ahn, D.H., Kobayashi, T., Morita, Y., Wakiyama, Y., Ono-
improved method for isolation of mouse pancreatic islets. Transplantation dera, M., Eto, K., Ema, H., and Nakauchi, H. (2009). Definitive proof for direct
40, 437–438. reprogramming of hematopoietic cells to pluripotency. Blood 114, 1764–1767.
Hirabayashi, M., Kato, M., Kobayashi, T., Sanbo, M., Yagi, T., Hochi, S., and Okita, K., Nakagawa, M., Hyenjong, H., Ichisaka, T., and Yamanaka, S. (2008).
Nakauchi, H. (2009). Establishment of rat embryonic stem cell lines that can Generation of mouse induced pluripotent stem cells without viral vectors.
participate in germline chimeras at high efficiency. Mol. Reprod. Dev. 77, 94. Science 322, 949–953.
James, D., Noggle, S.A., Swigut, T., and Brivanlou, A.H. (2006). Contribution of Rossant, J. (2007). Stem cells and lineage development in the mammalian
human embryonic stem cells to mouse blastocysts. Dev. Biol. 295, 90–102. blastocyst. Reprod. Fertil. Dev. 19, 111–118.
Kamel-Reid, S., and Dick, J.E. (1988). Engraftment of immune-deficient mice Rossant, J., and Frels, W.I. (1980). Interspecific chimeras in mammals:
with human hematopoietic stem cells. Science 242, 1706–1709. successful production of live chimeras between Mus musculus and Mus caroli.
Kroon, E., Martinson, L.A., Kadoya, K., Bang, A.G., Kelly, O.G., Eliazer, S., Science 208, 419–421.
Young, H., Richardson, M., Smart, N.G., Cunningham, J., et al. (2008). Pancre- Rossant, J., Mauro, V.M., and Croy, B.A. (1982). Importance of trophoblast
atic endoderm derived from human embryonic stem cells generates glucose- genotype for survival of interspecific murine chimaeras. J. Embryol. Exp.
responsive insulin-secreting cells in vivo. Nat. Biotechnol. 26, 443–452. Morphol. 69, 141–149.
Lai, L., Kolber-Simonds, D., Park, K.W., Cheong, H.T., Greenstein, J.L., Im, Satoh, M., Yasunami, Y., Matsuoka, N., Nakano, M., Itoh, T., Nitta, T., Anzai,
G.S., Samuel, M., Bonk, A., Rieke, A., Day, B.N., et al. (2002). Production of K., Ono, J., Taniguchi, M., and Ikeda, S. (2007). Successful islet transplantation

798 Cell 142, 787–799, September 3, 2010 ª2010 Elsevier Inc.


to two recipients from a single donor by targeting proinflammatory cytokines in Takahashi, K., and Yamanaka, S. (2006). Induction of pluripotent stem cells
mice. Transplantation 83, 1085–1092. from mouse embryonic and adult fibroblast cultures by defined factors. Cell
126, 663–676.
Shinohara, T., Kato, M., Takehashi, M., Lee, J., Chuma, S., Nakatsuji, N.,
Kanatsu-Shinohara, M., and Hirabayashi, M. (2006). Rats produced by inter- Tarkowski, A.K. (1962). Inter-specific transfers of eggs between rat and
species spermatogonial transplantation in mice and in vitro microinsemination. mouse. J. Embryol. Exp. Morphol. 10, 476–495.
Proc. Natl. Acad. Sci. USA 103, 13624–13628. Ueno, H., and Weissman, I.L. (2006). Clonal analysis of mouse development
reveals a polyclonal origin for yolk sac blood islands. Dev. Cell 11, 519–533.
Slack, J.M. (1995). Developmental biology of the pancreas. Development 121,
1569–1580. Ueno, H., Turnbull, B.B., and Weissman, I.L. (2009). Two-step oligoclonal
development of male germ cells. Proc. Natl. Acad. Sci. USA 106, 175–180.
Stanger, B.Z., Tanaka, A.J., and Melton, D.A. (2007). Organ size is limited by
Ying, Q.L., Stavridis, M., Griffiths, D., Li, M., and Smith, A. (2003). Conversion
the number of embryonic progenitor cells in the pancreas but not the liver.
of embryonic stem cells into neuroectodermal precursors in adherent mono-
Nature 445, 886–891.
culture. Nat. Biotechnol. 21, 183–186.
Stern, M.S. (1973). Letter: Chimaeras obtained by aggregation of mouse eggs Ying, Q.L., Wray, J., Nichols, J., Batlle-Morera, L., Doble, B., Woodgett, J.,
with rat eggs. Nature 243, 472–473. Cohen, P., and Smith, A. (2008). The ground state of embryonic stem cell
Takahashi, K., Tanabe, K., Ohnuki, M., Narita, M., Ichisaka, T., Tomoda, K., self-renewal. Nature 453, 519–523.
and Yamanaka, S. (2007). Induction of pluripotent stem cells from adult human Zeilmaker, G.H. (1973). Fusion of rat and mouse morulae and formation of
fibroblasts by defined factors. Cell 131, 861–872. chimaeric blastocysts. Nature 242, 115–116.

Cell 142, 787–799, September 3, 2010 ª2010 Elsevier Inc. 799


Profiling by Image Registration
Reveals Common Origin of Annelid
Mushroom Bodies and Vertebrate Pallium
Raju Tomer,1,* Alexandru S. Denes,1,2 Kristin Tessmar-Raible,1,3 and Detlev Arendt1,*
1Developmental Biology Unit, European Molecular Biology Laboratory, D-69117 Heidelberg, Germany
2Present address: Biozentrum, University of Basel, Klingelbergstrasse 50/70, CH-4056 Basel, Switzerland
3Present address: Max F. Perutz Laboratories, Campus Vienna Biocenter, Dr. Bohr-Gasse 9/4, A-1030 Vienna, Austria

*Correspondence: tomer@embl.de (R.T.), arendt@embl.de (D.A.)


DOI 10.1016/j.cell.2010.07.043

SUMMARY discrimination, as well as in olfactory learning and memory


(Heisenberg, 2003; Strausfeld et al., 2009). Anatomical, struc-
The evolution of the highest-order human brain tural, and functional similarities of the evolutionarily more ancient
center, the ‘‘pallium’’ or ‘‘cortex,’’ remains enigmatic. parts of the vertebrate pallium, the ‘‘paleopallium’’ and ‘‘archipal-
To elucidate its origins, we set out to identify related lium,’’ with, for example, mushroom bodies of neopteran insects
brain parts in phylogenetically distant animals, to have long been noted and interpreted as convergent acquisi-
then unravel common aspects in cellular composi- tions as a result of functional constraints in independent evolu-
tionary lineages (Farris, 2005, 2008; Strausfeld et al., 1998).
tion and molecular architecture. Here, we compare
Also, the evolution of characteristic folds and fissures and the
vertebrate pallium development to that of the mush- subpartitioning into functional units have occurred indepen-
room bodies, sensory-associative brain centers, in dently in the mammalian pallium and several times in insect
an annelid. Using a newly developed protocol for mushroom bodies (Farris, 2008). These multiple convergences,
cellular profiling by image registration (PrImR), we however, do not rule out the possibility that vertebrate pallium
obtain a high-resolution gene expression map for the and invertebrate mushroom bodies ultimately trace back to
developing annelid brain. Comparison to the verte- a common evolutionary precursor. Some less elaborate,
brate pallium reveals that the annelid mushroom sensory-associative brain centers may have already existed in
bodies develop from similar molecular coordinates the protostome ancestor (of, among others, insects and anne-
within a conserved overall molecular brain topology lids) or even in the last common ancestor of protostomes and
and that their development involves conserved deuterostomes (to which vertebrates belong). These may have
given rise to vertebrate pallium and invertebrate mushroom
patterning mechanisms and produces conserved
bodies alike.
neuron types that existed already in the proto- We set out to decide between the two alternatives—indepen-
stome-deuterostome ancestors. These data indicate dent evolution versus common origin of pallium and mushroom
deep homology of pallium and mushroom bodies bodies from ancient sensory-associative brain centers—by the
and date back the origin of higher brain centers to detailed molecular comparison of their development and differ-
prebilaterian times. entiation. Previous studies revealed conserved expression of
various transcription factors in vertebrate and invertebrate fore-
INTRODUCTION brains, but a specific comparison of gene expression in associa-
tive brain centers has not been undertaken. For this, we investi-
In human and other mammals, the pallium represents the most gated mushroom body development in the marine annelid
highly developed part of the forebrain, the site of learning and Platynereis dumerilii (Fischer and Dorresteijn, 2004), to then
memory (Kandel et al., 2000). It harbors huge densities of inter- compare to available vertebrate data. Platynereis is an ‘‘errant’’
neurons arranged in cortical layers around a central neuropil annelid with huge mushroom bodies (Müller, 1973) that actively
(hence, ‘‘cortex’’; Figure 1). The pallium is less elaborate in other explores the environment and shows some sort of learning
vertebrates and generally thought to function as a sensory-asso- (Evans, 1966). Fortunately, for the purpose of genetic compar-
ciative center integrating primarily olfactory information (Nieu- ison, the Platynereis transcriptome has proven to be ‘‘slow
wenhuys, 2002). Insects and spiders, but also annelids, form evolving,’’ exhibiting less evolutionary change than that of other
similar brain centers with densely packed neurons that send protostomes with mushroom bodies (Drosophila, Apis) (Raible
out thousands, in some species hundreds of thousands, of et al., 2005). Also, in line with a slower evolutionary pace of the
approximately parallel processes that form a central, lobed annelid lineage, we have shown that Platynereis neural
neuropil (Strausfeld et al., 1998) (Figure 1). These likewise repre- patterning exhibits ancient characteristics, many of which are
sent sensory-associative brain centers implicated in olfactory shared with the vertebrates but not with the faster-evolving fly

800 Cell 142, 800–809, September 3, 2010 ª2010 Elsevier Inc.


and nematode lineages (Denes et al., 2007; Tessmar-Raible
et al., 2007). Finally, Platynereis is easy to breed and thus acces-
sible to high-throughput approaches (Fischer and Dorresteijn,
2004), as a prerequisite for the analysis of multiple genes.
Previous comparisons of brain development between phylo-
genetically remote species have been restricted to gene-by-
gene comparisons that did not allow, or allowed only in a coarse
manner, relating expression data spatially for multiple genes. To
overcome this, we have established a new protocol for Profiling
by Image Registration (PrImR) that, for the first time, allows align-
ment of expression images with cellular resolution and thus
expression profiling for the whole brain at the single-cell level.
Our protocol builds on the highly stereotypical nature of Platyner-
eis neurodevelopment to align expression patterns for an unlim-
ited number of genes in an additive process.
Using PrImR, we first investigated whether the overall molec-
ular topography of the brain anlage is conserved between
annelid and vertebrate and whether the Platynereis mushroom
bodies develop from corresponding molecular regions when
compared to vertebrate pallium (as a test for homology) (Arendt,
2005; Woodger, 1945). We also tested whether the positioning of
both follows similar developmental patterning mechanisms.
Next, we used PrImR to determine the expression profile for
specific subregions, focusing on specifically expressed genes
making up the regulatory and differentiation signature of cells
developing from these regions and compared this ‘‘molecular
fingerprint’’ (Arendt, 2008) to that of vertebrate telencephalic
neuron types but also to that of the mushroom body neuroblasts
in Drosophila. We conclude that pallium and mushroom body
most likely evolved from the same (sensory associative) brain
center that was already present in the bilaterian ancestors.

RESULTS

In Silico Alignment of Gene Expression Patterns


To establish a high-resolution molecular topography of the
annelid brain and to investigate the molecular fingerprint of brain
subregions, we set out to obtain coexpression information for
a large number of specifically expressed genes. This is impos-
sible to achieve at large scale by two-color double whole-mount
Figure 1. Comparison of Vertebrate Pallium and Annelid Mushroom
in situ hybridization (Tessmar-Raible et al., 2005), because this
Body Development Based on Previous Work and on This Study
(A–E) Development of the mouse pallium from open neural plate early specifi-
would require a nonlinearly increasing amount of technically
cation (5 s; A and B) to E10.5 late specification stage (C and D) and adult (E). demanding experiments. To overcome this, we took advantage
(F–J) Development of the Platynereis mushroom bodies from 48 hpf specifica- of the observation that Platynereis development is highly stereo-
tion stage (F–I) to adult (J). Note that in mouse, elaborate morphogenesis typic and synchronous within and across batches (Fischer and
(neural tube and telencephalic vesicle formation) occurs between early and Dorresteijn, 2004) and developed a computational protocol for
late specification stages, which does not take place in the annelid.
(A) The mouse pallium (PAL) anlage with reference to the anterior-posterior
(otx, gbx, hox) and mediolateral (pax6, nkx2.1, nkx2.2) regional patterning
systems. (After Inoue et al. [2000] and Shimamura et al. [1995].) (F) The Platynereis mushroom body anlage with reference to the anterior-
(B) Expression of bf-1 (orange) outlining the telencephalon anlage including the posterior (otx, gbx, hox) and mediolateral (pax6, nkx2.1, nkx2.2) regional
pallium. Wnt5/8 and Shh indicate spatially restricted activity of signaling patterning systems. MB, mushroom bodies.
ligands implicated in telencephalon development. (G) Expression of bf-1 (orange) in the annelid brain at 48 hpf. Wnt5/8 and Hh
(C) Regionalization of telencephalic vesicles into pallium (PAL) and lateral and indicate spatially restricted expression of conserved ligands.
medial ganglionic eminences (LGE, MGE). Colored strip indicates level of (H) Brain molecular topography of the bf-1+ region at 48 hpf as revealed by this
cross-section in (D). M, mesencephalon; HT, hypothalamus, Eye, position of study. Color code refers to (I).
cutoff eye stalk. (I) Gene expression in the Platynereis brain indicated by black bars and color
(D) Gene expression along the telencephalic section in (C) indicated by black code referring to (H).
bars and color code. For references, see the main text. (J) The Platynereis adult mushroom bodies, parasagittal section.
(E) Parasagittal section of the mouse cortex. See also Figure S2.

Cell 142, 800–809, September 3, 2010 ª2010 Elsevier Inc. 801


Figure 2. In Silico Alignment of Expression
Patterns
(A) Maximum Z projection of average reference
axonal scaffold image of 48 hpf larval brain.
(B–D) Step-wise alignment of an axonal scaffold
image to the average reference scaffold (cyan)
using rigid, affine, and smoothened freeform
nonrigid transformations. Images are 20 mm thick
optical sections. See Movies S1 and S2 for details.
(E) A positive control experiment to test the accu-
racy of the alignment algorithm. All images are
maximum Z projections of aligned expression
patterns of r-opsin, acquired from three different
larvae, and an RGB merge of them. See Movie
S3 for details.
(F and G) Experimental validation of an in silico co-
expression (F) using double fluorescence WMISH
double staining (G). White, colocalization of green
and red pixels. Cyan and blue arrows point to veri-
fied coexpression.
(H) Violin plot illustrating the distribution of Pearson’s correlation coefficients of 171 gene expression images with their gene-specific average images. Magenta
shows the data density. Mean = 0.900, median = 0.904, standard deviation = 0.040.
Scale bars represent 50 mm. See also Figure S1 and Movies S1, S2, and S3.

accurately aligning the expression patterns of different genes The Molecular Topography of the Platynereis Brain
acquired from different stained individuals of identical develop- We used our PrImR protocol to determine the molecular topog-
mental stage (Profiling by Image Registration, PrImR, j,primərj). raphy of the Platynereis brain after 2 days of development, to test
In the first step, two-channel image stacks were acquired via whether any of the Platynereis forebrain regions would show
whole-mount reflection confocal laser-scanning microscopy ‘‘telencephalon-like’’ coordinates. At this stage, major subre-
(Jékely and Arendt, 2007). One channel contained information gions of the developing Platynereis nervous system are already
on the expression pattern of a given gene and the other on the established and larger populations of neurons have started
axonal scaffold of the Platynereis larval brain. More than three differentiation (Tessmar-Raible et al., 2007). For our purposes,
biological replicas were acquired for every gene. Next, we it was sufficient to focus on one stage only, because in the
used the axonal scaffold channel to align these images to a absence of drastic morphogenetic changes (such as neurulation
reference average axonal scaffold image (Figure 2A, Figure S1 and vesicle formation in vertebrates), gene expression patterns
available online), first via rigid alignment algorithms and then in the developing Platynereis brain remain spatially constant
by smooth nonrigid transformation (see the Experimental Proce- over time (as shown for example for pax6, six1/2 [Arendt et al.,
dures) (Figures 2B–2D). For each gene, a normalized average 2002] or dach and bf-1 [Figures 5 and 6]), indicating colocaliza-
expression image from (in most cases) five individuals was tion and temporal co-occurrence of differentiated neurons and
thus generated (Figure S1B) that was directly comparable to their precursors.
those of an unlimited number of other genes. As a test for accu- We had previously shown that both the annelid and vertebrate
racy, we obtained almost perfect overlap of signal for single cells brains exhibit a basic subdivision into medial nk2.1+ and lateral
stained for the same gene in different individuals (Figure 2E). We pax6+ subregions (Tessmar-Raible et al., 2007) (Figure 1A)
also found that the coexpression images obtained by our PrImR (transformed into a ventral-dorsal arrangement during vertebrate
protocol fully reproduced those obtained by double-fluorescent neurulation; Figures 1A–1C). It is within this conserved frame-
WMISH (Tessmar-Raible et al., 2005) (compare Figures 2F and work that the vertebrate telencephalon anlage is established,
2G) but tended to be more ‘‘complete,’’ reflecting the higher with the pallium developing from the anterior part of the pax6+
sensitivity of the NBT/BCIP staining and of the reflection micros- region (Figure 1A). The outline of the whole telencephalon anlage
copy technique (Jékely and Arendt, 2007). Finally, to challenge is demarcated early on by expression of bf-1 (Hébert and Fishell,
the accuracy of the protocol, we systematically estimated the 2008) (orange in Figure 1B), a crucial regulator of telencephalic
extent of overlap between individually aligned scans by calcu- development (Danesin et al., 2009). In the course of telence-
lating the Pearson’s correlation coefficient for 171 individually phalic morphogenesis, gsx+ and emx+ (Kimura et al., 2005)
aligned scans with the gene-specific average image (see the domains complement the nk2.1+ and pax6+ subregions (Figures
Experimental Procedures) and found that the probability is 1C and 1D) such that the gsx+, pax6+ overlap demarcates the
highest to obtain a value above 0.9, which implies that the future pallial-subpallial boundary (Danesin et al., 2009; Yun
average expression images used for this study should reliably et al., 2001) and pax6+, emx+ coexpression specifies pallial
reproduce endogenous gene expression. Our protocol thus telencephalic precursors (Kimura et al., 2005). Besides bf-1,
allows the comparison of any newly added expression pattern lhx2 (expressed broadly in the telencephalon and acting highly
to all preexisting patterns at once, with high accuracy and in up in the hierarchy of cortical induction (Hébert and Fishell,
cellular resolution. 2008), COUP-TF1/seven-up (contributing to telencephalic

802 Cell 142, 800–809, September 3, 2010 ª2010 Elsevier Inc.


Figure 3. Coexpression of ‘‘Telencephalic’’
and Other Marker Genes, Red, with bf-1,
Green, in the Platynereis 48 hpf Larval Brain
(A and B) Complete expression of bf-1 (green) (A).
Blue, reference axonal scaffold. Dotted circle
marks expression in the eye field, as indicated
by r-opsin coexpression in (B).
(C–E) genes covering the almost entire bf-1
domain.
(F–I) Lateral-to-medial sequence of regionalisa-
tion genes.
(J) Five-micron-thick optical section showing
lateral-to-medial coexpression of emx (green),
pax6(red), and gsx (cyan).
(K–O) Genes involved in mediolateral signaling.
(P–T) Genes showing mediolaterally restricted
expression.
All images are Maximum Z projections of slice
numbers 13 to 40, i.e., 28 mm thick (except K:
8–28, i.e., 21 mm thick) in the reference scaffold
image stack. White marks colocalizing pixels.
Scale bars represent 50 mm. See also Figure S5.

patterning (Hébert and Fishell, 2008) and soxB family members patterning can be perturbed by ectopic b-catenin activation
(sox1[Ekonomou et al., 2005] and sox2 [Bani-Yaghoub et al., (mimicking the dorsal signal), which accordingly triggers the
2006]) play important roles in early telencephalic development upregulation of dorsal pallial and downregulation of ventral sub-
(Figure 3E), although the spatial distribution and coexpression pallial markers (Backman et al., 2005). Investigating the expres-
of these factors along the anterior-posterior and mediolateral sion of orthologous signals and factors in Platynereis, we indeed
brain axes is not fully resolved for the vertebrates. detected Hh in the medial (Figure 3K) and Wnt8 in the lateral brain
We accordingly examined bf-1, lhx2, svp, soxB, pax6, emx, (Figure 3L), matching the vertebrate situation. We also observed
gsx, and nk2.1 expression in Platynereis and indeed identified lateral expression of Wnt5 (Figure 3M), another dorsal Wnt signal
a brain region with telencephalon-like molecular topography implicated in corticospinal axon guidance (Keeble et al., 2006), of
(Figures 3A–3I; summarized in Figure 1). First, we found bf-1 WntA (not found in vertebrates; Figure 3N) and Gli (Figure 3O),
expressed in a horseshoe-shaped domain in the Platynereis a zinc finger transcription factor essential for dorsal telencephalic
brain (Figure 3A), where it is specifically coexpressed with lhx2, fates (Tole et al., 2000). None of the other Platynereis Wnt genes
svp, and soxB (Figures 3C–3E). (The more lateral part of bf-1+ was expressed in the brain (data not shown; note that Wnt3 does
domain represents the eye anlage; Figure 3B.) Second, from not exist in annelids). For Platynereis, exposure to the chemical
lateral to medial, the Platynereis bf-1 brain domain is subdivided compound 1-Azakenpaullone has been established as an effi-
by spatially restricted coexpression of emx, pax6, gsx, and nk2.1 cient means to ectopically induce b-catenin activation (Schneider
(Figures 3F–3J) in a telencephalon-like fashion. Nowhere else and Bowerman, 2007). We accordingly tested the effect of 1-Aza-
in the developing Platynereis larva was a similar sequence of kenpaullone on Platynereis brain regionalization and observed
emx+/pax6+, pax6+, pax6+/gsx+, gsx+, and gsx+/nk2.1+ subre- upregulation of lateral emx expression (Figures 4B and 4G),
gions (Figure 3J) detected. The emx+/pax6+ and pax6+ regions downregulation of intermediate gsx (Figures 4D and 4I), and
within the bf1+ domain thus represented candidate counterpart medial nk2.1 expression (Figures 4E and 4J), while pax6 ap-
regions to the vertebrate pallium anlage, a notion that we peared unaffected (Figures 4C and 4H). These results mirror
decided to explore further. vertebrate telencephalic patterning (Figure 1) and are thus
strongly indicative of evolutionary conservation.
Comparison of Patterning Mechanisms
In vertebrates, bf-1 coordinates the activity of two opposing The Molecular Fingerprint of Mushroom Body Neurons
signaling centers that pattern the telencephalon anlage. bf-1 In vertebrates, distinct combinations of differentially expressed
acts downstream of the ventral signal, Hh, to induce ventral transcription factors control the fate of the various telencephalic
(subpallial) identities. At the same time, bf-1 inhibits dorsal subregions. We took advantage of PrImR to further determine
Wnt/b-catenin signaling through direct transcriptional repression and compare molecular fingerprints. In mouse, dorsal telence-
of Wnt8 (Danesin et al., 2009), which induces dorsal (pallial) phalic (pallial) pax6+ regions show differential activity of Dach
identities (Houart et al., 2002). Vertebrate telencephalic (Caubit et al., 1999) and ngn1/2, bHLH factors required for pallial

Cell 142, 800–809, September 3, 2010 ª2010 Elsevier Inc. 803


Figure 4. Differential Gene Regulation in the Developing Platynereis Brain
Expression of bf-1, emx, pax6, gsx, and nk2.1 in DMSO control (A–E) and 1-Azakenpaullone (1 mM final concentration) treated 48 hpf larval brain (F–J). Changes in
expression patterns are highlighted by dashed circles. Blue, nuclear stain DAPI; green, immunostaining against acetylated tubulin. n for bf-1: treated > 200,
control > 200; emx: treated > 380, control > 300; pax6: treated > 350, control > 310; gsx: treated > 150, control > 160; nk2.1: treated > 320, control > 200.
Scale bars represent 50 mm.

neurogenesis (Nieto et al., 2001). In Platynereis, dach and ngn fingerprint (bf1+, emx+, pax6+, dach+, svp+, tll+, ngn+, asc+)
are similarly restricted to emx+, pax6+ subregions (Figures 3P qualified as possible evolutionary counterpart to the vertebrate
and 3Q; compare Figures 1D and 1I). In mouse, ventral (subpal- pallium anlage (encircled by stippled line in Figures 1C and 1D),
lial) gsx+ regions coexpress vax1, required for GABAergic inter- we set out to determine whether this paired region indeed
neuron generation (Taglialatela et al., 2004), and coe/ebf1, an represented the Platynereis mushroom body anlagen by tracing
HLH transcription factor affecting subpallial development (Garel mushroom body development from adult (Figure 5, Figure S3,
et al., 1999). We accordingly found both genes coexpressed in Movie S4) and juvenile stages (10 days postfertilization [dpf],
Platynereis gsx+ regions (Figures 3R and 3S; compare Figures when they are fully developed and can be easily identified
1D and 1I). In mouse, telencephalic subregions express er81, histologically) back to earlier larval stages. We combined
an ETS factor active in distinct subpallial domains (Yun et al., anatomical (the specific shape of mushroom bodies), histolog-
2001) and direct target of pax6 in the dorsal pallium (Tuoc ical (the dense packing of neurons), and topological (the specific
and Stoykova, 2008). Platynereis er81 is similarly expressed spatial arrangement of mushroom bodies and palpal and
in the pax6+/gsx+ and gsx+/nk2.1+ subregions but not in antennal nerves) evidence, as well as the expression of mush-
between (Figure 3T; compare Figures 1D and 1I). Adding to room body-specific marker genes (dach and pax6), to reidentify
this, expression of Platynereis genes orthologous to ascl1/ and position the developing mushroom bodies at various stages
mash1 (bHLH downstream effector of gsx1 [Wang et al., 2009]; (6 weeks, 20, 10, 9, 8, 7, 6, 5, 4, 3, and 2 dpf; Figure 5 and data
expressed in ventral and dorsal telencephalon [Yun et al., not shown). This allowed unambiguous positioning of the mush-
2001]), to brn1/2/4 (POU domain transcription factors required room body anlagen to ventrolateral coordinates at 48 hr postfer-
for cortical migration in dorsal and ventral telencephalon [Ekono- tilization [hpf] that indeed matched the emx+, pax6+, dach+
mou et al., 2005; McEvilly et al., 2002]), and to many other genes candidate region.
implicated in telencephalon development by function or by In juvenile worms, the differentiating emx+, pax6+ mushroom
expression is detailed for Platynereis in Figure S2 and has bodies and the more medial gsx+, nk2.1+ brain tissue (referred
been used to generate the molecular map in Figure 1H. The over- to as ‘‘pars intercerebralis’’[Müller, 1973]) continued to express
all comparison suggests that the molecular fingerprints of bf-1 (Figure 6A) and started to specifically express arx (Fig-
annelid and vertebrate emx+/pax6+, pax6+, pax6+/gsx+, gsx+, ure 6B; expressed in mouse and fish telencephalon and ventral
and gsx+/nk2.1+ subregions are similar to large extent but may thalamus [Miura et al., 1997]). We also detected specific expres-
also differ in the detail (for example, dlx is expressed more sion of two conserved bilaterian microRNAs, miR-9 and mir-9*
broadly in annelid than in vertebrate; rx is expressed in the (Christodoulou et al., 2010) (Figures 6C and 6D). In the verte-
emx+, pax6+, and gsx+ subregions in the annelid but excluded brates, these microRNAs are expressed only in the telenceph-
in the vertebrate; Figure S2). alon, among all differentiated CNS tissues (Deo et al., 2006).
Regarding transmitter usage, large part of the Platynereis mush-
Positioning the Platynereis Mushroom Body Anlagen room bodies proved glutamatergic by expression of vglut,
Since our molecular comparison had identified one unique encoding the vesicular glutamate transporter (Figure 6F). In
candidate region in the Platynereis brain, which, by position, contrast, gad, a marker for GABAergic neurons, was restricted
similar responsiveness to Wnt signaling, and similar molecular to more medial brain tissue (Figure 6E), as was the dopaminergic

804 Cell 142, 800–809, September 3, 2010 ª2010 Elsevier Inc.


Figure 5. Platynereis Mushroom Body
Development
Three-dimensional models were generated for
various stages according to confocal image
stacks (see Movie S4 for details). Depicted struc-
tures represent mushroom body pedunculi (red),
mushroom body perikarya (transparent red),
antennal nerve (dark blue), pars intercerebralis
(purple), eyes (yellow), palpae (green), and
a glomerulus-like structure (mediates the connec-
tion between palpae and mushroom bodies; light
green). Continuous expression of dach (top and
bottow row) in mushroom body neurons consis-
tent with anatomical backtracing. Blue arrows
point to the antennal nerve. A, P, D, V, L, and R
indicate body axes.
Scale bars represent 50 mm. See also Figure S3
and Movie S4.

data not shown), indicative of evolu-


tionary conservation. To challenge
homology of insect and annelid mush-
room bodies, we looked for a combination
of transcription factors that, by
coexpression, uniquely define Drosophila
marker tyrosine hydroxylase (Figure 6H), again matching the mushroom body neuroblasts independent of pax6 and dach—to
vertebrate situation (Marı́n and Rubenstein, 2001). However, then test whether a similar coexpression would locate to the
we also detected a difference in the spatial distribution of trans- annelid mushroom body anlage in an equally specific manner.
mitter type in that Platynereis mushroom bodies also harbored Among the four Drosophila mushroom body neuroblasts, Pcd8
cholinergic neurons (Figure 6G; not found in the vertebrate andPcd9 are uniquely identified by the specific coexpression
pallium). of asc/l’sc, tll, svp, slp/bf-1, and otx (Urbach and Technau,
2003). We determined by PrImR where in the Platynereis 48
Molecular Fingerprint Comparison with Insect hpf brain these five transcription factors would be coexpressed,
Mushroom Bodies and indeed the only two bilateral spots of cells in the Platynereis
Mushroom bodies of similar cellular composition also exist in brain coexpressing them form part of the mushroom body
arthropods such as onychophorans (velvet worms), spiders, anlage (Figures 7C–7H). This is strong evidence in favor of an
and insects, yet ample molecular data exist only for the mush- evolutionary relatedness of insect and annelid mushroom
room bodies in insects. In Drosophila, mushroom body neuro- bodies. However, in contrast to annelid mushroom bodies
blasts require pax6 and dach for their specification but do not (Figure 7I) and to vertebrate pallium, the Drosophila dac+ mush-
express sine oculis (six1/2) or eyes absent (eya) (Noveen et al., room body precursors do not express emx, and the overall
2000). We found that the only region of the Platynereis brain molecular topography is conserved to lesser extent between
coexpressing pax6 and dach but devoid of six1/2 and eya is fly, annelid, and vertebrate (Urbach and Technau, 2003) than
indeed the mushroom body anlage (Figures 7A and 7B and eya between annelid and vertebrate. Our data suggest that this is

Figure 6. Gene Expression in the Brain of


Platynereis Juvenile Worms
Transcription factors (A and B), microRNAs (C and
D) and marker genes (E–H) for GABAergic (gad, E),
glutamatergic (vglut, F), cholinergic (vacht, G),
and dopaminergic (tyrosine hydroxylase, H) neu-
rons. Dashed circles outline mushroom bodies.
A, P, L, and R indicate body axes. Scale bars
represent 50 mm.

Cell 142, 800–809, September 3, 2010 ª2010 Elsevier Inc. 805


Figure 7. Coexpression of Insect Neuro-
blast Marker Genes, Red, with Dach, Green,
in the Platynereis 48 hpf Larval Brain
(A–J) Coexpression of dach (green) with pax6 (A),
six1/2 (B), ash2 (C), tll (D), svp (E), bf-1/slp (F), otx
(G), the intersection of ash2, tll, svp, bf-1/slp, and
otx (H), emx (I), and Wnt5 (J) (red). The intersection
of expression patterns was generated from
expression masks generated by smoothening
images with a Gaussian sigma of 2 and a signal
cutoff of 35. All images are Maximum Z projection
of slice numbers 13 to 40, i.e., 28 mm thick, in the
reference scaffold image stack. White marks
colocalizing pixels.
(K) Phylogenetic distribution of mushroom body
and pallium in Bilateria.
Scale bars represent 50 mm. See also Figure S4.

tions of additional transcription factors


(dach+, svp+, slp+, tll+, ngn+, asc+,
arx+) (Figures 1C, 1D, 1H, 1I, and 6).
They give rise to glutamatergic neurons,
while GABAergic and dopaminergic
neuron types emerge from more medial/
ventral brain regions. They are positive
for the same brain-specific microRNAs
(mir-9, -9*). We are confident that these
due to secondary modifications of mushroom body develop- similarities are too complex to be plausibly explained by indepen-
ment in the insect lineage. dent (convergent) evolution. Instead, we propose that they were in
place already in the protostome-deuterostome ancestor, the last
DISCUSSION common ancestor of annelids and vertebrates (Figure 7K).
To assess the statistical significance of our dataset, we con-
In this study, we have established a new protocol, PrImR, that ducted permutation tests by randomly assigning gene expres-
allows expression profiling by image registration with cellular sion to ‘‘pallium’’ or ‘‘mushroom body,’’ and we found the
resolution for the entire organism. Expression profiling at observed similarity in molecular fingerprint extremely unlikely to
single-cell resolution has been reported previously (Liu et al., arise by chance (Figure S4; p value less than 2.0E-05). It should
2009; Sugino et al., 2006), but our protocol is the first to allow be stressed, however, that this value might slightly overestimate
simultaneous profiling for the whole central nervous system, the significance of our comparison, because a subset of the
where cells are densely packed. Our protocol thus sets the stage involved genes may be coregulated and should be treated as
for a new chapter of evolutionary developmental research that a single observation.
has so far been restricted to gene-by-gene comparisons of Importantly, however, annelid mushroom body and vertebrate
expression patterns with restricted coexpression information, pallium development and specification also differ to consider-
necessarily remaining fragmentary and sometimes inconclusive. able extent, which is plausible given 600 million years of inde-
Instead, PrImR allows much broader comparisons, involving pendent evolution. First, vertebrate pallium development
hundreds of transcription factors and differentiation genes, and involves neurulation and telencephalic vesicle formation (Figures
a much higher level of integration. For the first time, molecular 1B and 1C), which does not occur in the annelid. Second, impor-
fingerprinting as introduced here allows testing homology tant differences are apparent in some expression domains. For
hypotheses such as the comparison of mushroom bodies example, annelid nk2.1 expression exhibits lateral ‘‘wings’’
between annelids and insects and of mushroom bodies with (Figure 1F), where it overlaps with the lateral pax6 domain (care-
vertebrate pallium (Strausfeld et al., 1998) that could not be ful 3D analysis, however, reveals that this overlap is outside of
resolved on merely anatomical and physiological grounds. the bf1+ mushroom body anlage). Also, the developing annelid
Taking advantage of this new protocol, our data reveal that mushroom body is positive for the rx gene (Figure S2), which is
mushroom bodies in annelid and vertebrate pallium develop excluded from the vertebrate telencephalon anlage (Stigloher
from the same, molecularly defined, subregion (emx+/pax6+) at et al., 2006). Finally, some genes are absent in the other species.
similar anterior-posterior and mediolateral coordinates within an For example, Wnt3, expressed in the dorsomedial pallium
overall conserved molecular topography of the brain (Figures 1A anlage, does not exist in annelids, and WntA, demarcating the
and 1F), which is established by similar patterning mechanisms lateral mushroom body anlage (Figure 3N), is not found in the
(Figures 1B and 1G). They express similar and unique combina- vertebrate genomes.

806 Cell 142, 800–809, September 3, 2010 ª2010 Elsevier Inc.


What is the significance of these findings for our under- repeatedly proposed (Farris, 2005, 2008; Strausfeld et al.,
standing of mushroom body and pallium/telencephalon evolu- 1998; Strausfeld et al., 2009). To settle this issue, it will be
tion? We can infer that the last common ancestor of protostomes rewarding to identify brain regions with similar molecular coordi-
and deuterostomes (and thus, of annelids and vertebrates) nates and fingerprint in species lacking overt mushroom body
already possessed distinct brain centers that later gave rise to morphology such as crustaceans or mollusks on the protostome
the pallium in the vertebrate and to mushroom bodies in the side or amphioxus on the deuterostome side (Figure 7K) and
annelid (and arthropod) lineage (Figure 7K). Based on our data, to investigate possible sensory input, integrative function and
it is likely that these centers harbored subsets of glutamatergic locomotor output of the corresponding brain parts. If, on one
neurons that presumably functioned in some sort of integration hand, such less complex, ‘‘mushroom body-related’’ brain parts
of sensory signals. Input was most likely chemosensory, as indeed exist, they will help defining a minimum set of anatomical
deduced from the primarily chemosensory input of pallium and and physiological characteristics associated with the conserved
mushroom bodies alike, in insects, spiders, and onychophorans, molecular fingerprints described here. This way, we would gain
as well as in annelids (Strausfeld et al., 2006). (The Platynereis more insight into anatomy and function of an ancient sensory-
mushroom bodies receive connections from the chemosensory integrative center present in the protostome-deuterostome
palpae [Movie S4].) Notably, however, the sensory input of ancestor. If, on the other, such regions do not exist, we can infer
arthropod mushroom bodies is received by distinct olfactory secondary loss of brain complexity. Possible evolutionary
appendages (Strausfeld et al., 2006) and can even be visual scenarios leading to reduced brain complexity include the tran-
(in spiders, honeybee and cockroach [Strausfeld and Barth, sition to sessile or hemisessile lifestyle with filter feeding, to inter-
1993]), indicating some plasticity in mushroom body connec- stitial burrowing with minute body size, or to parasitism.
tivity. Apparently, the sensory input into sensory associative Once a better and more complete picture of mushroom body-
centers can change according to the needs of changing ecolog- related brain parts in bilaterians is obtained, this will set the stage
ical niches. for a more comprehensive view on the evolution of insect and
We can speculate that the first function of these chemosen- annelid mushroom bodies and vertebrate pallium alike. What
sory integrative brain centers was to distinguish between food do these groups have in common that is not found in bilaterians
and nonfood, to decide about directed locomotion toward in which mushroom body-related brain parts are present but less
identified food sources, and, ultimately, to integrate previous elaborate? This will ultimately help to link mushroom body/
experiences into some sort of ‘‘learning’’ (Evans, 1966). Extrap- pallium evolution to changes in life style, locomotion, or food
olating from the similar motor output of today’s pallium and capture.
mushroom bodies, we can assume that these integrative centers
already produced coordinated locomotion of some sort (Farris, EXPERIMENTAL PROCEDURES
2005).
There is a long-standing notion that the vertebrate pallium Imaging Gene Expression Patterns
comprises distinct parts of different evolutionary age, paleo-, We used whole-mount reflection confocal microscopy (Jékely and Arendt,
archi-, and neopallium. Roughly, the paleopallium corresponds 2007) to acquire gene expression pattern images. Typically, an oil immersion
403 objective on the Leica TCS SPE Confocal Microscope was used.
to olfactory cortex and the archipallium to the hippocampus,
and the neopallium comprises the remainder of the cerebral
Alignment of Expression Patterns
hemispheres (Swanson, 2000). Paleo- and archipallium are
For achieving highly accurate alignments of axonal scaffolds, we developed
found in all vertebrates, while the neopallium is restricted to a multistep multiresolution image registration algorithm using Mutual Informa-
amniotes. In line with this concept, the cellular organization of tion (Viola and Wells, 1995) as the image similarity metric. In the first step,
the more ancient archi- and paleopallium strikingly resembles images were transformed rigidly with six degrees of freedom (three for trans-
that of annelid and arthropod mushroom bodies, in that densely lation and three for rotation) to roughly align the images. In the second step,
packed, basophilic somata send out parallel processes inter- affine transformation with 12 degrees of freedom (three for rotation, three for
translation, three for scaling, and three for shear) was used to compensate
sected by afferent and efferent networks (Strausfeld et al.,
for the shrinking and shearing artifacts introduced by in situ hybridization
1998). Intriguingly, when compared to neopallium, the hippo- and imaging procedures. In the last step, we used a uniform grid of control
campus and dentate gyrus each consist of only one layer of points to model freeform nonrigid local deformations and used third-order
principle neurons (Förster et al., 2006). This contrasts with the B-splines to smoothly interpolate the transformations. For the affine and
six-layered architecture of the neopallium but rather resembles nonrigid registration steps, we used a multiresolution approach in which 3D
the simple architecture of the mushroom bodies. We propose images were smoothened and downsampled to various degrees before the
that such simple architecture, ancient for vertebrates, might alignment.

have been in place in the urbilaterian mushroom body/pallium


sensory-associative precursor structure. Average Reference Axonal Scaffold
We generated an average Platynereis larval axonal scaffold using images
As mentioned in the introduction, it is unlikely that a mushroom
acquired from 36 different individual 48 hpf larvae (Figure S1), which was
body-like shape was already in place in the protostome-deutero- used as reference for all the alignments.
stome ancestor. Rather, it seems plausible that the common
evolutionary precursor structure of mushroom bodies and
Averaging Expression Patterns
pallium was morphologically less complex and that higher For direct comparison of the spatial expression patterns of different genes, we
degrees of histological (and thus, functional) complexity were generated normalized average expression patterns from three or more indi-
acquired independently in divergent evolutionary lineages, as vidual larvae (see Movie S3 and Figure S1B for examples). We used

Cell 142, 800–809, September 3, 2010 ª2010 Elsevier Inc. 807


‘‘Colocalization Highlighter’’ plug-in in ImageJ 1.41e to highlight the colocaliz- Arendt, D. (2008). The evolution of cell types in animals: emerging principles
ing pixels as white pixels. Default settings were used for most of the cases. from molecular studies. Nat. Rev. Genet. 9, 868–882.
Arendt, D., Tessmar, K., de Campos-Baptista, M.I., Dorresteijn, A., and
Alignment Accuracy Estimates Wittbrodt, J. (2002). Development of pigment-cup eyes in the polychaete
We conducted a systematic analysis by testing images from 171 different Platynereis dumerilii and evolutionary conservation of larval eyes in Bilateria.
larvae. Pearson’s correlation coefficient was calculated for each aligned Development 129, 1143–1154.
expression image with the corresponding gene-specific average. Backman, M., Machon, O., Mygland, L., van den Bout, C.J., Zhong, W.,
Taketo, M.M., and Krauss, S. (2005). Effects of canonical Wnt signaling
Modeling Platynereis Brain Anatomy on dorso-ventral specification of the mouse telencephalon. Dev. Biol. 279,
Amira 4.1.1 was used to segment various anatomical structures in the brain 155–168.
(Movie S4) and to generate 3D surface renderings shown in Figure 5. Bani-Yaghoub, M., Tremblay, R.G., Lei, J.X., Zhang, D., Zurakowski, B.,
Sandhu, J.K., Smith, B., Ribecco-Lutkiewicz, M., Kennedy, J., Walker, P.R.,
Ectopic Overexpression of Wnt Canonical Signaling and Sikorska, M. (2006). Role of Sox2 in the development of the mouse
We used an established procedure (Schneider and Bowerman, 2007) of treat- neocortex. Dev. Biol. 295, 52–66.
ment with 1-Azakenpaullone (cat. no. 191500, Calbiochem) to achieve ectopic Caubit, X., Thangarajah, R., Theil, T., Wirth, J., Nothwang, H.G., Rüther, U.,
overexpression of canonical Wnt signaling in Platynereis dumerilii larvae. and Krauss, S. (1999). Mouse Dac, a novel nuclear factor with homology to
A mixture of larvae from two to three different animal pairs was used for Drosophila dachshund shows a dynamic expression in the neural crest, the
treatment. The larvae were treated from 24 hpf to 48 hpf in 6 ml Natural Sea eye, the neocortex, and the limb bud. Dev. Dyn. 214, 66–80.
Water at a final concentration of 1 mM 1-Azakenpaullone or DMSO and were Christodoulou, F., Raible, F., Tomer, R., Simakov, O., Trachana, K., Klaus, S.,
grown at 18 C. The larvae were fixed in 4% PFA/PTW, stored in 100% meth- Snyman, H., Hannon, G.J., Bork, P., and Arendt, D. (2010). Ancient animal
anol at 20 C, and proceeded for in situ hybridization. The staining reactions microRNAs and the evolution of tissue identity. Nature 463, 1084–1088.
were stopped simultaneously for control and treated larvae, much before the
Danesin, C., Peres, J.N., Johansson, M., Snowden, V., Cording, A., Papalo-
saturation.
pulu, N., and Houart, C. (2009). Integration of telencephalic Wnt and hedgehog
signaling center activities by Foxg1. Dev. Cell 16, 576–587.
Cloning Genes
Denes, A.S., Jékely, G., Steinmetz, P.R., Raible, F., Snyman, H., Prud’homme,
We used degenerate primers to clone fragments of brn1/2/4, dach, emx, svp,
B., Ferrier, D.E., Balavoine, G., and Arendt, D. (2007). Molecular architecture of
tll, vglut, and gad and 50 ,30 -RACE extensions to extend the fragments (see the
annelid nerve cord supports common origin of nervous system centralization
Extended Experimental Procedures for primer sequences). We used cDNA
in bilateria. Cell 129, 277–288.
templates generated with SMART technology (SMART RACE cDNA amplifica-
tion kit, Clonetech) from total RNA extracted from larvae at several develop- Deo, M., Yu, J.Y., Chung, K.H., Tippens, M., and Turner, D.L. (2006). Detection
mental stages. Other genes investigated in this study were obtained either of mammalian microRNA expression by in situ hybridization with RNA oligonu-
as EST sequences or from previous publications. The GenBank IDs of arx, cleotides. Dev. Dyn. 235, 2538–2548.
ash2, bf-1, brn1/2/4, coe, dach, emx, er81, ets3, lhx1/5, svp, tll, vglut, Wnt5, Ekonomou, A., Kazanis, I., Malas, S., Wood, H., Alifragis, P., Denaxa, M.,
Wnt8, gad, and tyr hyd are GU169412–GU169428, respectively. Karagogeos, D., Constanti, A., Lovell-Badge, R., and Episkopou, V. (2005).
Neuronal migration and ventral subtype identity in the telencephalon depend
on SOX1. PLoS Biol. 3, e186.
SUPPLEMENTAL INFORMATION
Evans, S.M. (1966). Non-associative behavioural modifications in nereid poly-
Supplemental Information includes Extended Experimental Procedures, five chaetes. Nature 211, 945–948.
figures, and four movies and can be found with this article online at doi: Farris, S.M. (2005). Evolution of insect mushroom bodies: old clues, new
10.1016/j.cell.2010.07.043. insights. Arthropod Struct. Dev. 34, 211–234.
Farris, S.M. (2008). Evolutionary convergence of higher brain centers spanning
ACKNOWLEDGMENTS the protostome-deuterostome boundary. Brain Behav. Evol. 72, 106–122.
Fischer, A., and Dorresteijn, A.W.C. (2004). The polychaete Platynereis dumer-
We thank F. Christodoulou for providing larvae stained with mir-9, 9*, ilii (Annelida): a laboratory animal with spiralian cleavage, lifelong segment
H. Snyman for expert technical assistance, and the Arendt lab for support. proliferation and a mixed benthic/pelagic life cycle. Bioessays 26, 314–325.
The electron micrograph in Figure 5 was taken with C. Nielsen during a prac-
Förster, E., Zhao, S., and Frotscher, M. (2006). Laminating the hippocampus.
tical course. R.T. and K.T.-R. were supported by fellowships of the Marie Curie
Nat. Rev. Neurosci. 7, 259–267.
RTN ZOONET (MRTN-CT-2004-005624), R.T. by DFG Collaborative Research
Network SFB-488, and A.D. by a Louis Jeantet Foundation fellowship. R.T. Garel, S., Marı́n, F., Grosschedl, R., and Charnay, P. (1999). Ebf1 controls early
conceived and developed PrImR, designed and performed the experiments, cell differentiation in the embryonic striatum. Development 126, 5285–5294.
analyzed and interpreted the data, and wrote the paper. A.S.D. contributed Hébert, J.M., and Fishell, G. (2008). The genetics of early telencephalon
clones of vglut and gad. K.T.-R. contributed clones of brn124 and emx. D.A. patterning: some assembly required. Nat. Rev. Neurosci. 9, 678–685.
proposed to study annelid mushroom bodies, provided ideas, interpreted Heisenberg, M. (2003). Mushroom body memoir: from maps to models. Nat.
the data, and wrote the paper. Rev. Neurosci. 4, 266–275.
Houart, C., Caneparo, L., Heisenberg, C., Barth, K., Take-Uchi, M., and
Received: March 12, 2010
Wilson, S. (2002). Establishment of the telencephalon during gastrulation by
Revised: May 22, 2010
local antagonism of Wnt signaling. Neuron 35, 255–265.
Accepted: July 14, 2010
Published: September 2, 2010 Inoue, T., Nakamura, S., and Osumi, N. (2000). Fate mapping of the mouse
prosencephalic neural plate. Dev. Biol. 219, 373–383.

REFERENCES Jékely, G., and Arendt, D. (2007). Cellular resolution expression profiling using
confocal detection of NBT/BCIP precipitate by reflection microscopy. Bio-
Arendt, D. (2005). Genes and homology in nervous system evolution: techniques 42, 751–755.
comparing gene functions, expression patterns, and cell type molecular Kandel, E.R.S., James, H., and Jessell, T.M. (2000). Principles of Neural
fingerprints. Theory Biosci. 24, 185–197. Science, Fourth Edition (New York: McGraw-Hill Companies, Inc.).

808 Cell 142, 800–809, September 3, 2010 ª2010 Elsevier Inc.


Keeble, T.R., Halford, M.M., Seaman, C., Kee, N., Macheda, M., Anderson, Strausfeld, N.J., Hansen, L., Li, Y., Gomez, R.S., and Ito, K. (1998). Evolution,
R.B., Stacker, S.A., and Cooper, H.M. (2006). The Wnt receptor Ryk is required discovery, and interpretations of arthropod mushroom bodies. Learn. Mem. 5,
for Wnt5a-mediated axon guidance on the contralateral side of the corpus 11–37.
callosum. J. Neurosci. 26, 5840–5848.
Strausfeld, N.J., Strausfeld, C.M., Loesel, R., Rowell, D., and Stowe, S. (2006).
Kimura, J., Suda, Y., Kurokawa, D., Hossain, Z.M., Nakamura, M., Takahashi, Arthropod phylogeny: onychophoran brain organization suggests an archaic
M., Hara, A., and Aizawa, S. (2005). Emx2 and Pax6 function in cooperation relationship with a chelicerate stem lineage. Proc Biol Sci 273, 1857–1866.
with Otx2 and Otx1 to develop caudal forebrain primordium that includes
future archipallium. J. Neurosci. 25, 5097–5108. Strausfeld, N.J., Sinakevitch, I., Brown, S.M., and Farris, S.M. (2009). Ground
plan of the insect mushroom body: functional and evolutionary implications.
Liu, X., Long, F., Peng, H., Aerni, S.J., Jiang, M., Sánchez-Blanco, A., Murray,
J. Comp. Neurol. 513, 265–291.
J.I., Preston, E., Mericle, B., Batzoglou, S., et al. (2009). Analysis of cell fate
from single-cell gene expression profiles in C. elegans. Cell 139, 623–633. Sugino, K., Hempel, C.M., Miller, M.N., Hattox, A.M., Shapiro, P., Wu, C.,
Marı́n, O., and Rubenstein, J.L. (2001). A long, remarkable journey: tangential Huang, Z.J., and Nelson, S.B. (2006). Molecular taxonomy of major neuronal
migration in the telencephalon. Nat. Rev. Neurosci. 2, 780–790. classes in the adult mouse forebrain. Nat. Neurosci. 9, 99–107.
McEvilly, R.J., de Diaz, M.O., Schonemann, M.D., Hooshmand, F., and Swanson, L.W. (2000). Cerebral hemisphere regulation of motivated behavior.
Rosenfeld, M.G. (2002). Transcriptional regulation of cortical neuron migration Brain Res. 886, 113–164.
by POU domain factors. Science 295, 1528–1532.
Taglialatela, P., Soria, J.M., Caironi, V., Moiana, A., and Bertuzzi, S. (2004).
Miura, H., Yanazawa, M., Kato, K., and Kitamura, K. (1997). Expression of
Compromised generation of GABAergic interneurons in the brains of Vax1-/-
a novel aristaless related homeobox gene ‘Arx’ in the vertebrate telenceph-
mice. Development 131, 4239–4249.
alon, diencephalon and floor plate. Mech. Dev. 65, 99–109.
Müller, W.A. (1973). [Autoradiographic studies on the synthetic activity of Tessmar-Raible, K., Steinmetz, P.R.H., Snyman, H., Hassel, M., and Arendt, D.
neurosecretory cells in the brain of Platynereis dumerilii during sexual develop- (2005). Fluorescent two-color whole mount in situ hybridization in Platynereis
ment and regeneration]. Z. Zellforsch. Mikrosk. Anat. 139, 487–510. dumerilii (Polychaeta, Annelida), an emerging marine molecular model for
evolution and development. Biotechniques 39, 460, 462, 464.
Nieto, M., Schuurmans, C., Britz, O., and Guillemot, F. (2001). Neural bHLH
genes control the neuronal versus glial fate decision in cortical progenitors. Tessmar-Raible, K., Raible, F., Christodoulou, F., Guy, K., Rembold, M.,
Neuron 29, 401–413. Hausen, H., and Arendt, D. (2007). Conserved sensory-neurosecretory cell
Nieuwenhuys, R. (2002). Deuterostome brains: synopsis and commentary. types in annelid and fish forebrain: insights into hypothalamus evolution. Cell
Brain Res. Bull. 57, 257–270. 129, 1389–1400.
Noveen, A., Daniel, A., and Hartenstein, V. (2000). Early development of the Tole, S., Ragsdale, C.W., and Grove, E.A. (2000). Dorsoventral patterning of
Drosophila mushroom body: the roles of eyeless and dachshund. Develop- the telencephalon is disrupted in the mouse mutant extra-toes(J). Dev. Biol.
ment 127, 3475–3488. 217, 254–265.
Raible, F., Tessmar-Raible, K., Osoegawa, K., Wincker, P., Jubin, C., Tuoc, T.C., and Stoykova, A. (2008). Er81 is a downstream target of Pax6 in
Balavoine, G., Ferrier, D.E., Benes, V., de Jong, P., Weissenbach, J., et al. cortical progenitors. BMC Dev. Biol. 8, 23.
(2005). Vertebrate-type intron-rich genes in the marine annelid Platynereis
dumerilii. Science 310, 1325–1326. Urbach, R., and Technau, G.M. (2003). Molecular markers for identified neuro-
Schneider, S.Q., and Bowerman, B. (2007). beta-Catenin asymmetries after all blasts in the developing brain of Drosophila. Development 130, 3621–3637.
animal/vegetal- oriented cell divisions in Platynereis dumerilii embryos Viola, P.A., and Wells, W.M., III. (1995). Alignment by maximization of mutual
mediate binary cell-fate specification. Dev. Cell 13, 73–86. information. International Journal of Computer Vision 24, 137–154.
Shimamura, K., Hartigan, D.J., Martinez, S., Puelles, L., and Rubenstein, J.L.
Wang, B., Waclaw, R.R., Allen, Z.J., 2nd, Guillemot, F., and Campbell, K.
(1995). Longitudinal organization of the anterior neural plate and neural tube.
(2009). Ascl1 is a required downstream effector of Gsx gene function in the
Development 121, 3923–3933.
embryonic mouse telencephalon. Neural Dev 4, 5.
Stigloher, C., Ninkovic, J., Laplante, M., Geling, A., Tannhäuser, B., Topp, S.,
Kikuta, H., Becker, T.S., Houart, C., and Bally-Cuif, L. (2006). Segregation of Woodger, J.H. (1945). On biological transformations. In Essays on Growth and
telencephalic and eye-field identities inside the zebrafish forebrain territory is Form, presented to D’Arcy Wentworth Thompson, W.E. le Gros Clark and P.B.
controlled by Rx3. Development 133, 2925–2935. Medawar, eds. (Oxford: Oxford University Press), pp. 95–120.

Strausfeld, N.J., and Barth, F.G. (1993). Two visual systems in one brain: Yun, K., Potter, S., and Rubenstein, J.L. (2001). Gsh2 and Pax6 play comple-
neuropils serving the secondary eyes of the spider Cupiennius salei. mentary roles in dorsoventral patterning of the mammalian telencephalon.
J. Comp. Neurol. 328, 43–62. Development 128, 193–205.

Cell 142, 800–809, September 3, 2010 ª2010 Elsevier Inc. 809


Resource

The Protein Composition of Mitotic


Chromosomes Determined Using
Multiclassifier Combinatorial Proteomics
Shinya Ohta,1,6 Jimi-Carlo Bukowski-Wills,1,2,6 Luis Sanchez-Pulido,4 Flavia de Lima Alves,1 Laura Wood,1 Zhuo A. Chen,1
Melpi Platani,1 Lutz Fischer,1 Damien F. Hudson,1,5 Chris P. Ponting,4 Tatsuo Fukagawa,3 William C. Earnshaw,1,*
and Juri Rappsilber1,*
1Wellcome Trust Centre for Cell Biology
2Centre for Systems Biology at Edinburgh, School of Biological Sciences
University of Edinburgh, Edinburgh EH9 3JR, UK
3Department of Molecular Genetics, National Institute of Genetics and The Graduate University for Advanced Studies, Mishima,

Shizuoka 411-8540, Japan


4MRC Functional Genomics Unit, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3QX, UK
5Present address: Murdoch Children’s Research Institute, Royal Children’s Hospital, Melbourne, Victoria 3052, Australia
6These authors contributed equally to this work

*Correspondence: bill.earnshaw@ed.ac.uk (W.C.E.), juri.rappsilber@ed.ac.uk (J.R.)


DOI 10.1016/j.cell.2010.07.047

SUMMARY chores so that chromosomes can align and segregate on the


mitotic spindle. Our understanding of the mechanisms under-
Despite many decades of study, mitotic chromo- lying chromosome condensation is still fragmentary. These
some structure and composition remain poorly char- processes can be fully understood only when all components
acterized. Here, we have integrated quantitative pro- of mitotic chromosomes have been identified and functional
teomics with bioinformatic analysis to generate relationships between them determined. We have developed
a series of independent classifiers that describe the a new approach that we term multiclassifier combinatorial pro-
teomics (MCCP) to do this.
4,000 proteins identified in isolated mitotic chro-
The current list of mitotic chromatin proteins reported in pro-
mosomes. Integrating these classifiers by machine teomic studies is surprisingly short. Early analyses described
learning uncovers functional relationships between 62 and 79 proteins, respectively, in mitotic chromosome scaf-
protein complexes in the context of intact chromo- folds (Morrison et al., 2002; Gassmann et al., 2005). A later study
somes and reveals which of the 560 uncharacter- identified > 250 proteins that bound to sperm chromatin in
ized proteins identified here merits further study. Xenopus egg extracts in vitro, revealing the kinetochore protein
Indeed, of 34 GFP-tagged predicted chromosomal Bod1 (Porter et al., 2007). Other studies identified 240 proteins,
proteins, 30 were chromosomal, including 13 with subsequently corrected to roughly 50 bona fide putative struc-
centromere-association. Of 16 GFP-tagged pre- tural proteins (Uchiyama et al., 2005; Takata et al., 2007). In a tar-
dicted nonchromosomal proteins, 14 were confirmed geted study, 98 proteins were identified as shared in isolated
to be nonchromosomal. An unbiased analysis of the telomeres from wild-type and ALT cells (Dejardin and Kingston,
2009). Despite these efforts, currently available proteomics
whole chromosome proteome from genetic knock-
reports miss a significant fraction of known mitotic chromosomal
outs of kinetochore protein Ska3/Rama1 revealed proteins, particularly kinetochore components.
that the APC/C and RanBP2/RanGAP1 complexes Biochemical analysis of important chromosome substruc-
depend on the Ska complex for stable association tures such as kinetochores is extremely challenging. The
with chromosomes. Our integrated analysis predicts kinetochore is one of the most complex cellular substructures
that up to 97 new centromere-associated proteins (Cheeseman and Desai, 2008), with over 120 constituents
remain to be discovered in our data set. described by a range of approaches (Earnshaw and Rothfield,
1985), including targeted proteomic studies (Obuse et al.,
2004; Foltz et al., 2006; Okada et al., 2006; Hori et al., 2008;
INTRODUCTION Amano et al., 2009). Biochemical dissection of kinetochores is
complicated by the fact that it is not known to what extent
As cells enter mitosis, chromosomes undergo a remarkable the constituent protein complexes can be recovered in soluble
series of physiological and structural transformations known as form from chromosomes with their relevant intermolecular
chromosome condensation. This process involves individualiza- associations intact. As described here, those problems can
tion of the chromosomal territories to create the characteristic be circumvented by purifying and analyzing whole mitotic
mitotic chromosome morphology and maturation of the kineto- chromosomes.

810 Cell 142, 810–821, September 3, 2010 ª2010 Elsevier Inc.


Purifying large cellular structures or organelles free of con- A Classifier Approach to Identify Genuine Mitotic
taminants is virtually impossible. Genuine components have Chromosome Proteins
been distinguished from contaminants in such preparations by The presence of hitchhiker proteins complicates the definition
subtractive (Schirmer et al., 2003) or quantitative (Foster et al., of what constitutes a ‘‘true’’ chromosomal protein, as well as
2003) proteomics by determining the difference between two the design of biochemical control experiments. For example,
near-identical fractions, one enriched and the other depleted of comparing mitotic to interphase chromatin is of limited use,
the target structure. In protein correlation profiling, a set of known since the latter is shielded from the cytoplasm by the nuclear
components was used to define a common intensity profile envelope. In such a comparison, cytoplasmic hitchhiker proteins
across neighboring biochemical fractions from sucrose gradients would be scored as mitosis-specific chromosomal proteins.
during purification of organelles and this was used to select other Here, we describe an approach to study the complex chromo-
proteins that show a similar profile (Andersen et al., 2003). These somal proteome that both identifies proteins that merit further
methods do not recognize cellular background proteins that study and reveals functional relationships between all chromo-
adhere to the structure of interest due to nonspecific hydro- somal proteins. We quantify the chromosomal association of
phobic or electrostatic interactions. This type of contamination each protein in a series of quantitative proteomics experiments,
is particularly relevant for vertebrate mitotic chromosomes. mostly using SILAC technology (Ong et al., 2002). Each experi-
In the present study, we identified approximately 4000 polypep- ment provides an independent measure of a protein’s associa-
tides in highly purified chromosomes. We developed a statistical tion with mitotic chromosomes, which we term a ‘‘classifier.’’
approach for analysis of proteomic data to confirm which known Integration of the data obtained with all classifiers enables us
and uncharacterized proteins from this long list are chromosomal. to detect patterns in the behavior of groups of proteins that
An experimental test of our method led to the identification of 32 reveal shared membership in protein complexes as well as func-
chromosomal proteins, including 13 kinetochore-associated tional dependency relationships.
proteins. Key to our analysis is the innovative use of stable isotope The experimental protocols that define the five proteomic
labeling with amino acids in cell culture (SILAC) (Ong et al., 2002), classifiers are shown in Figure 1E and all classifiers are summa-
plus development of a framework to integrate data from multiple rized below.
classifiers, including nonproteomic classifiers, to reveal proteins Classifier I: Abundance estimation
of interest and determine functional relationships between protein To estimate the amounts of individual proteins in mitotic chromo-
complexes in the context of whole chromosomes. somes, we used an established protocol (Rappsilber et al., 2002;
Ishihama et al., 2005, 2008) to calculate a scaled protein abun-
dance index based on the number of peptides observed and
RESULTS the number of times that each peptide is observed (spectral
count) for each protein. This calculation and its validation are dis-
Identification of the Proteome of Isolated cussed in Extended Experimental Procedures.
Mitotic Chromosomes In the conventional pie chart of Figure 1D, all proteins are
We isolated mitotic chromosomes from chicken DT40 cells by weighted equally, independent of their actual abundance in iso-
a refinement of the polyamine method (Lewis and Laemmli, lated chromosomes. A more informative representation of chro-
1982) (Figures 1A and 1B). These preparations are negative for mosome composition is obtained by normalizing each class by
porin in immunoblots (Figure 1C), indicating that mitochondrial its mass, obtained by multiplying the estimated abundance by
contamination (common in chromosome preparations) is low. the predicted molecular mass of each protein (Figure 1F). As
Proteomic analysis (Cox and Mann, 2008; de Godoy et al., expected, histones comprise the bulk of mitotic chromosomal
2008) of 250 mg of total chromosomal protein identified 4029 protein (48%). Overall 68% of the protein mass is annotated as
proteins in 28 functional categories (Figure 1D and Table S1 chromosomal.
available online), including essentially all previously described Classifier II: Enrichment in Chromosomes
chromosomal proteins. We expected core chromosomal components like histones or
When vertebrate cells enter mitosis, the nuclear envelope structural proteins would be more abundant in isolated chromo-
breaks down and chromosomes are newly exposed to cyto- somes than in cytoplasmic extracts. The reverse would be true of
plasmic proteins, organelles and cellular membranes. Since background proteins. We therefore mixed isolated chromo-
highly positively charged histones contribute 38% of the chro- somes from mitotic DT40 cells grown in light medium with an
mosome mass and an equivalent amount is highly negatively equal mass of protein from post-chromosomal extracts of
charged DNA, many charged nonchromosomal proteins exhibit parallel cultures grown with heavy SILAC medium (Figure 1E).
strong adventitious binding to chromosomes. These ‘‘hitch- Classifier II was calculated as the ratio of light/heavy peaks for
hikers’’ differ from conventional contaminants (e.g., mitochon- each protein. Among the 20% most enriched proteins, chromo-
dria), as they are physically associated with the chromosomes somal proteins outnumbered nonchromosomal proteins by 3 to
before cell lysis and apparently cannot be separated by conven- 1. Conversely, among the 20% least enriched proteins, back-
tional purification protocols. Thus, the 1331 cytoskeletal, cyto- ground proteins outnumbered chromosomal proteins 6 to 1.
plasmic, mitochondrial, membrane and receptor proteins found Classifier III: In Vitro Exchange on Chromosomes
in our preparations, may be physically associated with chromo- We ranked proteins based on their ability to stably bind to chro-
somes following nuclear envelope disassembly, but many not be mosomes during an incubation in cytosol. A crude light chromo-
functionally relevant. some fraction obtained by gentle centrifugation was mixed with

Cell 142, 810–821, September 3, 2010 ª2010 Elsevier Inc. 811


Figure 1. Proteomic Analysis of Mitotic
Chromosome Proteins
(A) Outline of chromosome isolation procedure.
(B) Silver-stained gel of isolated chromosomes.
(C) Immunoblot with markers for mitochondria
(porin) and chromosomes.
(D) The 28 classes of proteins found in chromo-
somes.
(E) Experimental designs to prepare samples
for determination of classifiers I (abundance), II
(enrichment in chromosomes versus post-chro-
mosomal extract), III (exchange onto chromo-
somes from post-chromosomal cytosol), IV
(dependency on SMC2/condensin) and V (depen-
dency on Ska3/RAMA1).
(F–J) (F) Estimated percentages of total chromo-
somal protein mass in the major classes of
proteins. Sample spectra used to calculate classi-
fier III (exchange) values for (G) Ribosomal protein
SA (a hitchhiker protein), (H) MAD2, (I) BUBR1
and (J) MAD1. Spectra are plotted as the peak
intensity (a measure of abundance) against the
mass divided by charge. Peaks of light peptides
from chromosomes are indicated by blue bars
and heavy peptides bound from cell extracts
during the incubation are indicated by red bars.
See also Figure S1.

III = 61) and condensin I (average classi-


fier III = 59). Interestingly, condensin II
was more exchangeable (average classi-
fier III = 14). In contrast, the ratios for
75% of ribosomal proteins were in the
range from 0.45 to 3.0 (Figure 1G), indi-
cating significant binding to chromo-
somes from cytosol during the incubation.
Although we sought to optimize purity
rather than preserve functionality of
chromosomes, the exchange experiment
revealed that at least one aspect of kinet-
ochore function was retained in purified
chromosomes. Similar to one recent
study (Kulukian et al., 2009), kinetochores
of the purified chromosomes can recruit
spindle checkpoint proteins Mad2, Bub3,
and BubR1 from cytosol (Figures 1H and
1I) but not Mad1 (Figure 1J).
Classifier IV: SMC2 Dependency
We used a conditional genetic knockout
of SMC2 in DT40 SMC2ON/OFF cells (Hud-
son et al., 2003) to compare the composi-
an excess of heavy post-chromosomal extract and incubated tion of mitotic chromosomes formed in the presence or absence
to allow proteins to exchange at 14 C for 30 min (Figure 1E). of condensin, which is required for structural integrity of mitotic
The chromosomes were then subjected to rigorous purification. chromosomes. DT40 SMC2ON cells were cultured in SILACheavy
All heavy proteins identified must have bound to the chromo- medium. To obtain chromosomes depleted of condensin, cells
somes during the incubation in vitro. Classifier III is the light/ grown in SILAClight medium were cultured with doxycycline for
heavy ratio for each protein identified in this experiment. 30 hr to shut down SMC2 expression prior to the nocodazole
The most stable chromosomal-associated proteins were block (SMC2OFF). Equal numbers of mitotic cells from the two
histones (average classifier III = 63), topoisomerase IIa (classifier different populations were mixed and mitotic chromosomes

812 Cell 142, 810–821, September 3, 2010 ª2010 Elsevier Inc.


Figure 2. Combining Classifiers Increases
Specificity
(A) The rank of all proteins observed for proteomic
classifiers I (abundance), II (enrichment), III (reten-
tion), IV (SMC2 dependency), V (Ska3/Rama1
dependency) and RF (combined random forest
ranking) is plotted as vertical color coded lines
(rug plots). Proteins are ranked from the highest
(left) to lowest values for each classifier. Centro-
mere proteins are indicated by longer bars
(indicated).
(B) Cumulative curves show that combining classi-
fiers by RF significantly increases the specificity
of identifying chromosomal proteins without com-
promising the completeness of the analysis.
(C) 2D scatter graph plotting classifiers I versus II
flanked by the relevant rug plots. Proteins most
enriched in chromosomes relative to cytoplasm
and proteins most abundant in chromosomes are
above and to the right, respectively. Each spot is
color-coded by category (Figure 1D).
(D) Zoom of the region indicated in (C) showing
members of the chromosomal passenger (CPC),
Mis12, Ndc80, Nup107-160 and Cohesin com-
plexes plus the constitutive centromere-associ-
ated network (CCAN) and anaphase-promoting
complex/cyclosome (APC/C).
(E) Enrichment for chromosomal proteins in the
region indicated in (C & D).
See also Figure S2.

isolated (Figure 1E). Classifier IV is the heavy/light ratio Classifier VI: Domain Analysis
(SMC2ON/SMC2OFF) for each of the proteins identified in this We added an additional nonproteomic classifier to our analysis
experiment. using the protein domains found in chromosomal and nonchro-
SMC2-depleted chromosomes contained 5.8% of the wild- mosomal proteins (red/pink and green wedges in Figure 1D).
type level of SMC2. It is not possible to isolate chromosomes This made use of bioinformatic analysis in order to segregate
from cultures completely lacking SMC2 as SMC2 is an essential chromosomal from nonchromosomal proteins, but importantly
gene and dead cells do not accumulate in mitosis. These chro- did not consider a protein’s relevance to mitosis. We counted
mosomes were similarly depleted of all condensin I and II how often each domain was observed in chromosomal and
subunits. nonchromosomal proteins and assigned it this frequency as
Classifier V: Ska3/Rama1 Dependency a score (Table S2). Classifier VI was then determined for each
To demonstrate the targeted analysis possible with our protein based on the sum of its domain scores.
approach, we compared the association of kinetochore proteins
with chromosomes in cells with or without Ska3/Rama1/ Multiclassifier Combinatorial Proteomics
C13orf3. Ska3/Rama1, which was identified in this analysis as Traditional one-dimensional analysis (e.g., sorting the various
a chromosomal protein, was described in several recent publica- proteins according to their value for each classifier) was of
tions (Daum et al., 2009; Gaitanos et al., 2009; Raaijmakers et al., limited utility, as the data lacked a clear boundary between chro-
2009; Theis et al., 2009; Welburn et al., 2009). mosomal (red/pink in Figure 2A) and nonchromosomal proteins
To obtain chromosomes depleted of Ska complex, we isolated (green in Figure 2A) for each classifier (Figure 2A and Figures
a genetic knockout of the Ska3/Rama1 gene (Figures S5A and S2A–S2E).
S5B). Ska3/Rama1 / cells (homozygous knockouts are viable) By contrast, when classifiers were combined, our ability
were grown in SILAClight medium (Figure 1E). Equal numbers of to identify chromosomal proteins was vastly improved. For
Ska3/Rama1 / and wild-type DT40 (cultured in SILACheavy example, an enrichment of centromeric or chromosomal pro-
medium) mitotic cells from the two different populations were teins relative to nonchromosomal proteins was obtained when
mixed and mitotic chromosomes isolated. Classifier V is the classifiers I (abundance) and II (enrichment) were plotted
heavy/light ratio (wild-type/Ska3/Rama1 / ) for each protein (Figures 2C–2E). The clustering of protein complex subunits
identified in this experiment. in this plot (Figure 2D) reflects both their relative stoichiometry

Cell 142, 810–821, September 3, 2010 ª2010 Elsevier Inc. 813


(x axis), and the similar degree to which subunits in a complex are proteins lacking known domains are excluded from this boost,
all present either on or off chromosomes (y axis). Members of the Figure S3D). Now, 92.4% of the centromere proteins were
APC/C, Ndc80 and Mis12 complexes form closely knit clusters. assigned as chromosomal. In summary, RF analysis provides
It is important to note that this was achieved in the context of us with a tool for productively combining the outcomes of our
entire chromosomes and without requiring solubilization of the individual proteomics classifier experiments and further empow-
complexes. ering our analysis by including data from other sources.
We used random forest (RF) analysis, a machine learning If results of a random forest based on the five proteomic clas-
approach, to progress beyond two-dimensional analyses and sifiers plus the bioinformatics-based classifier VI were plotted
integrate the information present in all proteomics classifiers. against those from the initial random forest analysis (Fig-
This analysis offered two powerful benefits. First, it enabled us ure 3A), a near-perfect separation of the training data was
to work with data sets that contain missing values. This is a signif- achieved. Only a single chromosomal protein of the training set
icant advantage in proteomics studies where not every protein is and two nonchromosomal proteins were misassigned when
observed in every experiment, as seen in Figure 2A and Fig- placing manually a separation line.
ure S2F. Second, RF analysis allowed us to use any descriptor Using ten-fold cross-validation, we found that 118 centromere
of our proteins as a classifier and integrate it into our overall proteins positioned right and only 7 left of the separation line
analysis. Here, we also included a bioinformatic analysis of the (Figure 3B). This compares to 14 and 9 centromere proteins
distribution of protein domains in our analysis distinguishing being missed when using the one-dimensional ranked lists by
chromosomal from nonchromosomal proteins (classifier VI). classifiers I-V and I-VI, respectively. Accepting the line as a
In brief, RF is a decision tree analysis that separates data sets threshold returns known centromere proteins with a yield of
into ‘‘true’’ and ‘‘false’’ groups. The decision trees are trained on 94.4% and all other chromosomal proteins with 93.1% success.
defined data sets and randomly built to optimize the separation In contrast, 83.1% of nonchromosomal proteins are rejected.
between them. Analysis of the experimental data set then occurs Thus, the classifier approach is sufficiently powerful to suggest
by running each protein through all trees and adding up its overall chromosomal proteins from among hitherto uncharacterized
RF score (i.e., the fraction of trees that scored it as ‘‘true’’). RFs proteins.
perform much better on training data than application data, so
their performance is evaluated by ten-fold cross-validation. Identification of New Chromosomal Proteins
The training data are split into random sections of 90% for To test the predictive power of our RF analysis, we cloned and
training and 10% for evaluation, so that successively the entire tested the location in mitosis of 50 previously uncharacterized
set is used for evaluation. Here, the two training data sets chosen proteins including 15 without known domains: 34 predicted to
were ‘‘nonchromosomal’’ (green wedges in Figure 1D) and be and 16 predicted not to be on chromosomes in mitosis.
‘‘chromosomal’’ (red + pink wedges in Figure 1D), and the RF Reasoning that important proteins would be conserved, we
score for a given protein is the fraction of trees that scored it expressed GFP-tagged human homologs of these chicken
as ‘‘chromosomal.’’ proteins in U2OS cells. Remarkably, 30 of 34 cloned proteins
RF analysis readily discriminated chromosomal from nonchro- from the chromosomal region were confirmed as chromosomal,
mosomal proteins. In the RF rug plot of Figure 2A, which repre- contrasting with only 2 of the 16 predicted ab initio to be
sents the ranked list of proteins generated by RF analysis, the nonchromosomal. This confirms the power of our analysis and
left side is predominantly red, while the right side is predomi- indicates a success rate of 88%, with 44 of 50 tagged proteins
nantly green. To reach the 500th chromosomal protein on the localizing as predicted. Of 50 newly cloned proteins, 13 were
RF-ranked list only 229 nonchromosomal proteins are included associated with kinetochores in mitosis, 12 had a more general
(Figure 2B). In contrast, 410-671 nonchromosomal proteins distribution on mitotic chromosomes and 7 others were peri-
would be included when considering ranked lists from individual chromosomal, a class whose new members we propose to
classifiers. Note that the RF-based sorting was done on the term chromosome periphery proteins (cPERPs A-G) (Figures
complete data set, including proteins that failed to be observed S4B–S4G and Table S3). The chromosome periphery (perichro-
with some classifiers. Therefore, adding information from addi- mosomal layer) is enriched in ribosomal and nucleolar hitchhiker
tional experiments did not decrease the number of proteins proteins, and is of unknown function (Van Hooser et al., 2005).
covered. The new centromere proteins all appeared to localize to the
The advantage of combining classifiers can be statistically outer kinetochore, relative to CENP-C and HEC1 as standards
expressed by ROC curves (Figure S3A), with increased area (Figures 4B–4J, Figure S4A). In keeping with established nomen-
under the curve (AUC) for our combined analysis when com- clature, we propose to name these proteins CENP-Y, CENP-Z
pared to each of the individual classifiers (AUCRF(cI-V) = 0.81, and CENP-27 through CENP-33. Beyond ‘Z’, the 26th letter of
AUCscI-V = 0.41-0.76). The combined classifiers assigned 88.8% the basic modern Latin alphabet, we propose to designate the
of our gold standard, the 125 centromere proteins, correctly as new proteins with numbers starting with CENP-27.
chromosomal, at the cut-off that minimizes misassignment of
chromosomal versus nonchromosomal proteins. This specificity Functional Analysis of New Kinetochore-Associated
was further improved when bioinformatic domain analysis (cVI) Proteins
was integrated with our proteomic classifiers (AUCRF(cI-VI) = We focused our initial functional analysis on kinetochore
0.97; identification of the first 500 chromosomal proteins yields proteins. Clustering analysis (Gentleman et al., 2004) allowed
17 nonchromosomal proteins, Suppl. Figures S3B and S3C, us to combine data for proteins identified by all classifiers, and

814 Cell 142, 810–821, September 3, 2010 ª2010 Elsevier Inc.


complexes (Figure 4). Interestingly, our clustering sorted
CENP-27 as a component of the APC/C. This was confirmed
and the protein named APC16 in three recent reports (Hutchins
et al., 2010; Kops et al., 2010; Hubner et al., 2010). To further
test the predictive value of cluster analysis for proteomic data
sets, we examined two kinetochore proteins in greater detail.
Ska3/Rama1 and Functional Analysis of Kinetochore
Subcomplexes
C13orf3 was located adjacent to Ska2 in the cluster analysis.
This protein now known as Ska3/Rama1 has been suggested
to be involved in microtubule attachment to kinetochores (Gaita-
nos et al., 2009; Raaijmakers et al., 2009; Welburn et al., 2009) or
coordination of the spindle checkpoint response (Daum et al.,
2009; Theis et al., 2009). We analyzed the kinetochore proteome
in the presence or absence of Ska3/Rama1 (defined as classifier
V) in order to determine the role of this protein in kinetochore
structure (Figure 5).
A map of the Ska3/Rama1 locus in DT40 is shown in Figures
S5A and S5B, together with a targeting strategy for inactivating
the gene. The Ska3/Rama1 gene is not essential for life in DT40
cells (Figure S5C). However, these cells struggle to achieve
a normal chromosome alignment, and show a 33 increase in
mitotic index (Figure S5D and S5E), a 33 increase in the
percentage of apoptotic cells and a 6x increase in the number
of bi-nucleated cells.
Proteomic analysis of isolated chromosomes revealed that
loss of Ska3/Rama1 was accompanied by the loss of Ska1 and
Ska2. Loss of the Ska complex caused no systematic changes
in the chromosomal association of proteins of the constitutive
centromere-associated network (CCAN), Knl-1/Mis12/Ndc80
(KMN), Mis18, Ndc80, CPC, and Nup107-160 kinetochore sub-
complexes. However, striking changes were seen in the levels
of the APC/C, RanBP2/RanGAP1, spindle checkpoint, Rod/
Zw10/Zwilch (RZZ), and dynein/dynactin complexes. We con-
firmed that the RanBP2/RanGAP1 complex is indeed depleted
from kinetochores when Ska3/Rama1 is deleted in HeLa cells
(Figure S5F and S5G). Attempts to confirm the specific kineto-
chore depletion of the APC/C were uninformative, as we were
unable to reproducibly obtain kinetochore staining for the
APC/C in HeLa cells using four independent antibodies.
We conclude that combining genetic and SILAC analysis
provides a powerful new method for analysis of multicomplex
protein superstructures.
Figure 3. Random Forest Analysis Predicts New Proteins of Interest
(A) Separation of the two training sets (color-coded groups from Figure 1D) by A Protein Involved in Chromosome Alignment
RF analysis. The line provides optimal separation of the two training sets. and Spindle Organization
x axis: RF analysis of classifiers I–V; y axis: RF analysis of classifiers I–VI, A second new kinetochore-associated protein, CENP-32/
including the bioinformatic domain analysis. C9orf114, was sorted on our kinetochore cluster diagram next
(B) Positions of previously known centromere proteins from ten-fold cross- to CLASP1 and CLASP2, two paralogues known to be involved
validation.
in the regulation of microtubule dynamics. Like CLASPs,
(C) Position of cloned proteins and remaining uncloned proteins in the same 2d
analysis.
CENP-32/C9orf114 mapped to the outer kinetochore (Figure 6A),
(D) Classification of newly cloned proteins from regions to the left and right of and its depletion caused a significant accumulation of cells in
the dividing line, respectively. later prometaphase (Figure S6A) with misaligned chromosomes
See also Figure S3. (Figure 6B). These cells frequently had bipolar spindles, however
60% of those spindles exhibited remarkable abnormalities
where centrosomes appeared to have detached from the poles
look for informative groupings. This revealed a striking tendency (Figures 6C and 6D and Figure S6B). In one remarkable case,
for functionally related proteins to form clusters, as exemplified the centrosomes appeared at the midzone of a bipolar spindle
by members of the NDC80, CPC, Nup107-160 and APC/C (Figure 6C9-12).

Cell 142, 810–821, September 3, 2010 ª2010 Elsevier Inc. 815


Figure 4. Cluster Analysis and Imaging of New Kinetochore Proteins
(A) Heat map and cluster dendrogram for 101 centromere-associated proteins identified by all 5 proteomic classifiers. Known complexes are color-marked (see
bottom) and show a tendency to cluster in this analysis. Kinetochore proteins identified in this study are also marked (stars).
(B–J) Localization of GFP-tagged kinetochore-associated proteins (panels 2) relative to DNA detected with DAPI (panels 1) and CENP-C (panels 3 in [B]–[D], [F]–
[H], and [J] or ACA (panels 3 in [E] and [I]). Insets show merged images with blow-ups of representative kinetochores (GFP-novelCENP, green and CENP-C or
ACA, red).
See also Figure S4.

DISCUSSION complex proteome. The key approach of combining classifier


data can in principle be expanded indefinitely and can include
Multiclassifier Combinatorial Proteomics nonproteomics data sets such as the bioinformatic protein
The approach described here for analysis of the proteome of domain analysis used here. Other classifiers that could
vertebrate mitotic chromosomes can be used to study any be used in the future include microarray, protein interaction

816 Cell 142, 810–821, September 3, 2010 ª2010 Elsevier Inc.


(e.g., two hybrid screens or pull-down), protein phosphoryla-
tion, and localization data and, indeed, data from any experi-
mental approach in which the proteins of interest are sorted
systematically.
We first showed that plotting pairs of classifiers against one
another improved our ability to delineate potential chromosomal
proteins. As that approach could not be generalized when the
number of classifiers exceeded three, we adopted a random
forest (RF) analysis approach. This allowed us to integrate infor-
mation from all classifiers into decision trees on which known
and unknown proteins could be classified. Importantly, RF anal-
ysis handles missing values systematically. This is crucial when
not every protein is observed in every experiment. In contrast,
cluster analysis, which has been used both in this study and in
other recent work (Theis et al., 2009; Neumann et al., 2010),
can only integrate data for proteins that have a value for every
classifier.

Integrity of the Isolated Chromosomes


Our methods focused on optimizing the purity of the chromo-
somes. Thus, our list of proteins is likely to represent the minimal,
stably associated components of mitotic chromosomes. None-
theless kinetochores of isolated chromosomes retain some
function, as judged by their ability to recruit components of
the mitotic checkpoint complex from cytoplasm. This may be
because chromosomes were isolated from nocodazole-treated
cells, with kinetochores actively engaged in spindle checkpoint
signaling.

New Insights into Kinetochore Functional Organization


Remarkably, although no biochemical enrichment for centro-
meres was performed, our data set contained all known centro-
meric subcomplexes, with peptides from 125 reported cen-
tromere proteins (eight present as multiple isoforms) (Table S1).
We identified all members of the CCAN, KMN, Mis12 and Mis18
complexes and all members of the RZZ complex except Zwint
(which is not yet annotated in the chicken genome). Our success
in identifying centromere and telomere proteins may be
explained because 66 of the 78 chicken DT40 chromosomes
are microchromosomes whose purification provides a natural
enrichment for centromeres and telomeres since the chromo-
some arms are so short.
We combined genetics with whole proteome analysis in order
to identify complexes and structural dependencies in their
‘‘native environment’’ (e.g., kinetochore proteins in actual kinet-
ochores). Chromosomes lacking Ska3/Rama1 were depleted for
the entire Ska complex, confirming that these three proteins are
interdependent for chromosome binding. Similarly, depletion of
key condensin subunit SMC2 caused a loss of all seven subunits
of the condensin I and II complexes from chromosomes. Impor-
Figure 5. Ska3/Rama1 Dependency for Chromosomal Association
tantly, this analysis did not require tagging of any proteins or
and Analysis of CENP-27/APC16
Kinetochore proteins with increased abundance on chromosomes in the
attempts to solubilize functional complexes from large subcel-
absence of Ska3/Rama1 are shown with blue bars and proteins with lular structures.
decreased abundance are shown with red bars. Note covariance of protein In addition to these primary effects of depletion, loss of Ska3/
complexes. See also Figure S5. Rama1 also caused a significant secondary depletion of the
APC/C and RanBP2/RanGAP1 complexes from chromosomes
but had no consistent effect on most other kinetochore proteins.
Importantly, all members of the secondary-depleted complexes

Cell 142, 810–821, September 3, 2010 ª2010 Elsevier Inc. 817


Figure 6. Initial Characterization of CENP-32
(A) Localization of GFP-CENP-32 (panel 2, green) in the kinetochore relative to DNA (panel 1, blue) and CENP-C (panel 3, red).
(B) CENP-32 RNAi causes problems with chromosome alignment. DNA, red; CENP-C, green; microtubules, white.
(C) CENP-32 RNAi causes detachment of centrosomes from bipolar spindles. DNA (panels 1,5,9,13), blue; pericentrin (panels 2,6) or g-tubulin (panels 10,14),
green; a-tubulin (panels 3,7,11,15), red.
(D) Quantitation of spindle phenotypes following CENP-32 RNAi.
(E) Schematic representation of domain architectures for new kinetochore proteins tagged in this study (drawn to approx. scale). The locations of domains are
according to Pfam and SMART family databases (Letunic et al., 2004; Finn et al., 2010), complemented by REP web server analysis (Andrade et al., 2000).
The localization of the PEHE domain has been assigned from (Marı́n, 2003). Phyletic distributions of proteins are indicated in blue, yellow, green and violet
for Metazoa, Fungi, Plantae and Archaea, respectively. CENP-35 has been truncated. Abbreviations: BCNT, Bucentaur or craniofacial development protein;
Fbox, cyclin-F motif; G-Patch, Glycine-rich nucleic binding domain; JmjC, jumonji C domain; LRR, Leucine-rich repeats; Nua4, Nucleosomal acetyltransferase
of H4; OB, Oligonucleotide/oligosaccharide Binding; PEHE, Pro-Glu-His-Glu conserved region; PHD, Plant HomeoDomain; RA, Ras associated (RalGDS/AF-6)
domain; SPOUT, spoU and trmD RNA methyltransferase; WD40, WD/beta-transducin repeats; Zf-AD, Zinc finger-associated domain; and Zf-C2H2, Classical
zinc finger Cys(2)His(2).
See also Figure S6.

818 Cell 142, 810–821, September 3, 2010 ª2010 Elsevier Inc.


also behaved coordinately. Our results (e.g., the behavior of as specific for mitosis. Furthermore, their association might
CENP-27/APC16 as a component of the APC/C) confirm the depend on a fully assembled kinetochore and thus be lost
utility of single protein depletion analysis for the identification when attempting other than whole chromosome analysis.
of protein complexes and determination of their mutual interde-
pendencies for association with chromosomes. Characterization of a Kinetochore Protein
Our data suggest that the Ska complex may provide a docking CENP-32 is required both for chromosome alignment and for
site for the APC/C in the outer kinetochore. Alternatively, sumoy- association of the centrosomes with the poles of the bipolar
lation by RanBP2 may have a role in APC/C binding to chromo- spindle during metaphase. This latter phenotype is very similar
somes. The RanBP2-RanGAP1 complex is known to be involved to an unusual spindle morphology phenotype seen in Drosophila
in kinetochore-microtubule interactions and localization of cells following depletion of the CLASP homolog Mast/Orbit
several spindle checkpoint proteins (Joseph et al., 2004). We (Maiato et al., 2002). Indeed, in our analysis, CENP-32 clusters
note that among the recent spate of publications on Ska3/ with CLASP1 and CLASP2. A yeast homolog of CENP-32 inter-
Rama1, our observations appear to support a role in integration acts with CBF5, an enzyme involved in the posttranscriptional
and regulation of the spindle checkpoint response (Daum et al., modification of rRNA, that was shown to bind to budding yeast
2009; Theis et al., 2009). centromeres and microtubules (Jiang et al., 1993). Bioinformatics
Our whole-proteome analysis revealed that Cdc20 behaved analysis suggests that CENP-32 is a member of the SPOUT family
like a member of the APC/C and was distinct from other compo- of methyltransferases but is atypical in possessing a possible
nents of the spindle checkpoint pathway with respect to its Ska RNA-binding OB fold inserted into its catalytic domain (Tkaczuk
complex dependency. Spindle checkpoint components asso- et al., 2007). It is tempting to speculate that CENP-32 may func-
ciate with one another in cytoplasm as a mitotic checkpoint tion at kinetochores by interacting with an as-yet unknown RNA.
complex (MCC), containing BubR1, Bub3, Cdc20 and Mad2.
Our data suggest that once the MCC associates with chromo-
How Complex Is the Kinetochore?
somes, Cdc20 stably associates with the APC/C.
MCCP analysis allows us to predict how many more kinetochore
proteins remain to be identified in our data set. In the plot of
What Classes of Kinetochore Proteins Remain
Figure 3, where the chromosomal proteome is displayed in two
to Be Discovered?
dimensions, we found 35% of novel tagged proteins from region
We identified 13 kinetochore-associated proteins among previ-
R and 6% from region L to associate with the kinetochore during
ously uncharacterized proteins and, as discussed below, we
mitosis. These regions have 224 and 287 as-yet uncharacterized
predict that many more remain to be described. We therefore
novel proteins, respectively. Assuming no bias in the proteins we
asked whether there is any functional relationship between these
cloned, this suggests that approximately 97 more kinetochore
new proteins. That is, what sorts of kinetochore proteins had
proteins remain to be discovered. Taking into account the 13
been missed in the many previous genetic and biochemical
kinetochore-associated proteins confirmed in our work, this
screens? An interesting answer has emerged.
roughly doubles the currently known protein complexity of the
Since the new kinetochore proteins were identified solely
kinetochore during mitosis, confirming it as one of the most
based on their occurrence in chromosomes, they could poten-
complex cellular substructures.
tially represent a wide range of functions. Nevertheless, it is
striking that five of the new centromere proteins (namely,
Conclusions
CENP-28, 29, 31, 35, and 36) are subunits of complexes
Multiclassifier combinatorial proteomics and the data sets
that modify and/or bind histones (Figure 6E). Yeast orthologs
described here open the door to the identification of all functional
of two of these proteins (namely, CENP-28/C1orf149 and
components of mitotic chromosomes despite the adventitious
CENP-29/CFDP1) contribute to NuA4 histone acetyltransferase
binding of cellular background proteins during mitosis. Further-
(HAT) and SWR1 ATP-dependent chromatin remodelling com-
more, MCCP can be extended by adding additional classifiers
plexes, respectively. These complexes are known to share com-
to delineate protein complexes and define functional dependen-
ponents (Wu et al., 2005) and together stimulate the exchange of
cies between them in the context of intact mitotic chromosomes.
histone H2A for H2A.Z, following acetylation of H2A or H4 (Altaf
This will serve both as a starting point for systematic determina-
et al., 2010). A third centromere-associated protein, CENP-36/
tion of the full range of functions involved in mitotic chromosome
MSL1v1, is necessary for the activity of MOF, another HAT, on
segregation, and as a basis for the development of detailed
nucleosomal H4 (Li et al., 2009). Finally, CENP-31/PHD6 and
structural and functional interaction maps of key chromosomal
CENP-35/PHF2 each contain PHD (plant homeodomain) zinc
subdomains. MCCP should also prove useful for the analysis
fingers, which are usually associated with chromatin-mediated
of other cellular structures that lack defined boundaries, e.g.,
transcriptional regulation. The PHD of CENP-35, which also
membrane associated complexes like the post-synaptic density.
contains a JmjC (likely histone demethylase) domain, appears
to be required for demethylation of H3 at the promoters of ribo-
somal RNA genes (Wen et al., 2010). EXPERIMENTAL PROCEDURES

Why were these proteins not discovered earlier as kineto-


Preparation of Mitotic Chromosomes
chore-associated? One likely explanation is that they may have DT40 cells were incubated with Nocodazole for 13 hr, resulting in a mitotic
essential functions in other chromatin regions as well. Thus, index of 70%–90%. Mitotic chromosomes were isolated in the polyamine-
mutations might have pleiotropic phenotypes not recognized EDTA buffer system optimized for chicken DT40 cells (Lewis and Laemmli,

Cell 142, 810–821, September 3, 2010 ª2010 Elsevier Inc. 819


1982). 19.3 OD260 units were obtained from pooling the material of four inde- Andersen, J.S., Wilkinson, C.J., Mayor, T., Mortensen, P., Nigg, E.A., and
pendent preparations totaling 7.5 3 109 DT40 cells and solubilized in SDS- Mann, M. (2003). Proteomic characterization of the human centrosome by
polyacrylamide gel electrophoresis (SDS-PAGE) sample buffer. protein correlation profiling. Nature 426, 570–574.
Andrade, M.A., Ponting, C.P., Gibson, T.J., and Bork, P. (2000). Homology-
Preparation of Chromosome-Free Mitotic Cell Extracts based method for identification of protein repeats using statistical significance
Nocodazole blocked DT40 cells were dounce-homogenized under hypotonic estimates. J. Mol. Biol. 298, 521–537.
conditions. Mitotic chromosomes were removed by centrifuging the superna- Cheeseman, I.M., and Desai, A. (2008). Molecular architecture of the kineto-
tant twice at 10,000 x g and discarding the pellets to prepare a cell extract free chore-microtubule interface. Nat. Rev. Mol. Cell Biol. 9, 33–46.
of chromosomes. Cox, J., and Mann, M. (2008). MaxQuant enables high peptide identification
To measure the ratios between chromosomal and nonchromosomal rates, individualized p.p.b.-range mass accuracies and proteome-wide
proteins, SILAC based mass spectrometry was performed with 150 mg of protein quantification. Nat. Biotechnol. 26, 1367–1372.
labeled cell extract from 7.0 3 106 cells and 150 mg of nonlabeled proteins con-
Daum, J.R., Wren, J.D., Daniel, J.J., Sivakumar, S., McAvoy, J.N., Potapova,
tained in isolated chromosomes from 2.0 3 109 cells.
T.A., and Gorbsky, G.J. (2009). Ska3 is required for spindle checkpoint
To measure the exchange ratio, we isolated mitotic chromosomes from
silencing and the maintenance of chromosome cohesion in mitosis. Curr.
roughly 1.0 3 109 cells. Mitotic chromosomes were pelleted after centrifuging
Biol. 19, 1467–1472.
at 3000 3 g and mixed into 10 ml cell extract that were made from 3.0 3 108
de Godoy, L.M., Olsen, J.V., Cox, J., Nielsen, M.L., Hubner, N.C., Frohlich, F.,
cells. This mixture was incubated at 14 C for 30 min. Finally, we re-isolated the
Walther, T.C., and Mann, M. (2008). Comprehensive mass-spectrometry-
chromosomes as described above.
based proteome quantification of haploid versus diploid yeast. Nature 455,
1251–1254.
Mass Spectrometric Analysis Dejardin, J., and Kingston, R.E. (2009). Purification of proteins associated with
Proteins were separated into a high and a low molecular weight fraction by specific genomic Loci. Cell 136, 175–186.
SDS-PAGE, in-gel digested using trypsin (Shevchenko et al., 2006), and frac-
Earnshaw, W.C., and Rothfield, N. (1985). Identification of a family of human
tionated into 30 fractions each using SCX. The individual SCX fractions were
centromere proteins using autoimmune sera from patients with scleroderma.
desalted using StageTips (Rappsilber et al., 2003) and analyzed using
Chromosoma (Berl) 91, 313–321.
LC-MS on a LTQ-Orbitrap (Thermo Fisher Scientific) coupled to HPLC via
a nanoelectrospray ion source. The six most intense ions of a full MS acquired Finn, R.D., Mistry, J., Tate, J., Coggill, P., Heger, A., Pollington, J.E., Gavin,
in the orbitrap analyzer were fragmented and analyzed in the linear ion trap. O.L., Gunasekaran, P., Ceric, G., Forslund, K., et al. (2010). The Pfam protein
The MS data were analyzed using MaxQuant (Cox and Mann, 2008) and families database. Nucleic Acids Res. 38, D211–D222.
proteins identified by searching MS and MS/MS data using the MASCOT Foltz, D.R., Jansen, L.E., Black, B.E., Bailey, A.O., Yates, J.R., and Cleveland,
search engine (Matrix Science, UK). For more details, see the Extended Exper- D.W. (2006). The human CENP-A centromeric nucleosome-associated
imental Procedures. complex. Nat. Cell Biol. 8, 458–469.
Foster, L.J., De Hoog, C.L., and Mann, M. (2003). Unbiased quantitative pro-
teomics of lipid rafts reveals high specificity for signaling factors. Proc. Natl.
SUPPLEMENTAL INFORMATION
Acad. Sci. USA 100, 5813–5818.

Supplemental Information includes Extended Experimental Procedures, six Gaitanos, T.N., Santamaria, A., Jeyaprakash, A.A., Wang, B., Conti, E., and
figures, and five tables and can be found with this article online at doi:10. Nigg, E.A. (2009). Stable kinetochore-microtubule interactions depend on
1016/j.cell.2010.07.047. the Ska complex and its new component Ska3/C13Orf3. EMBO J. 28, 1442–
1452.
Gassmann, R., Henzing, A.J., and Earnshaw, W.C. (2005). Novel components
ACKNOWLEDGMENTS
of human mitotic chromosomes identified by proteomic analysis of the chro-
mosome scaffold fraction. Chromosoma 113, 385–397.
We dedicate this paper to Uli Laemmli on the occasion of his 70th birthday. We
thank Mayumi Takahashi for assistance with preparation of the Ska3/Rama1 Gentleman, R.C., Carey, V.J., Bates, D.M., Bolstad, B., Dettling, M., Dudoit, S.,
knockout, Frauke Melchior for anti-RanBP2 and David Tollervey, Margarete Ellis, B., Gautier, L., Ge, Y., Gentry, J., et al. (2004). Bioconductor: open soft-
Heck, Robin Allshire, Iain Cheeseman, Kumiko Samejima, Susana Ribeiro ware development for computational biology and bioinformatics. Genome
and Jan Bergmann for critical reading of the manuscript. This work was sup- Biol. 5, R80.
ported by a European Community Marie Curie Excellence Grant (JR), the Hori, T., Amano, M., Suzuki, A., Backer, C.B., Welburn, J.P., Dong, Y.,
MRC (CPP), EMBO (LSP) and the Wellcome Trust (WCE, JR). WCE and McEwen, B.F., Shang, W.H., Suzuki, E., Okawa, K., et al. (2008). CCAN makes
JR are Principal and Senior Research Fellows of The Wellcome Trust, multiple contacts with centromeric DNA to provide distinct pathways to the
respectively. outer kinetochore. Cell 135, 1039–1052.
Hubner, N.C., Bird, A.W., Cox, J., Splettstoesser, B., Bandilla, P., Poser, I.,
Received: December 7, 2009 Hyman, A., and Mann, M. (2010). Quantitative proteomics combined with
Revised: May 20, 2010 BAC TransgeneOmics reveals in vivo protein interactions. J. Cell Biol. 189,
Accepted: July 14, 2010 739–754.
Published: September 2, 2010 Hudson, D.F., Vagnarelli, P., Gassmann, R., and Earnshaw, W.C. (2003).
Condensin is required for nonhistone protein assembly and structural integrity
REFERENCES of vertebrate mitotic chromosomes. Dev. Cell 5, 323–336.
Hutchins, J.R., Toyoda, Y., Hegemann, B., Poser, I., Heriche, J.K., Sykora,
Altaf, M., Auger, A., Monnet-Saksouk, J., Brodeur, J., Piquet, S., Cramet, M., M.M., Augsburg, M., Hudecz, O., Buschhorn, B.A., Bulkescher, J., et al.
Bouchard, N., Lacoste, N., Utley, R.T., Gaudreau, L., et al. (2010). NuA4- (2010). Systematic analysis of human protein complexes identifies chromo-
dependent acetylation of nucleosomal histone H4 and H2A directly stimulates some segregation proteins. Science 328, 593–599.
incorporation of H2A.Z by the SWR1 complex. J. Biol. Chem., in press. Ishihama, Y., Oda, Y., Tabata, T., Sato, T., Nagasu, T., Rappsilber, J., and
Amano, M., Suzuki, A., Hori, T., Backer, C., Okawa, K., Cheeseman, I.M., and Mann, M. (2005). Exponentially modified protein abundance index (emPAI)
Fukagawa, T. (2009). The CENP-S complex is essential for the stable assembly for estimation of absolute protein amount in proteomics by the number of
of outer kinetochore structure. J. Cell Biol. 186, 173–182. sequenced peptides per protein. Mol. Cell. Proteomics 4, 1265–1272.

820 Cell 142, 810–821, September 3, 2010 ª2010 Elsevier Inc.


Ishihama, Y., Schmidt, T., Rappsilber, J., Mann, M., Hartl, F.U., Kerner, M.J., SILAC, as a simple and accurate approach to expression proteomics. Mol.
and Frishman, D. (2008). Protein abundance profiling of the Escherichia coli Cell. Proteomics 1, 376–386.
cytosol. BMC Genomics 9, 102. Porter, I.M., McClelland, S.E., Khoudoli, G.A., Hunter, C.J., Andersen, J.S.,
Jiang, W., Middleton, K., Yoon, H.J., Fouquet, C., and Carbon, J. (1993). An McAinsh, A.D., Blow, J.J., and Swedlow, J.R. (2007). Bod1, a novel kineto-
essential yeast protein, CBF5p, binds in vitro to centromeres and microtu- chore protein required for chromosome biorientation. J. Cell Biol. 179,
bules. Mol. Cell. Biol. 13, 4884–4893. 187–197.
Joseph, J., Liu, S.T., Jablonski, S.A., Yen, T.J., and Dasso, M. (2004). The Ran- Raaijmakers, J.A., Tanenbaum, M.E., Maia, A.F., and Medema, R.H. (2009).
GAP1-RanBP2 complex is essential for microtubule-kinetochore interactions RAMA1 is a novel kinetochore protein involved in kinetochore-microtubule
in vivo. Curr. Biol. 14, 611–617. attachment. J. Cell Sci. 122, 2436–2445.
Kops, G.J., van der Voet, M., Manak, M.S., van Osch, M.H., Naini, S.M., Brear, Rappsilber, J., Ishihama, Y., and Mann, M. (2003). Stop and go extraction tips
A., McLeod, I.X., Hentschel, D.M., Yates, J.R., 3rd, van den Heuvel, S., and for matrix-assisted laser desorption/ionization, nanoelectrospray, and LC/MS
Shah, J.V. (2010). APC16 is a conserved subunit of the anaphase-promoting sample pretreatment in proteomics. Anal. Chem. 75, 663–670.
complex/cyclosome. J. Cell Sci. 123, 1623–1633.
Rappsilber, J., Ryder, U., Lamond, A.I., and Mann, M. (2002). Large-scale pro-
Kulukian, A., Han, J.S., and Cleveland, D.W. (2009). Unattached kinetochores teomic analysis of the human spliceosome. Genome Res. 12, 1231–1245.
catalyze production of an anaphase inhibitor that requires a Mad2 template to
Schirmer, E.C., Florens, L., Guan, T., Yates, J.R., 3rd, and Gerace, L. (2003).
prime Cdc20 for BubR1 binding. Dev. Cell 16, 105–117.
Nuclear membrane proteins with potential disease links found by subtractive
Letunic, I., Copley, R.R., Schmidt, S., Ciccarelli, F.D., Doerks, T., Schultz, J., proteomics. Science 301, 1380–1382.
Ponting, C.P., and Bork, P. (2004). SMART 4.0: towards genomic data integra-
tion. Nucleic Acids Res. 32, D142–D144. Shevchenko, A., Tomas, H., Havlis, J., Olsen, J.V., and Mann, M. (2006). In-gel
digestion for mass spectrometric characterization of proteins and proteomes.
Lewis, C.D., and Laemmli, U.K. (1982). Higher order metaphase chromosome
Nat. Protoc. 1, 2856–2860.
structure: evidence for metalloprotein interactions. Cell 29, 171–181.
Takata, H., Uchiyama, S., Nakamura, N., Nakashima, S., Kobayashi, S., Sone,
Li, X., Wu, L., Corsa, C.A., Kunkel, S., and Dou, Y. (2009). Two mammalian
T., Kimura, S., Lahmers, S., Granzier, H., Labeit, S., et al. (2007). A comparative
MOF complexes regulate transcription activation by distinct mechanisms.
proteome analysis of human metaphase chromosomes isolated from two
Mol. Cell 36, 290–301.
different cell lines reveals a set of conserved chromosome-associated
Maiato, H., Sampaio, P., Lemos, C.L., Findlay, J., Carmena, M., Earnshaw, proteins. Genes Cells 12, 269–284.
W.C., and Sunkel, C.E. (2002). MAST/Orbit has a role in microtubule-kineto-
Theis, M., Slabicki, M., Junqueira, M., Paszkowski-Rogacz, M., Sontheimer,
chore attachment and is essential for chromosome alignment and mainte-
J., Kittler, R., Heninger, A.K., Glatter, T., Kruusmaa, K., Poser, I., et al.
nance of spindle bipolarity. J. Cell Biol. 157, 749–760.
(2009). Comparative profiling identifies C13orf3 as a component of the Ska
Marı́n, I. (2003). Evolution of chromatin-remodeling complexes: comparative complex required for mammalian cell division. EMBO J. 28, 1453–1465.
genomics reveals the ancient origin of ‘‘novel’’ compensasome genes.
J. Mol. Evol. 56, 527–539. Tkaczuk, K.L., Dunin-Horkawicz, S., Purta, E., and Bujnicki, J.M. (2007). Struc-
tural and evolutionary bioinformatics of the SPOUT superfamily of methyltrans-
Morrison, C., Henzing, A.J., Jensen, O.N., Osheroff, N., Dodson, H., Kandels-
ferases. BMC Bioinformatics 8, 73.
Lewis, S.E., Adams, R.R., and Earnshaw, W.C. (2002). Proteomic analysis of
human metaphase chromosomes reveals topoisomerase II alpha as an Aurora Uchiyama, S., Kobayashi, S., Takata, H., Ishihara, T., Hori, N., Higashi, T.,
B substrate. Nucleic Acids Res. 30, 5318–5327. Hayashihara, K., Sone, T., Higo, D., Nirasawa, T., et al. (2005). Proteome anal-
ysis of human metaphase chromosomes. J. Biol. Chem. 280, 16994–17004.
Neumann, B., Walter, T., Heriche, J.K., Bulkescher, J., Erfle, H., Conrad, C.,
Rogers, P., Poser, I., Held, M., Liebel, U., et al. (2010). Phenotypic profiling Van Hooser, A.A., Yuh, P., and Heald, R. (2005). The perichromosomal layer.
of the human genome by time-lapse microscopy reveals cell division genes. Chromosoma 114, 377–388.
Nature 464, 721–727. Welburn, J.P., Grishchuk, E.L., Backer, C.B., Wilson-Kubalek, E.M., Yates,
Obuse, C., Iwasaki, O., Kiyomitsu, T., Goshima, G., Toyoda, Y., and Yanagida, J.R., 3rd, and Cheeseman, I.M. (2009). The human kinetochore Ska1 complex
M. (2004). A conserved Mis12 centromere complex is linked to heterochro- facilitates microtubule depolymerization-coupled motility. Dev. Cell 16,
matic HP1 and outer kinetochore protein Zwint-1. Nat. Cell Biol. 6, 1135–1141. 374–385.
Okada, M., Cheeseman, I.M., Hori, T., Okawa, K., McLeod, I.X., Yates, J.R., Wen, H., Li, J., Song, T., Lu, M., Kan, P.Y., Lee, M.G., Sha, B., and Shi, X.
Desai, A., and Fukagawa, T. (2006). The CENP-H-I complex is required for (2010). Recognition of histone H3K4 trimethylation by the plant homeodomain
the efficient incorporation of newly synthesized CENP-A into centromeres. of PHF2 modulates histone demethylation. J. Biol. Chem. 285, 9322–9326.
Nat. Cell Biol. 8, 446–457. Wu, W.H., Alami, S., Luk, E., Wu, C.H., Sen, S., Mizuguchi, G., Wei, D., and Wu,
Ong, S.E., Blagoev, B., Kratchmarova, I., Kristensen, D.B., Steen, H., Pandey, C. (2005). Swc2 is a widely conserved H2AZ-binding module essential for
A., and Mann, M. (2002). Stable isotope labeling by amino acids in cell culture, ATP-dependent histone exchange. Nat. Struct. Mol. Biol. 12, 1064–1071.

Cell 142, 810–821, September 3, 2010 ª2010 Elsevier Inc. 821


Scientific Editor Position Available
Cell is seeking a scientist to join its editorial team. The minimum qualification for this position is
a PhD in a relevant area of biomedical research, although additional post-doctoral or editorial
experience is preferred. This is a superb opportunity for a talented individual to play a critical role
in promoting science by helping researchers disseminate their findings to the wider community.

As an editor, you would be responsible for assessing submitted research papers and overseeing
the refereeing process, and you would commission and edit material for Cell’s Leading Edge.
You would also travel frequently to scientific conferences to follow developments in research and
to establish and maintain close ties with the scientific community. You would have opportunities
to pioneer and contribute to new trends in scientific publishing. The key qualities we look for
are breadth of scientific interest and the ability to think critically about a wide range of scientific
issues. The successful candidate will be highly motivated and creative and able to work inde-
pendently as well as in a team.

This is a full-time, in-house editorial position, based at the Cell Press office in Kendall Square
near MIT in Cambridge, Massachusetts. Cell Press offers an attractive salary and benefits pack-
age and a stimulating working environment that encourages innovation.

To apply, please send your CV and a letter describing your qualifications, general research inter-
ests, and motivation for pursuing a career in scientific publishing as soon as possible to our online
jobs site: http://reedelsevier.taleo.net/careersection/51/jobdetail.ftl?lang=en&job=CEL0000R.

No phone inquiries, please. Cell Press in an equal opportunity/affirmative action employer, m/f/d/v.
The American Journal of Human Genetics
Editor Position Available

The American Society of Human Genetics is seeking an Editor for The American Journal of Human
Genetics. The Editor leads one of the world’s oldest and most prestigious journals publishing pri-
mary human genetics research.

Among the Editor’s responsibilities are determining the scope and direction of the scientific con-
tent of The Journal, overseeing manuscripts submitted for review and their publication, selecting
and supervising a staff consisting of an Editorial Assistant and doctoral-level Deputy Editor, direct-
ing interactions with the publisher (currently Cell Press), reviewing quarterly reports provided by the
publisher, evaluating the performance of the publisher, and if required, supervising the process of
the selection a new publisher. The Editor serves as a member of the Board of Directors of the Ameri-
can Society of Human Genetics (ASHG), as well as the ASHG Finance Committee, and presents
semiannual reports to the Board. All Associate Editors of The Journal are appointed by the Editor,
who also determines their duties. At the ASHG annual meeting, the Editor presides over a meeting
of the Associate Editors and presents an annual report to the ASHG membership.

The term of the appointment is five years and includes a yearly stipend. The new Editor will be
selected by the end of 2010 and will begin receiving manuscripts approximately in September 2011;
there will be partial overlap with the Boston office. Applicants should be accomplished scientists
in the field of human genetics and should have a broad knowledge and appreciation of the field.
Nominations, as well as applications consisting of a letter of interest and curriculum vitae, should be
sent to:

AJHG Editorial Search Committee


American Society of Human Genetics
9650 Rockville Pike
Bethesda, MD 20814
Cell Host & Microbe
Scientific Editor Position Available

Cell Host & Microbe, the premier microbiology primary research journal, is seeking a full-time sci-
entific editor to join its editorial team. As a scientific editor, you would be responsible for assessing
submitted research papers, overseeing the refereeing process, and choosing and commissioning
review material. You would also travel frequently to scientific conferences to follow developments
in research and establish and maintain close ties with the scientific community. This is a superb
opportunity for a talented individual to play a critical role in the research community away from the
bench.

This is a full-time in-house editorial position, based at the Cell Press office in Cambridge, Mas-
sachusetts. Cell Press offers an attractive salary and benefits package and a stimulating working
environment that encourages innovation.

The minimum qualification for this position is a PhD in a relevant area of biomedical research,
although additional postdoctoral or editorial experience is preferred. We will consider qualified can-
didates with scientific expertise in any area that falls within the scope of the journal, including molec-
ular and cell biology of microbes, host immune response to microbes, immune evasion by microbes,
microbiology, therapeutics, and vaccine design. The key qualities we look for are breadth of scien-
tific interest and the ability to think critically about a wide range of scientific issues. The successful
candidate will also be highly motivated and creative and able to work independently as well as in a
team.

Applications will be accepted through August 31, 2010. To apply, please visit the Careers page at
http://www.elsevier.com and search on keywords “Cell Host & Microbe.” Please submit a resume
and cover letter describing your qualifications, general research interests, and motivation for pursu-
ing a career in scientific publishing. Applications will be held in the strictest confidence.
Cell Press Business Project Editor
Position Available

Cell Press is seeking a Business Project Editor to plan, develop, and implement projects that have
commercial or sponsorship potential. By drawing on existing content or developing new material, the
Editor will work with Cell Press’s commercial sales group to create collections of content in print or
online that will be attractive to readers and sponsors. The Editor will also be responsible for leverag-
ing new online opportunities for engaging the readers of Cell Press journals.

The successful candidate will have a PhD in the biological sciences, broad scientific interests, a
fascination with technology, good commercial instincts, and a true passion for both science and
science communication. They should be highly organized and dedicated, with excellent written and
oral communication skills, and should be willing to work to tight deadlines.

The position is full time and based in Cambridge, MA. Cell Press offers an attractive salary and
benefits package and a stimulating work environment. Applications will be considered on a rolling
basis. For consideration, please apply online and include a cover letter and resume. To apply, visit
the career page at http://www.elsevier.com and search on keywords “Business Project Editor.”
Positions Available
Positions Available

TENURE TRACK FACULTY POSITION in NEUROSCIENCE

The Dept of Neurobiology & Anatomy at the University of Utah (http://www.neuro.utah.edu/) is seeking an outstanding scientist for a tenure track
faculty position at the Assistant Professor level. After a successful faculty search this past year, we continue with our expansion of the department in the
area of neuroscience.

We are interested in candidates who are using innovative combinations of molecular, genetic, and cellular approaches to pursue fundamental problems
in neuroscience. Areas of interest include but are not limited to in vivo imaging, genetic and epigenetic mechanisms underlying neural circuitry plasticity,
and behavior, as well as aging, regeneration and repair.

Individuals holding Ph.D. and/or M.D., or equivalent degrees, with two or more years of postdoctoral experience are encouraged to apply. Applicants
should demonstrate excellence in research and strong potential for securing and sustaining independent and collaborative extramural funding.
The University of Utah offers excellent resources to support new faculty, including competitive salary and start-up support, a highly collegial research
environment, core facilities and strong interdepartmental graduate training programs. A successful applicant will be expected to develop an innovative,
independent research program, and to share our commitment to excellence in graduate and medical education.

Only electronic applications will be accepted. Please submit a single PDF document including: 1) cover letter, 2) curriculum vitae, 3) research statement
4) one recent publication. Email the application to: facsearch@neuro.utah.edu Three letters of reference should be sent independently to: facsearch@
neuro.utah.edu

For full consideration, applications should be received by October 29, 2010.

The University of Utah is an Affirmative Action/Equal Opportunity employer and does not discriminate based upon race, national origin, color, religion, sex,
age, sexual orientation, gender identity/expression, disability, or status as a Protected Veteran. Upon request, reasonable accommodations in the application
process will be provided to individuals with disabilities. To inquire about the University’s nondiscrimination policy or to request disability accommodation,
please contact: Director, Office of Equal Opportunity and Affirmative Action, 201 S. Presidents Circle, Rm 135, (801) 581-8365.

The University of Utah values candidates who have experience working in settings with students from diverse backgrounds, and possess a demonstrated
commitment to improving access to higher education for historically underrepresented students.

ASSISTANT PROFESSOR
DEPARTMENT OF GENETICS
HARVARD MEDICAL SCHOOL

The Department of Genetics at Harvard Medical School invites applicants for a tenure-track faculty position at the
rank of Assistant Professor. The Department of Genetics consists of faculty working on diverse problems using a
variety of approaches and model organisms, unified in their focus on the genome as an organizing principle for
understanding biological phenomena. We are seeking outstanding applicants, with a demonstrated potential for
imaginative research and a clear vision for the future, who are working on exciting problems in any area of genet-
ics, broadly defined. The successful candidate is expected to direct innovative and independent research and
participate in the teaching of graduate and medical students. Significant scholarly and scientific resources are
readily available with this position. Our highly interactive Department provides the opportunity to interact and
collaborate with other dedicated researchers within the diverse Harvard research community. For further informa-
tion about our Department, please see our Web Page: http://genetics.med.harvard.edu.

Applicants should submit electronic copies of curriculum vitae, bibliography, a brief description of research
accomplishments and future research interests (limit to 500 words) by October 31, 2010, and ask three refer-
ences to provide letters of recommendation. These materials should be sent to the following email address:
faculty_search@genetics.med.harvard.edu.

We strongly encourage applications from women and minority candidates.

Harvard University is an Equal Opportunity/Affirmative Action Employer.


Positions Available

THE UNIVERSITY OF TEXAS


SOUTHWESTERN MEDICAL CENTER
AT DALLAS

The Department of Molecular Biology at the University of


Texas Southwestern Medical Center invites applications for
Assistant Professor tenure-track faculty positions. We are
seeking individuals with innovative research programs in
the areas of stem cell biology, cell signaling, development
and gene regulation. Successful applicants should be capa-
ble of establishing a vigorous independent research program
and teaching in one of several active graduate programs.
Attractive start-up packages through the Endowed Scholars
Program, state-of-the-art core facilities, and exceptional
laboratory space are available.
Applicants should submit a curriculum vitae containing a
summary of past research accomplishments, a statement of
future objectives, and names of three references to:
Eric N. Olson
Chairman
Department of Molecular Biology
University of Texas
Southwestern Medical Center at Dallas
5323 Harry Hines Boulevard
Dallas, Texas 75390-9148
UTSW is an Equal Opportunity Employer
Positions Available

Department of Pharmacology
(with potential joint appointment in the Institute for Translational Neuroscience)
University of Minnesota Medical School
TENURE/TENURE TRACK POSITION
(Assistant Professor, Associate Professor, Professor)

The Department of Pharmacology at the University of Minnesota invites applications for a tenure/tenure track faculty position (Assistant,
Associate or Full Professor). Applicants using molecular, biochemical, cellular, and/or integrative translational approaches to study
problems relevant to pharmacological sciences are encouraged to apply. Positions are also available in this joint recruitment with the
Institute for Translational Neuroscience, whose interest is in how basic science discoveries may lead to new therapeutic principles of
certain neurological disorders (see their website for more details). Qualifications include a Ph.D. in biomedical science, or an M.D.
degree, and relevant postdoctoral research experience. Applicants must have a strong record of research accomplishments, as docu-
mented by publications in leading peer-reviewed journals. The successful Assistant Professor candidate will be expected to develop
an innovative, competitive research program supported by extramural funding and to participate in departmental teaching activities.
Applicants for Associate Professor and Professor positions must demonstrate distinction in published research, evidence of consistent
extramural funding, and a commitment to teaching. For additional information about the department and the Institute for Translational
Neuroscience, visit: www.pharmacology.med.umn.edu and http://www.itn.umn.edu.

Interested applicants should apply online at http://employment.umn.edu/applicants/Central?quickFind=88959 for the Assistant


Professor position or at http://employment.umn.edu/applicants/Central?quickFind=88961 for the Assoc. Prof. or Prof. position, and
attach a letter of interest, curriculum vitae, brief statement of research interests, and contact information for three references.

DEPARTMENT OF STRUCTURAL BIOLOGY

ASSISTANT PROFESSOR OF STRUCTURAL BIOLOGY

Applications are invited for a tenure-track junior faculty appointment in the Department of Structural Biology, Stanford University School of
Medicine. Candidates should have expertise and a commitment to future research in the broad area of structural biology and biophysics. The
predominant criterion for appointment in the University Tenure Line is a major commitment to research and teaching.
To be considered for this position, interested individuals should:
1. Fully complete the Structural Biology Faculty Recruitment Form available at: https://med.stanford.edu/survey/sbio_fclty_recruit.
2. Prepare the application materials as one .pdf file and e-mail the file to sbio_faculty_recruit@lists.stanford.edu. PDF file must contain
all of the following documents:
i) Cover letter
ii) Curriculum vitae
iii) Description of research interests (4 page limit)
iv) List of publications
v) Maximum of 5 representative reprints
vi) Names and contact information of three referees
3. Arrange for three reference letters to be e-mailed by each referee directly to: sbio_fclty_reference@lists.stanford.edu
or send by mail to:
Chair, Faculty Search Committee, Department of Structural Biology, Stanford University School of Medicine, 299 Campus Drive West, D100
Fairchild Bldg., Stanford, CA 94305-5126
Candidates must have a PhD and/ or MD degree and a minimum of two years of postdoctoral research experience. Application material and
reference letters must be received by November 1, 2010.
Stanford University is an equal opportunity employer and is committed to increasing the diversity of its faculty. It welcomes
nominations of and applications from women and members of minority groups, as well as others who would bring
additional dimensions to the university's research, teaching and clinical missions.
Positions Available
Positions Available

FACULTY POSITION IN
MOLECULAR, CELLULAR, AND
DEVELOPMENTAL BIOLOGY
University of Colorado at Boulder

The Department of MCD Biology invites applications for a POSTDOCTORAL POSITIONS


tenure-track Assistant Professor in the area of molecular, cel- AVAILABLE AT SLOAN-KETTERING
lular, or developmental biology with an emphasis on basic
molecular biological problems. Applicants must have a Ph.D., INSTITUTE
M.D., or equivalent; and postdoctoral research experience.
The candidate is expected to develop a vigorous and innova- Seeking research scientists with postdoctoral experi-
tive research program, and have enthusiasm for teaching at the ence to join a laboratory in order to study the molecular
undergraduate and graduate levels. pathogenesis of multiple myeloma and utilize mouse
models to develop new therapeutic approaches.
Review of applications will begin on November 1, 2010 and
continue until the position is filled. Application materials are
Applicants must have a thorough knowledge of plasma
accepted electronically at https://www.jobsatcu.com, posting cell biology and expertise using genetically modified
number 810875. Applicants should submit a curriculum vitae mice to study cancer pathogenesis. Send a description
and a concise statement of research and teaching interests, and of your research interests, curriculum vitae, and the
arrange to have three reference letters sent. names and phone numbers of three references to

For questions or concerns on submitting your materials elec- Dr. Stephen D. Nimer
tronically, please contact mcdbsrch@Colorado.EDU. Memorial Sloan-Kettering Cancer Center
1275 York Avenue, Box 575
The University of Colorado at Boulder is committed to diver-
New York, NY 10065
sity and equality in education and employment.

See www.colorado.edu/ArtsSciences/Jobs/ for full job The Memorial Sloan-Kettering Cancer Center is an
description. Affirmative Action/Equal Opportunity Employer.
Positions Available

THE UNIVERSITY OF CALIFORNIA AT BERKELEY

Department of Molecular and Cell Biology,


Helen Wills Neuroscience
Institute and Center for Computational Biology

The Department of Molecular and Cell Biology (MCB), the Helen Wills Neuroscience Institute (HWNI)
and the Center for Computational Biology (CCB) are seeking applications for four faculty positions in
the areas listed below. We seek candidates with Ph. D. and/or M.D. degrees who have a strong inter-
est in undergraduate and graduate teaching and demonstrated excellence, originality and productivity
in research.

Biomedical Sciences (MCB)


We seek candidates interested in molecular and cellular mechanisms of disease processes. Areas of
research could include cancer, human genetics, aging, molecular physiology, animal models of
human disease, infectious and neurodegenerative diseases, or any other area of biomedical science.
This position is open at the Assistant Professor level (tenure-track).

Molecular, Cellular or Developmental Neuroscience (MCB / HWNI)


We seek candidates in any area of molecular, cellular, and developmental neurobiology, with a particular
emphasis on molecular genetic approaches to understanding neural circuit function, development, or
regeneration. Approaches could include (but are not limited to) human genetics, use of genetic model
organisms, genomics, behavioral analysis, optical imaging, and/or computational modeling. This posi-
tion is open at the Assistant Professor level (tenure-track).

Human Genome Variation (MCB / CCB)


We seek candidates whose research focuses on an understanding of genome and epigenome variation
of humans as it relates to phenotype, particularly disease predisposition, using both computational and
laboratory approaches. This position is open at the Assistant Professor level (tenure-track).

Stem Cell Biology (MCB)


We seek candidates whose research focuses on any aspect of stem cell biology, including (but not
limited to) the use of stem cells to develop models of human biology or disease, molecular mecha-
nisms of transcriptional regulation in pluripotent and differentiating stem cells, the regulation of stem
cell renewal and differentiation during development, and the biology of cancer stem cells. This posi-
tion is open at any level (tenured or tenure-track).

Applications and letters of reference should be submitted online through http://mcb.berkeley.edu.


Applications should include a curriculum vitae; a list of publications; copies of three significant
publications; a brief description of research accomplishments; and a statement of research objectives
and teaching interests. In addition, junior applicants applying for a non-tenured position should
arrange to have three letters of reference submitted online. Potential reviewers should be referred to
the Statement of Confidentiality found at: http://apo.chance.berkeley.edu/evalltr.html. The deadline
for applications is November 15, 2010.

We are interested in candidates who will contribute to diversity and equal


opportunity in higher education through their teaching, research, and service. We are
also committed to addressing the family needs of faculty. The University of California
is an Affirmative Action/Equal Opportunity Employer.
Positions Available

The Cell Biology Department seeks exceptional


candidates at the rank of Assistant and Associate
Professor who wish to engage in independent research
programs in the field. Successful candidates will have
PhD, and/or M.D. degrees and a demonstrated record
of originality and productivity in research during gradu-
ate training and/or post-doctoral research, as well as
well-formulated plans for independent research.

60
Although the focus is on tenure-track junior faculty
appointments, applicants for senior faculty ranks may
also be considered. The Department is undergoing a
major expansion under the Chair, Dr. James E. Rothman.
Exceptional candidates in all areas of cell biology will
be considered.

Please email your CV with a list of publications, a years of leadership in


summary of doctoral research [1 page], a summary of
post-doctoral research [max. 2 pages], and a research human genetics research,
plan [max. 3 pages], along with the names and addresses education and service.
(including email) of three potential references by
October 25, 2010 to: cellbio.search@yale.edu
1948–2008
Applications from, or nominations of, women and
minority scientists are encouraged. Yale is an www.ashg.org
Affirmative Action/Equal Opportunity Employer.
EuPA now has
http://www.eupa.org/
its own journal!

Editor in Chief:
Juan J. Calvete, Valencia, Spain Covered by
PubMed
Executive Editors:
Proteomics in Cell Biology
Jean-Jacques Diaz, Lyon, France

Proteomics in Microbiology
Concha Gil, Madrid, Spain

Proteomics in Plant Systems


Jesus V. Jorrín, Córdoba, Spain

Proteomics in Animal Models


Dario Neri, Zürich, Switzerland

Proteomics in Protein Science


Jasna Peter-Katalinic, Münster,
Germany

Biomedical Applications of
Proteomics and Congress
Proceedings
Jean-Charles Sanchez, Geneva,
Switzerland

Proteomics of Body Fluids and


Proteomic Technologies
Pier Giorgio Righetti, Milan, Italy Submitting Authors
Manuscripts can be submitted to the
Bioinformatics in Proteomics Journal of Proteomics at
Peter Højrup, Odense, Denmark http://ees.elsevier.com/jprot
For a complete listing of the
editorial board, visit the
journal’s homepage

To receive more information register at:


http://www.elsevier.com/locate/jprot
For the past 40 years
FEBS Letters has
published award-winning
research papers crafted
by distinguished
experts in the field.

www.febsletters.com
Just Published!

John H. Byrne and James L. Roberts, who treated us to a superb textbook in


their first edition of From Molecules to Networks, have now returned with another
outstanding effort in the second edition.
— Eric R. Kandel, Howard Hughes Medical Institute; Columbia
University College of Physicians and Surgeons, New York, USA

From Molecules to Networks


An Introduction to Cellular and Molecular
Neuroscience, Second Edition

Editors-in-Chief:
John H. Byrne, W.M. Keck Center for the
Neurobiology of Learning and Memory,
University of Texas Medical School at Houston;
James L. Roberts, Trinity University,
San Antonio, Texas

s Now includes an introduction to


mathematical modeling and simulation
approaches and a chapter on the
Neurobiology of Disease
s Full color, professional graphics throughout
s Two expansive, frequently updated
support websites:
- For students—study materials, updated
further reading references, and quizzes
- For instructors—all figures, solutions,
PowerPoint lecture slides, and more

Feb 2009, hardback 656 pp.


ISBN: 9780123741325
$94.95 USD / €69.95 EUR / £47.99 GBP

Secure online ordering at elsevierdirect.com/9780123741325

Register at our instructor site: http://textbooks.elsevier.com


This site allows registered/approved instructors to browse our list of cutting-edge books at
student-friendly prices, request print review copies, access ancillaries, view selected books
electronically, and contact your textbook representative.
60
years of leadership in
human genetics research,
education and service.
1948–2008
www.ashg.org
T H E N E W E D I T I O N O F F U N D A M E N TA L N E U R O S C I E N C E I S N O W AVA I L A B L E !

FUNDAMENTAL
NEUROSCIENCE
THIRD EDITION
EDITED BY:
Larry R. Squire, VA Medical Center and University
of California, San Diego, La Jolla, California, USA

February 2008 SENIOR EDITORS:


Darwin Berg, Floyd E. Bloom, Sascha du Lac,
Hardback Anirvan Ghosh, and Nicholas C. Spitzer
1,280 pp.
9780123740199 This comprehensive text reference presents the entire spectrum of modern neuroscience.
Print list price: Addressing feedback on the previous edition, Fundamental Neuroscience, Third Edition is
$99.95 now more concise and reader-friendly than ever before. Each chapter is heavily illustrated
and provides clinical boxes describing experiments, disorders, and methodological
approaches and concepts. A companion site contains an image bank of the figures for
use in poster presentations, slides, and handouts.
Capturing the promise and excitement of this fast-moving field, Fundamental Neuroscience,
Third Edition is the book that you will reference throughout your neuroscience career!

NEW TO THIS EDITION:


• 30% new material including new chapters on Dendritic Development and Spine
Morphogenesis; Chemical Senses; Cerebellum; Eye Movements; Circadian Timing;
Sleep and Dreaming; and Consciousness
• Companion Web site with all figures and Web links to additional material
• Multiple model system coverage beyond rats, mice, and monkeys
• Extensively expanded index for easier referencing

WHY BUY THIS EDITION?


• It presents the entire spectrum of modern neuroscience as a cohesive reference
rather than just a collection of review articles
• It is written at a level appropriate for graduate-level students and researchers with
various science backgrounds who need a solid foundation in neuroscience principles
• Clinical conditions are discussed in neuroscience to help readers establish the
connection between the basic biology and the neurological symptoms and syndromes
• It includes more than 650 illustrations, including color photographs, imaging, and
micrographs, also available on the companion Web site

SECURE ONLINE ORDERING AT ELSEVIERDIRECT.COM


.%7

Find Your Ideal Job!

så3EARCHåJOBSåBYåKEYWORD å
LOCATIONååTYPEå

så0OSTåYOURåRESUMEåå
ANONYMOUSLYå

så#REATEåAå*OBå!LERTåå
ANDåLETåYOURåIDEALåå
JOBålNDåYOU

careers.cell.com
,OOKING¬FOR¬WHAT¬#ELL¬
HAS¬PUBLISHED¬IN¬¬
%VOLUTION¬LATELY

TION
LIANå%VOLU
O å6 IS IO N åI Nå-AMMA å   å PPån
å
D A P TS åT PR å 
Tå3PECIES
å!  å!
Rå#ELLS å#ELL å
Få 2 O D å0 H OTORECEPTO REMERå4 å'UCKå* å*OFFEå" " E TW E E N å4WOå9EAS
åO å# RILITYå
RCHITECTURE å+ÙSEMå
3 å0EICHLå, å(YBRIDå3TE
.UCLEARå! - å,ANCTØTå# N O M ESå#AUSES  åPPån R
YS IN Gå Lå ' E ESISå-OTO
3OLOVEIå) å+
RE HONDRIA ECå å EDå#YTOKIN
. U C LE A Rå ANDå-ITOC EU å* 9 å# ELL åå$
A å# O N S E RV
BILITYåOFå å9ANGå39 å,
LLSå$EPRIV
EDåOFå
)NCOMPATI
åå
å#HANGå.( DELå#7 å,Iå2
å# HO Uå *9 å#HEONGå, O LU TI O N åO Få9EASTå#E G ( AM PTONå+ å3EI
,EEå( 9 TIVEå% V ! å3TAEH LIN
APIDå!DAP Nå+ å0ERERAå
P LO ID Yå 5 NDERLIESå2 å.OLLå! å4RIMBLEå2 å7ALTO Gå9EAST +2 åå
!NE U å&LEHARTYå" O N åINå"UDDIN '2 å&OSTERå
2ANCATIå' å0
AV ELKA å.
å  n  IK E å# O O P E RA TI
# å, AT GÎå*0 å&INKå
 å PP IOlLM , Nå! å0REVO
ST å-
å.OVå å Tå$RIVESå" Så-$ å*ANSE
#ELL å E A RD å'ENEåTHA å9ANå# å6INCE
N å" QUENCING
RE E GN INIå3
å6ARIABLEå' HETå. å"EAUVAISå! å'UA
DA
UGHPUTå3E å3IEBAUERå- å"URBANO
å
&,/åISåA RA å- å0 OC å  n  åB Yå ( IG H 4 H RO
FERå+
å# AL DA   å PP
ETERM IN E D EL å5 å0Rß åå
3MUKALLAå3 å.OVå
 å
EQUENCEå$ å4 å3TENZ å0ÊÊBOå3
å+*åå#ELL å *- å-ARICIC å* å3LATKINå-
6ERSTREPEN H O N D RI A Lå 'ENOMEå3 HLERå# å-EYERå- å'OODå å, AA KK ON ENå, å+ELSO
RTALå-ITOC NSONå0, å5 | ) å7IKSTRÙ
Må-
TEå.EANDE RAUSEå* å"RIGGSå!7 å*OH OVICå | Nå: å'US
ICå
!å#OMPLE å+ RA JK | $ å+UCA ON
CEå%VOLUTI
Så ! 3 å0 å"
ALASPINA å- å2UDAN
'REENå2% å-
OT HB ER Gå *- å%GHOLM O N å# O D IN G 3EQUEN
(! å2ONAN
å- å2 n NSTRAINTå
 åPPå MINANTå#O
#ELLå
å!UGå å
IS FO LD IN GåASåAå$O ROSOPHILA

N D U C E D å0 ROTEINå-  å PP ån IM O RP H IC å4RAITSåINå$
TION ) å*ULå å
 UALLYå$
-ISTRANSLA #/å#ELL å OLLINGå3EX ån
M ON Då $! å7ILKEå N E TI C å3 W ITCHå#ONTR å!UGå å åPP ,,å&AMILY

+./8"%
$RU M Få A å' E å# EL L å
V O LU TI O N åO å# AR RO LLå 3 "
E å0 LA N Tå
å% ! NåOFåTH
LATIONåAND ERNERå4 å'OMPELå. å+OP

4HEå2EGU Då%VOLUTIO
4- å3 EL EG UEå*% å7
TE RO D IM E RIZATIONåAN
7ILLIAMSå INå(E n
MEOPROTE  åPPå
E X U A Lå / RI GINSåOFå(O 5å#ELL åå-AYå å
%ARLYå3 OODENOUGHå
å( å*OOå3 å'
,EEå*( å,IN

,OOKå!GAIN
$ISCOVERå-ORE
WWWCELLCOM
SnapShot: Nuclear Receptors I
Neil J. McKenna and Bert W. O’Malley
Baylor College of Medicine, Houston, TX 77030, USA

Receptor/ Disease
Family* Symbols Ligands Major Functions Associations Target Genes
Estrogen ERα/NR3A1; Endogenous: 17β-estradiol Regulation of cell growth and proliferation Cancer, cardiovascular, ↑ MYC, NGF, BCL2, CXCL2,
receptors* ERβ/NR3A2 in multiple tissues (e.g., female reproductive immune and IGF1, TYMS; ↓ CD36, NDRG1,
Clinical: Mixed agonists (e.g. tamoxifen, tissues, bone, and CNS) inflammatory, NCOR1, NCOA3
raloxifene, and toremifene in breast cancer) metabolic, neurological,
Xenobiotics: Bisphenol A, PCBs reproductive

Androgen AR/NR3C4 Endogenous: Testosterone, Key role in male reproductive organs in Cancer, cardiovascular, ↑ MYC, VEGF, BCL2, IGF1,
receptor dihydrotestosterone addition to other systems (e.g., CNS) immune, metabolic, MUC1, P66(Shc), CCND1;
neurological, ↓ TSHA, TSHB, PTEN, FAS,
Clinical: Flutamide and bicalutamide for reproductive CASP2, CTNND2, ESR2,
prostate cancer and alopecia TMPRSS2

Glucocorticoid GR/NR3C1 Endogenous: Cortisol (hydrocortisone) Diverse developmental and physiological Metabolic, ↑ SCNN1A, GADD45B, GILZ,
receptor roles (e.g., antagonism of inflammatory cardiovascular, immune TAT; ↓ BGLAP, POMC, INS
Clinical: Fluticasone and prednisolonein signaling pathways, mediation of the stress and inflammatory,
inflammatory disorders response, and gluconeogenesis) memory

Vitamin D VDR/NR1I1 Endogenous: Calcitriol (1′,25′ dihdroxy Maintenance of serum calcium and Bone, cancer, ↑ FGF23, CYP24A1, CALB1,
receptor vitamin D3) phosphate levels for skeletal integrity; cardiovascular, BGLAP, SPP1; ↓ IL2, PHEX
antiproliferative in many tissues metabolic, immune and
Clinical: Paracalcitol for 2o inflammatory, renal,
hyperparathyroidism in renal patients; neurological
Tacalcitol for psoriasis

Thyroid hormone TRα/NR1A1; Endogenous: Thyroxine Regulation of oxygen consumption; protein, Thyroid conditions, ↑ ADRB1, PCK2, GH1, UCP1; ↓
receptors* TRβ/NR1A2 (T4),Triiodothyronine (T3) carbohydrate, lipid, and vitamin metabolism cancer DIO2, EHHADH, PRL, EGFR

Clinical: Levo-thyroxine, triiodothyroacetic


acid (TRIAC) in resistance to thyroid
hormone

Progesterone PR/NR3C3 Endogenous: Progesterone Diverse reproductive functions (e.g., Cancer, metabolic, ↑ SERPINB14, FAS, MT2A,
receptor establishing and maintaining pregnancy, reproductive PGC, EGFR , IHH; ↓ ESR1, PGR,
Clinical: RU486 (Mifepristone) as an developing breast tissue, and stopping ANXA6
abortifacient proliferation in the uterus)

Mineralocorticoid MR/NR3C2 Endogenous: Aldosterone Regulating electrolyte and fluid balance in Metabolic ↑ SCNN1A, ATP1A1, ATP1B1,
receptor the kidney; specific roles in the CNS GILZ, SGK1, NDRG2
Clinical: Spironolactone in hypertensive
cardiovascular disease

Peroxisome- PPARγ/NR1C3 Endogenous: FAs and FA intermediates Regulation of adipocytes, insulin sensitivity Cardiovascular, ↑ FABP4, UCP1, AP2, PCK1,
proliferator- and lipogenesis, and broader integration of metabolic, cancer, LPL, ADIPOQ, CD36, AQP7
activated Dietary: FAs and PUFAs energy, lipid, and carbohydrate metabolism neurological
receptor-γ
Clinical: Thiazolidinediones (e.g.,
rosiglitazone) in type II diabetes

Peroxisome- PPARα/ Endogenous: FAs and FA intermediates Regulating energy expenditure; modulating Cardiovascular, ↑ ACBP, ACOX1, APOA1,
proliferator- NR1C1 fatty acid oxidation systems (mitochondria), metabolic, cancer, CPT1A, CYP1A1, CYP4A1,
activated Clinical: Fibrates (e.g., fenofibrate) in peroxisome β-oxidation, and microsomal neurological CYP7A1, SLC27A1, LCAS,
receptor-α hyperlipidemia ω-oxidation MLYCD, SCD, FADS2, RETN,
Dietary: FAs and PUFAs MYC, CCND1, IGFBP1, UCP1,
KRT23, IL6, TF, PEX11A
Xenobiotics: DEHP, DEHA

Peroxisome- PPARδ/ Endogenous: FAs and FA intermediates Regulating cell proliferation, differentiation, Cardiovascular, ↑ ACSL3, CPT1A, RGS3, RGS4,
proliferator- NR1C2 and migration in wound healing and metabolic, cancer, RGS5, ISG20, CXCL7, CCL21,
activated Dietary: FAs and PUFAs inflammatory processes neurological RETN, CPT1A
receptor-δ (β)

Retinoic acid RARα/NR1B1; Endogenous: All-trans and 9-cis retinoic Pleiotropic control of embryonic patterning Neurological and ↑ Numerous HOX genes, STRA6,
receptors* RARβ/NR1B2; acid and organogenesis, cell proliferation, psychiatric, cancer HNF3A, CRABP2, ACADM,
RARγ/NR1B3 differentiation, apoptosis and homeostatic MECOM; ↓ CYP1A1, HOXB9
Clinical: Tretinoin for treating acne and control
acute promyelocytic leukemia

Liver X receptors* LXRα/NR1H3; Endogenous: Oxysterols Cholesterol and steroid sensors with roles in Metabolic ↑ SREBP1C, CYP7A1, ABC8,
LXRβ/NR1H2 lipid and carbohydrate metabolism APOA1, APOE, LPL, PLTP

Retinoid X RXRα/NR2B1; Endogenous: 9-cis retinoic acid Embryonic cell patterning and Neurological and ↑↓ many genes as heterodimers
receptors* RXRβ/NR2B2; organogenesis, cell proliferation and psychiatric, immune with other receptors (e.g.,LXRs,
RXRγ/NR2B3 differentiation, other functions as PPARs, FXR, TRs, RARs; ↑ ABC1
heterodimers with other nuclear receptors (with LXR); ↓ CYP7A1 (with FXR)

Pregnane X PXR/NR1I2 Endogenous: Bile acids Metabolism and transport of pharmaceutical Immune ↑ Multiple CYP2 and CYP3
receptor drugs, xenobiotics, and toxic bile acids in gene family members, MDR1,
Xenobiotic: St. John’s Wort (hyperforin), the liver and GI tract MRP2, OATP2, UGT1A1, SULT,
Taxol, rifampicin, phenobarbital ↓ CYP7A1
Dietary: Vitamin E, sulforaphane, Gugulipid

Constitutive CAR/NR1I3 Endogenous: Androstanol, androstenol Metabolism of xenobiotics and endogenous Involved in ↑ CYP2B10, CYP311A, CYP3A4,
androstane lipids by regulating expression of hepatotoxicity of CYP1A2, CYP2B6 THRSP,
receptor Xenobiotics: Phenobarbital, DEHP, cytochrome P450 genes acetaminophen SLC21A6, MRP2, MDR1, OATP2
Meclizine

Farnesoid X FXR/NR1H4 Endogenous: Bile acids (e.g., A sensor for bile acid that helps regulate bile Metabolic ↑ SLC10A2, ABCB1, ABCB11,
receptor chenodeoxycholic acid) acid homeostasis NR0B2, HSD3B2, FETUB,
ABCB4, FGF19, NOS2; ↓
Dietary: Cafestol, guggulsterone CYP7A1, HNF1A, HNF4A,
SLCO1B1, SLC10A2

822  Cell 142, September 3, 2010 ©2010 Elsevier Inc.  DOI 10.1016/j.cell.2010.08.026 See online version for legend and references.
4HEå"IOPHYSICALå3OCIETYå
ANDå#ELLå0RESS
!åNEWåPARTNERSHIP

3EEåWHATSåNEWåAT
WWWBIOPHYSJORG
SAN TA CR UZ B I OT ECH N O LO GY, I N C . SANTA CRUZ
®

Leading (#1 worldwide) supplier BIOTECHNOLOGY

of specialty biochemicals for


The Power to Question

proteomics research.

Products include over 71,300 unique biochemicals


sorted alphabetically, by CAS number, by formula
and on the basis of functional activity.

See our web site for product details


and ordering information.

www.scbt.com

HEADQUARTERS
TOLL FREE: 800.457.3801 TEL: 831.457.3800
FAX: 831.457.3801 E-MAIL: scbt@scbt.com

EUROPEAN SUPPORT
TOLL FREE: +00800.4573.8000 PHONE: +49.6221.4503.0
FAX: +49.6221.4503.45 E-MAIL: europe@scbt.com

ASIAN SUPPORT
JAPAN TOLL FREE: (010) 800.40402026 S. KOREA TOLL FREE: 00798.1.1.002.0297
N. CHINA TOLL FREE: 10.800.711.0752 S. CHINA TOLL FREE: 10.800.110.0694
FAX: 831.457.3801 E-MAIL: asia@scbt.com

ChemCruz ™

Das könnte Ihnen auch gefallen