Sie sind auf Seite 1von 14

Loss of Fancc Impairs Antibody-Secreting

Cell Differentiation in Mice through


Deregulating the Wnt Signaling Pathway
This information is current as Mathieu Sertorio, Surya Amarachintha, Andrew Wilson and
of July 12, 2017. Qishen Pang
J Immunol 2016; 196:2986-2994; Prepublished online 19
February 2016;
doi: 10.4049/jimmunol.1501056
http://www.jimmunol.org/content/196/7/2986

Downloaded from http://www.jimmunol.org/ by guest on July 12, 2017


Supplementary http://www.jimmunol.org/content/suppl/2016/02/19/jimmunol.150105
Material 6.DCSupplemental
References This article cites 44 articles, 23 of which you can access for free at:
http://www.jimmunol.org/content/196/7/2986.full#ref-list-1
Subscription Information about subscribing to The Journal of Immunology is online at:
http://jimmunol.org/subscription
Permissions Submit copyright permission requests at:
http://www.aai.org/About/Publications/JI/copyright.html
Email Alerts Receive free email-alerts when new articles cite this article. Sign up at:
http://jimmunol.org/alerts

The Journal of Immunology is published twice each month by


The American Association of Immunologists, Inc.,
1451 Rockville Pike, Suite 650, Rockville, MD 20852
Copyright © 2016 by The American Association of
Immunologists, Inc. All rights reserved.
Print ISSN: 0022-1767 Online ISSN: 1550-6606.
The Journal of Immunology

Loss of Fancc Impairs Antibody-Secreting Cell


Differentiation in Mice through Deregulating the Wnt
Signaling Pathway

Mathieu Sertorio, Surya Amarachintha, Andrew Wilson, and Qishen Pang


Fanconi anemia (FA) is characterized by a progressive bone marrow failure and an increased incidence of cancer. FA patients have
high susceptibility to immune-related complications such as infection and posttransplant graft-versus-host disease. In this study, we
investigated the effect of FA deficiency in B cell function using the Fancc mouse model. Fancc2/2 B cells show a specific defect in
IgG2a switch and impaired Ab-secreting cell (ASC) differentiation. Global transcriptome analysis of naive B cells by mRNA
sequencing demonstrates that FA deficiency deregulates a network of genes involved in immune function. Significantly, many
genes implicated in Wnt signaling were aberrantly expressed in Fancc2/2 B cells. Consistently, Fancc2/2 B cells accumulate high
levels of b-catenin under both resting and stimulated conditions, suggesting hyperactive Wnt signaling. Using an in vivo Wnt GFP
reporter assay, we verified the upregulation of Wnt signaling as a potential mechanism responsible for the impaired Fancc2/2

Downloaded from http://www.jimmunol.org/ by guest on July 12, 2017


B cell differentiation. Furthermore, we showed that Wnt signaling inhibits ASC differentiation possibly through repression of
Blimp1 and that Fancc2/2 B cells are hypersensitive to Wnt activation during ASC differentiation. Our findings identify Wnt
signaling as a physiological regulator of ASC differentiation and establish a role for the Wnt pathway in normal B cell function
and FA immune deficiency. The Journal of Immunology, 2016, 196: 2986–2994.

T
he B cells are essential for the humoral-based immunity. Pax5, which is responsible for the expression of genes involved in
After encountering an Ag, B cells undergo genomic B cell function and the repression of genes involved in ASC dif-
mutation and recombination, proliferation, and differen- ferentiation, such as the master regulator of ASC differentiation,
tiation. At the genomic level after encountering an Ag, B cells Blimp1 (2, 3). After induction, Blimp1 represses Pax5, allowing
undergo two induced cytidine deaminase processes called somatic ASC differentiation while blocking proliferation through repres-
hypermutation (SHM) and class switch recombination (CSR). sion of c-Myc (4) and by indirect induction of Xbp-1 (5). There
SHM results in introduction of point mutations in the V regions of are two types of ASCs: a first wave of low-affinity and short-term
the Ig gene to enhance Ig affinity for Ags. CSR leads to recom- ASC-producing IgM and a second type of high-affinity switched
bination by nonhomologous end joining (NHEJ) DNA repair of ASCs that can migrate from secondary lymphoid organs to the
the IgM C region (Cm) with one of the downstream C regions to bone marrow (BM) to become long-term nondividing ASCs (6).
generate different classes of Ab (IgD, IgG, IgE, or IgA) (1). After Fanconi anemia (FA) is characterized by a progressive BM
being selected, the high-affinity B cells differentiate either into failure and a high susceptibility to develop leukemia and solid
memory B cells, which allow a faster immune response in case of tumors. The disease is due to a mutation in one of the 19 already
a second encounter with the same Ag, or into Ab-secreting cells identified genes (A to Q) (7). Deficiency in any one of these FA
(ASC; also called plasma cells), which are able to produce a high gene-encoding proteins leads to genomic instability and high
quantity of Ig. Differentiation into plasma cells is inhibited by susceptibility to cancer development (8). FA proteins are mainly
involved in DNA repair after DNA damage or replicative stress.
Upon activation of the FA pathway, eight FA proteins (FANCA,
Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s -B, -C, -E, -F, -G, -L, and -M) interact to form the FA core
Hospital Medical Center, Cincinnati, OH 45229 complex that activates FANCD2 and FANCI by mono-
ORCID: 0000-0003-1076-2802 (S.A.). ubiquitination (8). The activation of FA pathway is thought to
Received for publication May 8, 2015. Accepted for publication January 20, 2016. favor the homologous recombination while inhibiting the error-
This work was supported by National Institutes of Health, National Heart, Lung, and prone NHEJ DNA repair (9, 10). Aside from DNA repair, other
Blood Institute Grant R01 HL076712 and National Cancer Institute Grant R01
CA157537. Q.P. was supported by a Leukemia and Lymphoma Scholar award.
specific functions have been described for some FA proteins. For
example, FANCC is able to interact with heat shock protein 70 to
The RNAseq data in this article have been submitted to the National Center for
Biotechnology Information’s Gene Expression Omnibus (http://www.ncbi.nlm.nih. inhibit TNF-a–induced apoptosis (11, 12), with STAT-1 to allow a
gov/geo/query/acc.cgiacc=GSE76634) under accession number GSE76634. normal IFN-g response (13, 14) and with CtBP1 and b-catenin to
Address correspondence and reprint requests to Dr. Qishen Pang, Division of Exper- modulate the WNT signaling pathway (15, 16).
imental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical A lot of effort has been made to understand, improve, and try to
Center, 3333 Burnet Avenue, Cincinnati, OH 45229. E-mail address: qishen.
pang@cchmc.org cure the BM failure of FA patients. Most of the studies on FA
The online version of this article contains supplemental material. proteins are focused on their roles in DNA repair function and
Abbreviations used in this article: ASC, Ab-secreting cell; BM, bone marrow; CSR, hematopoietic stem cell maintenance. To date few studies have
class switch recombination; FA, Fanconi anemia; FC, fold change; NHEJ, nonho- addressed the immune function of FA proteins (17). Because high
mologous end joining; NP, (4-hydroxy-3-nitrophenyl) acetyl; PB, peripheral blood; susceptibility to general infection has been reported for a group of
RNAseq, mRNA sequencing; SHM, somatic hypermutation; WT, wild-type.
FA patients (17), the question of immune function in the context
Copyright Ó 2016 by The American Association of Immunologists, Inc. 0022-1767/16/$30.00 of FA deficiency seems of interest to understand and predict

www.jimmunol.org/cgi/doi/10.4049/jimmunol.1501056
The Journal of Immunology 2987

possible complications aside from the development of BM failure (eBioscience). Ten minutes before flow cytometry acquisition, propidium
and cancer. More recently, the study of APCs has demonstrated iodide (eBioscience) was added to all samples for detection and/or ex-
clusion of dead cells. Cells were analyzed with a FACSFortessa or a
impaired function of Fancc-deficient macrophages (18). It has also FACSCanto (BD Biosciences) and Diva (BD Biosciences) or FLowJo
been reported that a subgroup of FA patients has an impaired (Tree Star) softwares.
immunization after pneumococcal vaccination (19), whereas an-
other recent study reported a normal immunization of FA-deficient RNA isolation, RNA NextGen sequencing analysis, and
women vaccinated with human papillomavirus vaccine (20). In real-time quantitative PCR
mice, a study has reported an impaired Ab response in Fancc- Total RNAwas purified with RNeasy Plus mini kit from B cells ex vivo, after
deficient animals immunized with only a human papillomavirus culture or after CFSE peak-based cell sorting by FACSAria (3 division
vaccine formulation containing a TLR4 adjuvant (21). The dif- peak). For mRNA sequencing (RNAseq), poly-A+ isolation, library, single-
end sequencing, and alignment of reads on mouse genome (mm9 version)
ferences seen in immunization efficiency in FA patients and vac- were done by the Cincinnati Children’s Hospital Medical Center DNA and
cine formulation in mice raise the question of a specific deficiency sequencing core according to their standard protocol and QC filters. Bam
of B cells for certain complementation groups and specificity for files provided by the DNA and sequencing core were analyzed using
certain pathogens or adjuvants. In this study, we show that B cells GeneSpring GX v12 (Agilent). Quantification of mRNA expression was
done using the RefSeq database. Gene expression and fold change (FC)
from mice deficient for the Fancc gene have a specific defect in were evaluated between WT and Fancc2/2 sample. The RNAseq data have
Ab-secreting cell (ASC) differentiation. We further demonstrate been deposited in National Center for Biotechnology Information’s Gene
that Fancc deficiency deregulates a network of genes involved in Expression Omnibus under the accession number GSE76634 (http://www.
B cell activation and ASC differentiation and identify hyperactive ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE76634). The genes with a
Wnt signaling as a potential mechanism responsible for the im- significant difference (moderate-t test, p # 0.05) and a FC $ 1.5 were
selected to run the pathway analysis module of GeneSpring GX v12 using
paired Fancc2/2 ASC differentiation. the curated WikiPathway database (http://www.wikipathways.org). For

Downloaded from http://www.jimmunol.org/ by guest on July 12, 2017


quantitative PCR experiments, reverse transcription was performed on 500
ng RNA with the High-Capacity cDNA Reverse Transcription kit (Life
Materials and Methods Technology), as recommended by the manufacturer. cDNA were used for
Mice and treatment real-time PCR with the primer listed in Supplemental Table I. Relative
expression of each mRNA was calculated using the 22d d CT method using
Fancc2/2 mice (C57BL/6 background) were described elsewhere (22).
ActB and Hprt1 as endogen controls and average of WT mRNA sample
Heterozygous Fancc+/2 mice were bred to obtain Fancc2/2 and Fancc+/+
expression as reference sample.
(wild-type [WT]) littermate offspring. Animals were maintained in the
animal barrier facility at Cincinnati Children’s Hospital Medical Center. ELISA quantification of total IgM and NP-specific anti-IgG
Six- to 8-wk-old mice were used for the entire study. BM was flushed with
PBS from two tibias and two femurs of each mouse and labeled for flow Total IgM Abs from B cell culture supernatant (5 d) and serum of LPS-
cytometry analysis of B cell lineage or for LSK cell sorting. Splenocytes injected mouse (3 d) were measured with the Mouse IgM ELISA
were obtained by smashing mouse spleens on a 70-mm strainer placed in a Ready-Set-Go kit (eBioscience), according to the manufacturer, protocol
35-mm petri dish. After osmotic lysis of RBCs by ACK buffer, splenocytes using 3,39,5,59 tetramethylbenzidine as HRP substrate. NP-specific IgG
were either used for B cell isolation or flow cytometry analysis. For in vivo Abs from serum of NP-LPS–immunized mice were measured by ELISA.
treatment, 6- to 8-wk-old mice were injected i.v. with 15 mg LPS (LPS Ninety-six–well Elisa plates (Costar) were coated with 10 ug/ml NP29-
from Salmonella typhosa; Sigma-Aldrich). For immunization, mice were BSA (BIOSEARCH Technologies) overnight. After 1-h blocking with PBS
injected i.p. with 100 mg (4-hydroxy-3-nitrophenyl) acetyl (NP)-LPS 1% BSA, the wells were incubated with diluted sera for 2 h, and NP-
(BIOSEARCH Technologies) in PBS or with 200 ml PBS only as con- specific IgG were detected by a secondary HRP-linked anti-mouse IgG
trol. All experiments were approved by the Veterinary Services at Cin- (BioLegend) and 3,39,5,59 tetramethylbenzidine. Reaction was stopped by
cinnati Children’s Hospital Medical Center and conducted in accordance 1 M phosphoric acid, and absorbance was measured at 450 nm.
with the National Institutes of Health guidelines for animal care.
Lentivirus transduction and LSK transplantation
Cell culture
Mononuclear BM cells from WT or Fancc2/2 mice (CD45.2) were labeled
B cells were purified by negative selection with anti-CD43 magnetic beads with the lineage marker (Lin) biotinylated Abs mixture (BD Biosciences)
(Miltenyi Biotec; .95%) from RBC-lysed mouse splenocytes using au- including the following: anti-CD3ε (clone 145-2C11), CD11b (clone M1/
tomatic labeling and magnetic separation on an AutoMacs-Pro (Miltenyi 70), CD45R/B220 (clone RA3-6B2), Ly-76 (clone Ter119), Ly6G, and Ly-
Biotec). B cells were cultured at 5 3 105 cells/ml in complete RPMI 1640 6C (clone RB6-8C5). Cells were then labeled with anti–Sca1-PeCy7 (D7)
medium (10% FBS, 25 mM HEPES, 2 mM L-glutamine, nonessential and anti–cKit-allophycocyanin (2B8) (BD Biosciences). After washing,
amino acid, 50 mM 2-ME, 1% penicillin/streptomycin) with 20 mg/ml LPS biotinylated Abs were revealed by streptavidin PerCP-Cy5.5 (BD Biosci-
alone (serotype 0111:B4 from Escherichia coli; Sigma-Aldrich) to induce ences). Lin2Sca1+Ckit+ (LSK) cells were sorted using FACSAria (BD
IgG3 CSR or in combination with 5 ng/ml murine rIL-4 (PeproTech) to Biosciences) from the Cincinnati Children’s Hospital Medical Center Flow
induce IgG1 CSR or 50 ng/ml murine rIFN-у (PeproTech) to induce Cytometry Core. Sorted LSKs ($98%) were activated in StemSpan me-
IgG2a. For CFSE labeling, B cells were loaded before culture with 1.5 mM dium (Stemcell Technologies) in presence of 25 ng/ml murine rTpo
CFSE (Life Technology). For culture in presence of Wnt3a, 25% of culture (Peprotech) and 50 ng/ml murine recombinant stem cell factor (Pepro-
volume was replaced by Wnt3a conditioned medium (gift of Dr. Helmrath, Tech). After 12 h, LSKs were transduced by the 7TGC-eGFP Wnt reporter
Cincinnati Children’s Hospital Medical Center) or control medium. lentivirus (23) with 2 hits at multiplicity of infection = 5. Lentiviral par-
ticles were produced by the Cincinnati Children’s Hospital Medical Center
Flow cytometry analysis Viral Vector core using 293T cells. At 72 h, transduction efficiency was
evaluated by mCherry detection by flow cytometry (transduction efficiency
Mononuclear BM cells, B cells after culture, or splenocytes were treated 10
70–80%). Five thousand LSK cells were transplanted into lethally irradi-
min with FC block solution (BD Biosciences) and then labeled 20 min on ice
ated Boy/J recipient mice (CD45.1). GFP expression of naive splenic
with combination of different anti-mouse Abs, as indicated in legends of
B cells was detected at 10 wk posttransplantation by flow cytometry.
figures. Abs included anti–B220-eFluor660 (eBioscience; clone RA3-6B2),
anti–IgM-Fitc (eBioscience; clone eB121-15F9), anti–IgG1-PE (eBio-
science; clone m1-14D12), biotin anti-IgG2ab (BD Biosciences; clone 5.7), Results
biotin anti-mouse IgG3 (BD Biosciences; clone R40-82), anti–CD138-
Normality of peripheral B cell compartment despite differences
BrillantViolet421 (BD Biosciences; clone 281-2), anti–CD43-PE (BD
Biosciences; clone S7), anti–CD45.1-PeCy7 (BD Biosciences; clone A20), of B cell population in the BM of Fancc2/2 mice
and anti–CD45.2-allophycocyanin-Cy7 (BD Biosciences; clone 104). In FA patients, a previous study reported a decrease of peripheral
Biotinylated primary Abs were detected by incubation of labeled cells with
streptavidin-PE (BD Biosciences). For apoptosis analysis, B cells were first blood (PB) B cell number (24). We thus first analyzed B cell
surface labeled for ASC detection, then washed and resuspended in 13 development in the BM and PB of Fancc2/2 mice and WT con-
binding buffer (eBioscience), and labeled with anti-annexin V FITC trol littermates. In the BM, adult Fancc2/2 mice exhibited a sig-
2988 ASC DIFFERENTIATION DEFECT IN Fancc2/2 MICE

nificant difference in the proportion of B cell lineage subpopula- or ASCs (also called plasma cells) (25). ASCs are the principal
tions compared with WT mice. Specifically, Fancc2/2 mice source of Abs, and extensive efforts have been made to charac-
showed a significant decrease in the frequencies of immature terize the process and factors involved in ASC differentiation (6).
(IgM+B220dim) and mature B cells (IgM+B220+) accompanied by We evaluated ASC differentiation in vitro, using B220 and CD138
an increase of pre/pro-B cells (IgM2B220dim) (Fig. 1A, 1B). labeling. As shown in Fig. 2A and 2B, 5 d of culture led to a
However, no significant difference was observed for total BM significant decrease in ASC (B220dimCD138+) cells in Fancc-de-
cellularity or even total B cell count in PB between Fancc2/2 and ficient B cells compared with WT cells for all three conditions
WT mice (Fig. 1C, Supplemental Fig. 1A). Interestingly, we found tested. We next evaluated the expression of Blimp1, which is the
that the percentage and the absolute number of total B cell lineage major transcription factor controlling ASC differentiation (6).
population (B220dim/+) were significantly increased in the BM of Consistent with the observed decrease in B220dimCD138+ cells,
Fancc2/2 mice compared with WT mice (Fig. 1D, 1E). We next the expression of Blimp1 mRNA was downregulated in Fancc2/2
analyzed the B cell subpopulations in the spleen. As shown in B cells (Fig. 2C). To further substantiate these observations, we
Supplemental Fig. 1B–D, no difference was observed in spleen measured the production of IgM in supernatants of B cells cul-
cellularity and in the percentage of total B (B220+) cells in live tured for 5 d. As expected, LPS-stimulated B cells from both
splenocytes. We also evaluated the B cell subpopulations in the genotypes produced an increased amount of IgM compared with
spleen using IgM, IgD, or CD21 and CD23 labeling and observed resting cells (Fig. 2D). However, we observed a significant de-
no significant difference in the proportion of the different B cell crease of IgM production in supernatants from stimulated Fancc2/2
subpopulations in Fancc2/2 mice compared with WT mice B cells compared with stimulated WT cells (Fig. 2D). We next
(Supplemental Fig. 1E–G). These data indicate that, despite the evaluated the ability of Fancc2/2 B cells to differentiate into
aberrant B cell development seen in the BM, Fancc2/2 mice have ASCs in vivo. To this end, we treated Fancc2/2 and WT mice with

Downloaded from http://www.jimmunol.org/ by guest on July 12, 2017


normal B cell number in periphery and normal B cell subpopu- LPS and determined the proportion of CD138+B200dim/2 cells in
lations in the spleen in steady state. The increase in BM B cells live splenocytes 3 d postinjection. As shown in Fig. 2E and 2F,
could compensate for the imbalance of B cell development ob- LPS injection induced a burst in the CD138+B200dim/2 population
served in Fancc2/2 mice and thus maintain B cell number and in the spleen of both Fancc2/2 and WT mice. However, stimulated
characteristics in periphery. Fancc2/2 mice exhibited much weaker induction of ASC differ-
entiation than their WT controls (Fig. 2E, 2F). Consistently,
Impaired ASC differentiation of Fancc2/2 B cells Fancc2/2 mice showed a significant decrease in serum IgM
Upon encountering an Ag, B cells go through SHM and CSR and concentration compared with WT mice (Fig. 3G). Furthermore,
enter differentiation programs to differentiate into memory B cells immunization of mice with NP-LPS, a T cell–independent Ag,

FIGURE 1. Abnormality of B cell lineage in bone marrow of Fancc2/2. BM was harvested from tibias and femurs of 6- to 8-wk-old WT or Fancc2/2
mice. Mononuclear cells were separated by centrifugation on Ficoll and labeled with anti–B220-eFluor660 and anti–IgM-FITC Abs. Before FACS ac-
quisition, propidium iodide (PI) was added to detect and exclude dead cells. (A) Representative FACS plot of the evaluation of pre/pro (B220dimIgM2),
immature (B200dimIgM+), and mature (B220+IgM+) among live B220dim/+ mononuclear BM cells from Fancc2/2 or WT littermate mice. (B) Quantification
of BM B cell populations (n = 6 mice/genotype), as described in (A). (C) Bone marrow cellularity from two tibias and femurs of WT and Fancc2/2 mice
(n = 6 mice/genotype). (D) Representative FACS histogram of total B cell lineage percentage in live mononuclear BM cells from Fancc2/2 or WT mice. (E)
Total B cell (B220+) number from two tibias and femurs (n = 6 mice/genotype). For all graphics, error bar represents mean value 6 SD. Evaluation of
differences between genotype has been conducted using Student t test (*p # 0.05).
The Journal of Immunology 2989

induced significantly lower levels of NP-specific IgG in the serum indicating the activation of Wnt signaling after LPS stimulation
of Fancc2/2 mice compared with WT mice (Fig. 2H). Together, (Fig. 3D), as previously reported (30). Interestingly, we observed
these results indicate that Fancc2/2 B cells have a decreased higher levels of b-catenin in Fancc2/2 B cells than WT cells at
ability to differentiate into ASCs and to produce Abs. both resting and stimulated conditions, suggesting a possible
hyperactivation of Wnt signaling in Fancc2/2 B cells. To verify
Deregulation of Wnt signaling pathway in Fancc2/2 B cells
the activation of Wnt signaling in vivo, we transduced Fancc2/2
We next investigated the molecular mechanism responsible for the and WT (CD45.2) hematopoietic stem and progenitor cells (Lin2
impaired Fancc2/2 B cell-to-ASC differentiation by conducting a Sca1+Ckit+; LSK) with a Wnt GFP reporter lentivirus and trans-
global transcriptome analysis of naive WT and Fancc2/2 B cells planted the transduced LSK cells into lethally irradiated Boy/J
using RNAseq. We found deregulation of 488 genes (224 upreg- mice (CD45.1). Strikingly, we observed a decrease in spleen
ulated and 264 downregulated, moderate t test, FC $ 1.5; cellularity (Fig. 3E) and a profound decrease in B cell number
Supplemental Fig. 2). Pathway analysis showed that among the among donor splenocytes (CD45.2+) of animals transplanted with
top five pathways, the PG and calcium pathways, two known Fancc2/2 LSKs at 4 and 10 wk posttransplant (Fig. 3F). More-
regulators of B cell activation (26) and ASC differentiation (27), over, evaluation of Wnt activity by flow cytometric analysis of
were deregulated in Fancc2/2 B cells (Fig. 4A). Interestingly, we reporter GFP expression showed that WT donor-derived B cells
also observed a significant enrichment of genes involved in Wnt expressed almost no GFP at 10 wk posttransplant (Fig. 3G, 3H),
signaling (Fig. 3A). We decided to evaluate this pathway, as indicating no or weak activity of Wnt pathway. In contrast,
previous studies have suggested a possible interaction of Fancc Fancc2/2 donor-derived B cells exhibited a significant increase in
with Wnt signaling (15, 16) and the involvement of Wnt signaling the proportion of GFP-positive cells (Fig. 3G, 3H), indicating an
in B cell development (28, 29). Upregulation and downregulation increased activation of Wnt signaling. Taken together, these re-

Downloaded from http://www.jimmunol.org/ by guest on July 12, 2017


of both inhibitors (Camk2b, Ankrd6, Sfrp1, Axin2, Nkd1, sults identify deregulation of Wnt signaling as a potential mech-
Tax1bp3) and activators (Dlg2, Fhl2) of the Wnt pathway and anism responsible for the impaired Fancc2/2 B cell differentiation
deregulated expression of Wnt receptor genes (Fzd-9, 2, and 4) and functions.
were detected in Fancc2/2 B cells (Fig. 3B). We confirmed by
quantitative PCR upregulation of Fzd9 and Axin2 and downreg- Wnt signaling inhibits ASC differentiation
ulation of Sfrp1 and Fzd2 in Fancc2/2 B cells (Fig. 3C). To clarify Because our results demonstrated an upregulation of Wnt signaling
the activation state of Wnt signaling, we first evaluated by Western in Fancc2/2 B cells that showed an impaired ASC differentiation,
blot the stabilization of b-catenin in B cells in vitro. Stimulation of we next evaluated the effect of Wnt activation on B cell differ-
B cells with LPS led to the accumulation of b-catenin at 2 and 4 h, entiation into ASCs. We used Wnt3a to activate Wnt signaling in

FIGURE 2. Impaired Fancc2/2 B cell differentiation into plasma cells. (A–D) Naive untouched B cells (CD432 cells) were purified from splenocytes of
6- to 8-wk-old mice and cultured in presence of LPS (20 mg/ml) alone or in combination with IL-4 (5 ng/ml) or IFN-g (50 ng/ml) for 5 d. (A and B) ASCs
(B220dimCD138+) were detected by FACS analysis. (A) Representative FACS plot of ASC detection in cultures of WT or Fancc2/2 B cells. (B) Quan-
tification of ASC percentage as represented in (A) (n = 6/genotype, three independent experiments). (C) The mRNAs from B cell in culture were extracted
for analysis of Blimp1 mRNA expression. Data were normalized on the mean value of WT Blimp1 expression (n = 5/genotype, two independent ex-
periments). (D) IgM production was measured by ELISA in supernatant of 5-d B cell cultures (n = 5/genotypes, three independent experiments). (E–H)
Mice were injected i.v. with 15 mg LPS. (E and F) ASC population (B220dim/2CD138high) was evaluated in spleen of mice by FACS analysis. (E) Rep-
resentative plot of ASC detection in spleen of mice injected with PBS or LPS. (F) Quantification of ASC percentage as represented in (A) (n = 4 mice/
condition, two independent experiments). (G) IgM serum concentration was measured by ELISA at different times after a single i.p. injection of 100 mg
LPS (n = 4 mice/group, two independent experiments). (H) Anti-NP–specific IgG was measured at different times by ELISA in serum of mice immunized
by a single i.p. injection of 100 mg NP-LPS (n = 5 mice/group, two independent experiments). For all graphics, error bar represents mean value 6 SD.
Evaluation of differences between genotype has been conducted using Student t test (*p # 0.05, **p # 0.01, ***p # 0.001).
2990 ASC DIFFERENTIATION DEFECT IN Fancc2/2 MICE

Downloaded from http://www.jimmunol.org/ by guest on July 12, 2017


FIGURE 3. Deregulation of WNT signaling in Fancc2/2 B cells. (A and B) Total RNA was extracted from naive B cells of spleen of WT or Fancc2/2
mice and stimulated 4 h in vitro with 20 mg/ml LPS. The mRNAs were analyzed by RNAseq to determine transcript abundance and differential expression
using GeneSpring GX (FC $ 1.5, p # 0.05). (A) Top five deregulated pathways determined by Genespring Pathway analysis module using the Wiki
Pathway curated database. (B) Heatmap representation (two independent experiments) of mean normalized expression values of WNT signaling deregulated
genes identified by RNAseq. (C) Quantitative PCR validation of candidate WNT deregulated genes from WT and Fancc2/2 B cell culture activated 4 h by
LPS (n = 4 mice/genotype, two independent experiments). (D) Western blot analysis of b-catenin protein level in whole B cell lysate for indicated time of
culture (representative of three independent experiments with pooled B cells of at least two mice for each genotype). Values under the blot represent the
density ratio of b-catenin/b-actin. (E–H) LSK cells were isolated from WT and Fancc2/2 BM by FACS sorting, transduced in vitro with 7TGC reporter
lentivirus, and transplanted into lethally irradiated Boy/J mice. Donor-derived splenocytes (CD45.2+) were analyzed at 4 and 10 wk posttransplantation. (E)
Spleen cellularity of mice at 4 and 10 wk after BM transplantation with Fancc2/2 or WT LSKs. (F) Quantification of proportion of naive donor B cell
percentage at 4 wk (n = 4 mice/genotype, two independent experiments) and 10 wk (n = 5 mice/genotype, two independent experiments) post-
transplantation. (G) Representative overlay histogram of GFP expression from donor-derived naive B cells at 10 wk posttransplantation. (H) Quantification
of GFP+ donor-derived B cells, as indicated in (F) (n = 5 mice/genotype, two independent experiments). For all graphics, error bar represents mean value 6
SD. Evaluation of differences between genotype has been conducted using Student t test (*p # 0.05, **p # 0.01).

WT B cells. As shown in Fig. 4A and 4B, cultures of LPS-stimulated was observed in the B220+Cd1382 and B220dimCd138+ subpop-
WT B cells in presence of Wnt3a resulted in a significant decrease in ulations between LPS-stimulated WT and Fancc2/2 B cells in the
the proportion of CD138+B200dim cells compared with control me- absence of Wnt3a treatment (Fig. 5A, 5B, 5D). This indicates that
dium (Fig. 4A, 4B), indicating an inhibitory effect of Wnt signaling the previously observed decrease in the frequencies of Fancc2/2
on ASC differentiation. Consistent with this result, we observed a B cells and ASCs upon LPS activation was unlikely due to in-
diminution of IgM production (Fig. 4C) and Blimp1 mRNA ex- creased apoptosis of mature undifferentiated B cells. We observed
pression (Fig. 4D) in cells cultured in presence of Wnt3a. Interest- a decrease in apoptosis of WT B cells treated with Wnt3a
ingly, we also observed an upregulation of Pax5 (Fig. 4D), a known (Fig. 5A, 5B), suggesting an antiapoptotic function of Wnt acti-
repressor of Blimp1 (31) and a mediator of Wnt signaling (32). vation during B cell stimulation. Unlike the B220+Cd1382 sub-
These results indicate that Wnt signaling plays an inhibitory effect population, we observed a significant increase in apoptosis in the
on ASC differentiation, possibly through repression of Blimp1. plasmablast and ASC subpopulations in WT and Fancc2/2 B cell
cultures containing Wnt3a (Fig. 5A, 5C, 5D). Significantly, Wnt3a
Fancc2/2 B cells are hypersensitive to Wnt activation during induced a greater augmentation of apoptosis in all three subpop-
ASC differentiation ulations of Fancc2/2 cells than in WT cells (Fig. 5), indicating
We next evaluated the sensitivity of ASC differentiation to the hypersensitivity of Fancc2/2 B cells to Wnt3a. These data dem-
activation of Wnt signaling by analyzing apoptosis in three B cell onstrate differential effects of Wnt activation on apoptosis of
differentiation subpopulations (B220+Cd1382 = non-ASC dif- B cells in these three different differentiation stages and suggest
ferentiated B cells; B220+ Cd138dim/+ = plasmablasts; B220 dim that the inhibitory effect of Wnt3a on ASC differentiation could be
Cd138+ = ASCs). It is noticeable that no difference in apoptosis due in part to an apoptotic mechanism.
The Journal of Immunology 2991

Downloaded from http://www.jimmunol.org/ by guest on July 12, 2017


FIGURE 4. Canonical WNT signaling pathway activation inhibits ASC differentiation in vitro. Naive B cells from spleen of 6- to 8-wk-old WT mice
were isolated and cultured in vitro in the presence of 20 mg/ml LPS with control medium or Wnt3a conditioned medium (25% of culture volume) for 5 d.
(A) Representative FACS plot of B cells in culture labeled with anti–B220-eFluor660, anti–CD138-BV421, and propidium iodide. (B) Quantification of
ASC percentage in culture, as indicated in (A) (n = 6/genotypes, three independent experiments). (C) ELISA evaluation of IgM concentration in B cell
culture supernatants at 6 d of stimulation by LPS with control medium or Wnt3a conditioned medium (n = 5/genotypes, three independent experiments).
(D) Quantitative PCR analysis of Pax5 and Blimp1 mRNA expression from 5-d LPS-stimulated B cells (n = 4, two independent analyses). For all graphics,
error bar represents mean value 6 SD. Evaluation of differences between genotype has been conducted using Student t test (*p # 0.05, **p # 0.01).

Fancc-deficient B cells show a specific defect in IgG2a switch Discussion


Finally, as Fancc is a component of the FA core complex, which In this study, we have investigated the effect of FA deficiency in
is involved in double-strand break repair, and because Ig CSR B cell function using the Fancc mouse model. We have shown that
produces double-strand breaks, we evaluated the potency of loss of Fancc impairs B cell-to-ASC differentiation. We identified
Fancc-deficient B cells to undergo CSR. Fancc2/2 B cells stim- hyperactive Wnt signaling as a potential mechanism responsible
ulated with LPS or LPS plus IL-4 produced a comparable pro- for the impaired Fancc2/2 ASC differentiation. There are several
portion of CSR as WT cells, as evaluated by IgG3 or IgG1 labeling, findings that highlight the significance of FA deficiency in B cell
respectively (Fig. 6A, 6B). However, we observed a significant defects, as follows: first, Fancc 2/2 mice produce significantly
decrease in the percentage of IgG2a-switched Fancc2/2 B cells fewer ASCs and consequently less Abs; second, Fancc loss de-
after stimulation with LPS plus IFN-g (Fig. 6A, 6B). To examine regulates genes essential for B cell activation and ASC differen-
the observed IgG2a switch defect at the molecular level, we tiation; third, hyperactive Wnt signaling is identified as a potential
evaluated the expression of different germline and postswitch mechanism responsible for the impaired Fancc2/2 B cell differ-
IgG isotype mRNA, including Iy3-Cy3 and Iu-Cy3 (IgG3 entiation; fourth, Fancc2/2 B cells exhibit a specific defect in
switch), Iy1-Cy1 and Iu-Cy1 (IgG1 switch), and Iy2a-Cy2a and IgG2a switch; lastly, Fancc2/2 B cells are hypersensitive to Wnt
Iu-Cy2a (IgG2 switch). We also evaluated the expression of the activation during ASC differentiation. These findings identify Wnt
major CSR regulator protein Aicda (25). Because CSR is de- signaling as a physiological regulator of ASC differentiation, es-
pendent on cell division and it is well known that the FA- tablish a role for the Wnt pathway in normal B cell development
deficient lymphocytes have an impaired cell cycle progression and FA immune deficiency, and suggest that modulation of Wnt
(33), we extracted RNA from WT or Fancc2/2 B cells that un- activity could be beneficial for improving FA B cell–dependent im-
derwent three divisions (representative CFSE labeling for Facs munity. In addition, this study extends the understanding of the role
sorting; Supplemental Fig. 3). We observed no difference in the of Wnt signaling in primary B cell differentiation into plasma cells.
expression of IgG germline or postswitch mRNA after LPS (Fig. An important finding of this study is the observation of impaired
6C) and LPS plus IL-4 (Fig. 6D) stimulations, indicating no ASC differentiation in Fancc2/2 B cells. The differentiation of
defect in IgG3 switch or IgG1 switch. In contrast, we observed a B cells to ASC is the main event leading to Ab production. Unlike
profound decrease in the expression of germline and postswitch the specific defect of IgG2a switch CSR observed in Fancc2/2
IgG2a mRNA in the Fancc2/2 B cells compared with WT cells B cells, this deficiency is not specific to the type of costimulatory
stimulated with LPS plus IFN-g (Fig. 6E). Interestingly, we cytokines. This difference is most likely responsible for the
observed an increase in Aicda mRNA expression in Fancc2/2 diminution of IgM production after LPS stimulation and probably
B cells for all three conditions tested (Fig. 6C–E). However, this contributes to the impaired IgG response after immunization of
increase in Aicda expression had no consequence on the CSR, as the mice with NP-LPS. In an effort to understand the mechanism
we observed similar percentages of IgG1- and IgG3-switched underlying this difference, we identified hyperactive Wnt signal-
cells and equal postswitch mRNA levels in both Fancc2/2 and ing in Fancc2/2 B cells. Wnt signaling pathway has been shown to
WT B cells (Fig. 6A–D). Together these data indicate a specific regulate cell differentiation, migration, polarity, and development
IgG2a switch defect in Fancc-deficient B cells and that the im- of organs during embryogenesis (review in 34). Wnt signaling has
paired plasma cell differentiation is not a consequence of a also been implicated in B cell development. In fact, impairment of
general defect in CSR. Wnt signaling in Fzd92/2 mice leads to depletion of pre-B cells in
2992 ASC DIFFERENTIATION DEFECT IN Fancc2/2 MICE

Downloaded from http://www.jimmunol.org/ by guest on July 12, 2017


FIGURE 5. Canonical WNT signaling pathway activation has different effect on apoptosis of WT and Fancc2/2 B cells in culture. Naive B cells were
isolated from spleen of 6- to 8-wk-old WT or Fancc2/2 mice and cultured 5 d in presence of LPS (20 mg/ml) in control or Wnt3a conditioned medium
(25%). Cells in culture were labeled with anti-B220, anti-CD138, anti-annexin V, and PI. (A–D) Apoptosis was determined by FACS analysis (annexin
V+PI+or2 cells) for non-ASC–differentiated B cells (B220+CD1382), plasmablasts (B220+CD138+), and ASCs (B220dimCD138bright). (A) Represen-
tative FACS plot of apoptotic cell percentage in the different cell population after 5 d of B cell culture. (B and C) Apoptotic cell quantification as
presented in (A) among (B) non-ASC–differentiated B cells, (C) plasmablast cells, and (D) ASCs (n = 4 mice/genotype, two independent experiments).
For all graphics, error bar represents mean value 6 SD. Evaluation of differences between genotype has been conducted using Student t test (*p # 0.05,
**p # 0.01).

BM (28). It has also been described that Wnt signaling is im- Pax-5, we found upregulation of Aicda, whose expression is posi-
portant for pro-B cell proliferation and survival and these effects tively regulated by Pax-5 (37), in the cultures of Fancc2/2 and WT
are mediated by Lef-1 (29). The role of Wnt signaling on mature B cells in the presence of Wnt3a (data not shown). Together, we
B cell function is mostly unknown, and, to our knowledge, only propose that the WNT canonical pathway is an inhibitor of ASC
one study reported that knockdown of b-catenin inhibited ASC differentiation and that overactivation of the WNT pathway may
differentiation in vitro (35). Our evaluation of Wnt activity by lead to impaired ASC differentiation seen in Fancc-deficient B cells.
Western blot and in vivo Wnt reporter assay demonstrates an in- It has recently been proposed that the FA pathway plays a role in
crease in constitutive and induced activation of the canonical Wnt the regulation of Wnt signaling. Specifically, two recent studies
signaling in Fancc2/2 B cells. have shown that FANCC is able to interact with Ctbp1 and
To demonstrate that the effect of Wnt signaling on ASC dif- b-catenin and regulates the expression of the Wnt inhibitor Dkk1,
ferentiation is not specific for Fancc2/2 B cells, we used Wnt3a to and that the integrity of the FA core complex is indispensable for a
activate WNT canonical pathway in WT B cells. We observed an normal Wnt activity (15, 16). We observed no alteration in Dkk1
inhibition of ASC differentiation of WT B cells in the culture expression in our transcriptome analysis, but found deregulation
exposed to Wnt3a, recapitulating the phenotype of Fancc2/2 of several genes implicated in the Wnt signaling. Because the
B cells. These studies also enabled us to identify potential medi- previous studies were conducted in human cell line, we cannot
ators of Wnt signaling on ASC differentiation. Specifically, we rule out a difference between human and mouse and/or between
found that activation of Wnt signaling by Wnt3a led to decreased cell types. Nevertheless, our data demonstrate that, in mouse
expression of Blimp-1 and IgM secretion in LPS-stimulated primary cells, disruption of Fancc could deregulate Wnt signaling.
B cells. Interestingly, we also observed an upregulation of the In addition, because oxidative DNA damage is able to increase
expression of Pax5, which has been described as a mediator of Wnt activity by ATM activation (38), and because FA-deficient
Wnt signaling (32) and as an inhibitor of Blimp-1 (3). It is pos- cells have an increased intracellular level of reactive oxygen
sible that hyperactive Wnt signaling in Fancc2/2 B cells leads to species (39), it would be interesting to evaluate the implication of
an increased activity of Pax5, which in turn represses Blimp1 reactive oxygen species in hyperactive Wnt signaling observed in
transcription. As Blimp-1 represses Pax5 transcription (36), the Fancc2/2 B cells.
observed upregulation of Pax-5 could be the result of Blimp1 Our study also points to other defects in Fancc2/2 B cells. Our
repression. Moreover, in accordance with an increased activity of in vitro and in vivo B cell differentiation experiments indicate an
The Journal of Immunology 2993

Downloaded from http://www.jimmunol.org/ by guest on July 12, 2017


FIGURE 6. Specific IgG2a class switch impairment in Fancc2/2 B cells. (A–E) Naive untouched B cells (CD432 cells) were purified from splenocytes of
6- to 8-wk-old mice and cultured 3 d in presence of LPS (20 mg/ml) alone or in combination with IL-4 (5 ng/ml) or IFN-g (50 ng/ml). (A and B) Isotype-
specific CSR was evaluated for Fancc2/2 and WT B cells by FACS analysis. (A) Representative FACS plot of percentage of B cells that underwent CSR
(switched). (B) Quantification of switched B cells as represented in (A) (n = 5 mice/group, three independent experiments). (C–E) Quantitative PCR analysis
of mRNA expression of Aicda, isotype-specific IgG germline (ly3-Cy3, ly1-Cy1, and ly2a-Cy2a), or postswitched (lm-Cy3, -Cy1, or Cy-2a) from 3 division
peak (CFSE)-sorted B cells (n = 4/genotype, two independent experiments). For all graphics, error bar represents mean value 6 SD. Evaluation of dif-
ferences between genotype has been conducted using Student t test (**p # 0.01).

augmentation of the pre/pro-B cell population and a diminution of possible explanation for the impairment of germline IgG2a mRNA
the more differentiated B cells in the BM of Fancc2/2 mice. expression could be related to the deficiency in IFN-g signaling
However, no difference was observed in the periphery of Fancc2/2 that has been previously reported for Fancc deficiency (13, 14). In
mice compared with WT controls, indicating that in steady state, fact, the downstream effector of IFN-g signaling T-bet and a
the hematopoietic system might be able to compensate probably proper activation of Stat1 are required for an efficient IgG2a
by increasing the B lineage populations in the BM. At the pre- and germline mRNA expression (42, 43). The IgG2a switch deficiency
pro-B cell stages of B cell differentiation, the cells are highly most likely contributes to the decrease in immunization response
expanding and are going through rearrangement at the locus of the observed in Fancc 2/2 mice, as previously reported (17) and
Ig H and L chain (1). It is possible that this high rate of prolif- shown in this study after NP-LPS immunization. TLR4 activation
eration in combination with genomic DNA breakage and rear- is known to stimulate the Th1 immune response (44), which is
rangement causes a replicative stress in Fancc2/2 B cells in which dependent on IFN-g signaling and secretion. Moreover, as Th1
the FA repair pathway is deficient. We also note that other and IFN-g are important mediators of intracellular pathogen im-
mechanisms, such as changes in B cell migration/sequestration, munity, it would be important to evaluate this deficiency in FA
may also be responsible for the B cell defects in the Fancc2/2 patients. In effect, it has been reported that a subgroup of FA
BM. However, our results with transplantation seem to indicate an patients is more sensitive to infections (17) and has an impaired
impairment of B cell differentiation when the hematopoietic immunization response (19). Therefore, the choice of vaccine
system is under stress and might not be able to compensate the formulation without TLR4 adjuvant might be beneficial for FA
B cell compartment without a functional FA pathway. This B cell patients.
differentiation defect is in accordance with a previous report
showing a diminution of B cell number in a subgroup of FA pa- Acknowledgments
tients (24). We thank Dr. Maxime Mahe from Dr. Michael Helmrath’s laboratory at the
The similarity in periphery of WT and Fancc2/2 mice allowed Cincinnati Children’s Hospital Medical Center Pediatric Surgery Depart-
us to evaluate the CSR efficiency in B cells without bias. CSR ment for providing the Wnt3a conditioned medium; Dr. Damien Renaud
from the Cincinnati Children’s Hospital Medical Center Experimental
involves the NHEJ DNA repair pathway (1), and the FA pathway
Hematology Department for providing Abs for BM B cell lineage exper-
has been proposed to inhibit the NHEJ in favor of the homologous iments; Danielle Davis for reviewing the manuscript; the Cincinnati Child-
recombination (9, 10). As previously reported for Fanca (40)- and ren’s Hospital Medical Center DNA Sequencing and Genotyping Core for
Fancg (41)-deficient mice, we did not observe a general impair- mRNA sequencing; the Cincinnati Children’s Hospital Medical Center
ment of CSR in Fancc2/2 B cells. In contrast, we observed a Research Cytometry Core for cell sorting; the Cincinnati Children’s Hos-
specific defect of IgG2a CSR in Fancc2/2 B cells. However, this pital Medical Center Viral Vector Core for lentivirus production; and the
is unlikely due to the deficiency in DNA recombination process, as Cincinnati Children’s Hospital Medical Center Comprehensive Mouse and
the expression of specific Ig germline sequence is required to Cancer Core for the mouse transplantation experiments.
initiate CSR, and we observed a profound decrease in the germline
expression of IgG2a. This result indicates that an impairment of Disclosures
CSR occurs before initiating the DNA recombination process. A The authors have no financial conflicts of interest.
2994 ASC DIFFERENTIATION DEFECT IN Fancc2/2 MICE

References defects and hematopoietic hypersensitivity to gamma-interferon in mice with a


targeted disruption of the Fanconi anemia C gene. Blood 88: 49–58.
1. Alt, F. W., Y. Zhang, F. L. Meng, C. Guo, and B. Schwer. 2013. Mechanisms of 23. Fuerer, C., and R. Nusse. 2010. Lentiviral vectors to probe and manipulate the
programmed DNA lesions and genomic instability in the immune system. Cell Wnt signaling pathway. PLoS One 5: e9370.
152: 417–429. 24. Myers, K. C., J. J. Bleesing, S. M. Davies, X. Zhang, L. J. Martin, R. Mueller,
2. Shapiro-Shelef, M., and K. Calame. 2005. Regulation of plasma-cell develop- R. E. Harris, A. H. Filipovich, M. B. Kovacic, S. I. Wells, and P. A. Mehta. 2011.
ment. Nat. Rev. Immunol. 5: 230–242. Impaired immune function in children with Fanconi anaemia. Br. J. Haematol.
3. Usui, T., Y. Wakatsuki, Y. Matsunaga, S. Kaneko, H. Koseki, and T. Kita. 1997. 154: 234–240.
Overexpression of B cell-specific activator protein (BSAP/Pax-5) in a late B cell 25. Pieper, K., B. Grimbacher, and H. Eibel. 2013. B-cell biology and development.
is sufficient to suppress differentiation to an Ig high producer cell with plasma J. Allergy Clin. Immunol. 131: 959–971.
cell phenotype. J. Immunol. 158: 3197–3204. 26. Scharenberg, A. M., L. A. Humphries, and D. J. Rawlings. 2007. Calcium sig-
4. Lin, Y., K. Wong, and K. Calame. 1997. Repression of c-myc transcription by nalling and cell-fate choice in B cells. Nat. Rev. Immunol. 7: 778–789.
Blimp-1, an inducer of terminal B cell differentiation. Science 276: 596–599. 27. Lydyard, P. M., J. Brostoff, B. N. Hudspith, and H. Parry. 1982. Prostaglandin
5. Shaffer, A. L., M. Shapiro-Shelef, N. N. Iwakoshi, A. H. Lee, S. B. Qian, E2-mediated enhancement of human plasma cell differentiation. Immunol. Lett.
H. Zhao, X. Yu, L. Yang, B. K. Tan, A. Rosenwald, et al. 2004. XBP1, down- 4: 113–116.
stream of Blimp-1, expands the secretory apparatus and other organelles, and 28. Ranheim, E. A., H. C. Kwan, T. Reya, Y. K. Wang, I. L. Weissman, and
increases protein synthesis in plasma cell differentiation. Immunity 21: 81–93. U. Francke. 2005. Frizzled 9 knock-out mice have abnormal B-cell development.
6. Oracki, S. A., J. A. Walker, M. L. Hibbs, L. M. Corcoran, and D. M. Tarlinton. Blood 105: 2487–2494.
2010. Plasma cell development and survival. Immunol. Rev. 237: 140–159. 29. Reya, T., M. O’Riordan, R. Okamura, E. Devaney, K. Willert, R. Nusse, and
7. Dong, H., D. W. Nebert, E. A. Bruford, D. C. Thompson, H. Joenje, and R. Grosschedl. 2000. Wnt signaling regulates B lymphocyte proliferation
V. Vasiliou. 2015. Update of the human and mouse Fanconi anemia genes. Hum. through a LEF-1 dependent mechanism. Immunity 13: 15–24.
Genomics 9: 32. 30. Christian, S. L., P. V. Sims, and M. R. Gold. 2002. The B cell antigen receptor
8. Kee, Y., and A. D. D’Andrea. 2012. Molecular pathogenesis and clinical man- regulates the transcriptional activator beta-catenin via protein kinase C-mediated
agement of Fanconi anemia. J. Clin. Invest. 122: 3799–3806. inhibition of glycogen synthase kinase-3. J. Immunol. 169: 758–769.
9. Adamo, A., S. J. Collis, C. A. Adelman, N. Silva, Z. Horejsi, J. D. Ward, 31. Shapiro-Shelef, M., K. I. Lin, D. Savitsky, J. Liao, and K. Calame. 2005. Blimp-
E. Martinez-Perez, S. J. Boulton, and A. La Volpe. 2010. Preventing nonho- 1 is required for maintenance of long-lived plasma cells in the bone marrow. J.
mologous end joining suppresses DNA repair defects of Fanconi anemia. Mol. Exp. Med. 202: 1471–1476.

Downloaded from http://www.jimmunol.org/ by guest on July 12, 2017


Cell 39: 25–35. 32. Machon, O., M. Backman, O. Machonova, Z. Kozmik, T. Vacik, L. Andersen,
10. Pace, P., G. Mosedale, M. R. Hodskinson, I. V. Rosado, M. Sivasubramaniam, and S. Krauss. 2007. A dynamic gradient of Wnt signaling controls initiation of
and K. J. Patel. 2010. Ku70 corrupts DNA repair in the absence of the Fanconi neurogenesis in the mammalian cortex and cellular specification in the hippo-
anemia pathway. Science 329: 219–223. campus. Dev. Biol. 311: 223–237.
11. Pang, Q., W. Keeble, T. A. Christianson, G. R. Faulkner, and G. C. Bagby. 2001. 33. Dutrillaux, B., A. Aurias, A. M. Dutrillaux, D. Buriot, and M. Prieur. 1982. The
FANCC interacts with Hsp70 to protect hematopoietic cells from IFN-gamma/ cell cycle of lymphocytes in Fanconi anemia. Hum. Genet. 62: 327–332.
TNF-alpha-mediated cytotoxicity. EMBO J. 20: 4478–4489. 34. Grigoryan, T., P. Wend, A. Klaus, and W. Birchmeier. 2008. Deciphering the
12. Pang, Q., T. A. Christianson, W. Keeble, T. Koretsky, and G. C. Bagby. 2002. function of canonical Wnt signals in development and disease: conditional loss-
The anti-apoptotic function of Hsp70 in the interferon-inducible double-stranded and gain-of-function mutations of beta-catenin in mice. Genes Dev. 22: 2308–
RNA-dependent protein kinase-mediated death signaling pathway requires the 2341.
Fanconi anemia protein, FANCC. J. Biol. Chem. 277: 49638–49643. 35. Yu, Q., W. J. Quinn, III, T. Salay, J. E. Crowley, M. P. Cancro, and J. M. Sen.
13. Pang, Q., S. Fagerlie, T. A. Christianson, W. Keeble, G. Faulkner, J. Diaz, 2008. Role of beta-catenin in B cell development and function. J. Immunol. 181:
R. K. Rathbun, and G. C. Bagby. 2000. The Fanconi anemia protein FANCC 3777–3783.
binds to and facilitates the activation of STAT1 by gamma interferon and he- 36. Lin, K. I., C. Angelin-Duclos, T. C. Kuo, and K. Calame. 2002. Blimp-1-
matopoietic growth factors. Mol. Cell. Biol. 20: 4724–4735. dependent repression of Pax-5 is required for differentiation of B cells to im-
14. Fagerlie, S. R., T. Koretsky, B. Torok-Storb, and G. C. Bagby. 2004. Impaired munoglobulin M-secreting plasma cells. Mol. Cell. Biol. 22: 4771–4780.
type I IFN-induced Jak/STAT signaling in FA-C cells and abnormal CD4+ Th 37. Alinikula, J., K. P. Nera, S. Junttila, and O. Lassila. 2011. Alternate pathways for
cell subsets in Fancc-/- mice. J. Immunol. 173: 3863–3870. Bcl6-mediated regulation of B cell to plasma cell differentiation. Eur. J.
15. Huard, C. C., C. S. Tremblay, K. Helsper, M. C. Delisle, D. Schindler, G. Lévesque, Immunol. 41: 2404–2413.
and M. Carreau. 2013. Fanconi anemia proteins interact with CtBP1 and modulate 38. Svegliati, S., G. Marrone, A. Pezone, T. Spadoni, A. Grieco, G. Moroncini,
the expression of the Wnt antagonist Dickkopf-1. Blood 121: 1729–1739. D. Grieco, M. Vinciguerra, S. Agnese, A. J€ungel, et al. 2014. Oxidative DNA
16. Huard, C. C., C. S. Tremblay, A. Magron, G. Lévesque, and M. Carreau. 2014. damage induces the ATM-mediated transcriptional suppression of the Wnt in-
The Fanconi anemia pathway has a dual function in Dickkopf-1 transcriptional hibitor WIF-1 in systemic sclerosis and fibrosis. Sci. Signal. 7: ra84.
repression. Proc. Natl. Acad. Sci. USA 111: 2152–2157. 39. Zhang, X., D. P. Sejas, Y. Qiu, D. A. Williams, and Q. Pang. 2007. Inflammatory
17. Fagerlie, S. R., and G. C. Bagby. 2006. Immune defects in Fanconi anemia. Crit. ROS promote and cooperate with the Fanconi anemia mutation for hematopoi-
Rev. Immunol. 26: 81–96. etic senescence. J. Cell Sci. 120: 1572–1583.
18. Liu, Y., K. Ballman, D. Li, S. Khan, E. Derr-Yellin, W. Shou, and L. S. Haneline. 40. Nguyen, T. V., L. Riou, S. Aoufouchi, and F. Rosselli. 2014. Fanca deficiency
2012. Impaired function of Fanconi anemia type C-deficient macrophages. J. reduces A/T transitions in somatic hypermutation and alters class switch re-
Leukoc. Biol. 91: 333–340. combination junctions in mouse B cells. J. Exp. Med. 211: 1011–1018.
19. Roxo, P., Jr., L. K. Arruda, A. T. Nagao, M. M. Carneiro-Sampaio, and 41. Krijger, P. H., N. Wit, P. C. van den Berk, and H. Jacobs. 2010. The Fanconi
V. P. Ferriani. 2001. Allergic and immunologic parameters in patients with anemia core complex is dispensable during somatic hypermutation and class
Fanconi’s anemia. Int. Arch. Allergy Immunol. 125: 349–355. switch recombination. PLoS One 5: e15236.
20. Alter, B. P., N. Giri, Y. Pan, S. A. Savage, and L. A. Pinto. 2014. Antibody 42. Gerth, A. J., L. Lin, and S. L. Peng. 2003. T-bet regulates T-independent IgG2a
response to human papillomavirus vaccine in subjects with inherited bone class switching. Int. Immunol. 15: 937–944.
marrow failure syndromes. Vaccine 32: 1169–1173. 43. Xu, W., and J. J. Zhang. 2005. Stat1-dependent synergistic activation of T-bet for
21. Holmgren, S. C., E. M. Goren, B. L. Wood, P. S. Becker, and J. A. Taylor. 2012. IgG2a production during early stage of B cell activation. J. Immunol. 175: 7419–
Immune defects in a mouse model of Fanconi anaemia. Br. J. Haematol. 159: 7424.
246–250. 44. Netea, M. G., J. W. Van der Meer, R. P. Sutmuller, G. J. Adema, and
22. Whitney, M. A., G. Royle, M. J. Low, M. A. Kelly, M. K. Axthelm, C. Reifsteck, B. J. Kullberg. 2005. From the Th1/Th2 paradigm towards a Toll-like receptor/T-
S. Olson, R. E. Braun, M. C. Heinrich, R. K. Rathbun, et al. 1996. Germ cell helper bias. Antimicrob. Agents Chemother. 49: 3991–3996.
Primers 5'-->3'
Target mRNA Forward Reverse
Iu-C3 ACCTGGGAATGTATGGTTGTGGCTT AGCCAGGGACCAAGGGATAGAC
Iu-Cy1 CCAGGCACCGCAAATGCC GGACAGTCACTGAGCTGCTC
Iu-Cy2a ACCTGGGAATGTATGGTTGTGGCTT GCTGGGCCAGGTGCTCGAGGTT
Iy3-Cy3 GAGGTGGCCAGAGGAGCAAGAT AGCCAGGGACCAAGGGATAGAC
Iy1-Cy1 TCGAGAAGCCTGAGGAATGTG GGATCCAGAGTTCCAGGTCACT

Iy2a-Cy2a GCTGATGTACCTACCGAGAGA GCTGGGCCAGGTGCTCGAGGTT


Iy3-Cy3 GAGGTGGCCAGAGGAGCAAGAT AGCCAGGGACCAAGGGATAGAC
Aicda GCCACCTTCGCAACAAGTCT CCGGGCACAGTCATAGCAC
Blimp1 TGACTTTGTGGACAGAGGCCGAGT CTGTTGTTGGCAGCATACTTGAAA

Pax5 CCATCAGGACAGGACATGGAG GGCAAGTTCCACTATCCTTTGG


Fzd9 CCAGTACGTGGAGAAGAGTC GTGGAGAAGAAACACAACGC
Axin2 TGACTCTCCTTCCAGATCCCA TGCCCACACTAGGCTGACA
Sfrp1 CAACGTGGGCTACAAGAAGAT GGCCAGTAGAAGCCGAAGAAC

Fzd2 ATCTTTCTGTCCGGCTGCTA GATGGCTAGGCTCTTGCAGT


Hprt1 CGTCGTGATTAGCGATGATG ACAGAGGGCCACAATGTGAT-
Actb GGCTGTATTCCCCTCCATCG CCAGTTGGTAACAATGCCATGT

Supplemental Table 1: Primer sequences used for RT-Q-PCR.


Supplemental Figure 1: Normality of B cells numbers and B cells populations in
periphery in Fancc-/- mice (A) Measurement of B cells number in tail vein blood of mice (n=6
mice/group). Data represented as percentage of mean WT B cells blood count. (B-G) Spleens
were harvested from 6-8 weeks old mice and splenocytes were collected after centrifugation on
Ficoll and labelled with anti-B220-eFluor660, anti-IgM-Fitc, anti-IgD-Pe anti-CD23-PeCy7 and
anti-CD121-EFLUOR450 Abs. Before Facs acquisition, PI was added to the cells to the tubes to
detect and exclude dead cells (n=5 mice/group). (B) Spleen cellularity of Fancc-/- and WT mice
(n=6). (C) Representative Facs histogram of B cells (B220+ cells) percentage into live
splenocytes of WT or Fancc-/- mice. (D) Quantification of B cells percentage as shown in (C). (E)
Representative Facs Plot of B cells subpopulation evaluation among live B220+ splenocytes.
Left panel = Mature B cells (IgMdimIgD+), T1+Marginal zone (MZ) B cells (IgDdim/-IgM+-) and T2 B
cells (IgD+IgM+). Right panel = MZ B cells (CD21+CD23-) and follicular B cells (CD21+/dimCD23+).
(F-G) Quantification of B cells population represented in (E), (F) left panel and (G) right panel.
For all graphics, Bar represents mean value ± SD. Evaluation of differences between genotype
has been conducted using Student’s T-test.
Supplemental Figure 2: RNAseq evaluation of deregulated genes in Fancc-/- mice B cells.
The mRNA expressions were filtered to keep genes with a raw expression value >1 in 50% of
samples. For the remaining genes, fold changed was calculated in GeneSpring GX and
selected with a 1.5 cut-off and moderate T-test analysis was done to detect significantly
deregulated genes. Heat map representation of total deregulated genes in 2 independent
experiments (normalized expression value).
Supplemental Figure 3: Cell divisions analysis and sorting strategy by CFSE staining. (A-
C) Naïve untouched B cells (CD43- cells) were purified from splenocytes of 6-8 weeks old mice
loaded with 1.5µM CFSE and then cultured for 3 days. Cell in 3 division peaks were sorted by
FACS for qPCR analysis (Figure 2) (A) Representative histogram detection of CFSE
fluorescence form LPS stimulated B cells in presence of IFN-γ or IL-4. (B-C) Quantification of
cell division as indicated in (A) for LPS stimulated B cells in presence of (B) IFN- γ or (C) IL-4.

Das könnte Ihnen auch gefallen