Sie sind auf Seite 1von 29

Hum Genet (2012) 131:535563

DOI 10.1007/s00439-011-1119-1

REVIEW PAPER

Modeling human neurodegenerative diseases


in transgenic systems
Miguel A. Gama Sosa Rita De Gasperi
Gregory A. Elder

Received: 13 October 2011 / Accepted: 23 November 2011 / Published online: 14 December 2011
Springer-Verlag (outside the USA) 2011

Abstract Transgenic systems are widely used to study


the cellular and molecular basis of human neurodegenerative diseases. A wide variety of model organisms have been
utilized, including bacteria (Escherichia coli), plants (Arabidopsis thaliana), nematodes (Caenorhabditis elegans),
arthropods (Drosophila melanogaster), Wsh (zebraWsh,
Danio rerio), rodents (mouse, Mus musculus and rat, Rattus
norvegicus) as well as non-human primates (rhesus monkey, Macaca mulatta). These transgenic systems have enormous value for understanding the pathophysiological basis
of these disorders and have, in some cases, been instrumental in the development of therapeutic approaches to treat
these conditions. In this review, we discuss the most commonly used model organisms and the methodologies available for the preparation of transgenic organisms. Moreover,
we provide selected examples of the use of these technolo-

M. A. Gama Sosa (&) R. De Gasperi


Research and Development Service, 3F-20, James J. Peters
Department of Veterans AVairs Medical Center,
130 West Kingsbridge Road, Bronx, NY 10468, USA
e-mail: miguel.gama-sosa@mssm.edu
M. A. Gama Sosa R. De Gasperi G. A. Elder
Department of Psychiatry, Mount Sinai School of Medicine
of New York University, New York, NY, USA
M. A. Gama Sosa R. De Gasperi G. A. Elder
The Friedman Brain Institute, Mount Sinai School of Medicine
of New York University, New York, NY, USA
G. A. Elder
Neurology Service, James J. Peters Department of Veterans
AVairs Medical Center, Bronx, NY, USA
G. A. Elder
Department of Neurology, Mount Sinai School of Medicine
of New York University, New York, NY, USA

gies for the preparation of transgenic animal models of neurodegenerative diseases, including Alzheimers disease
(AD), frontotemporal lobar degeneration (FTLD), amyotrophic lateral sclerosis (ALS), Huntingtons disease (HD) and
Parkinsons disease (PD) and discuss the application of
these technologies to AD as an example of how transgenic
modeling has aVected the study of human neurodegenerative diseases.

Introduction
Inherited neurodegenerative diseases are a heterogeneous
group of genetic disorders characterized by loss of neuronal
structure and function, and are generally accompanied by
neuronal loss. These diseases may result directly from
degeneration of particular neuronal populations or indirectly from alterations in glial support cells. The particular
topological pattern of brain involvement determines the
speciWc clinical manifestations of each disease. Several of
these diseases are characterized by an accumulation of
abnormal or aggregated proteins or other biological materials either extracellularly or within neurons. These accumulations take various forms and result in the neuritic plaques
or neuroWbrillary tangles in Alzheimers disease (AD),
Lewy bodies in Parkinsons disease (PD), glycogen and
polyglucosan bodies in Lafora disease, or glycolipids and
complex carbohydrates in the lysosomal storage diseases.
As with other genetic disorders, once a pattern of inheritance has been identiWed, the chromosomal loci can be
determined by linkage studies. Sequencing of putative loci
within members of an established pedigree can be used to
identify the speciWc genetic changes associated with the
disease. This process has been greatly facilitated by recent
advances in DNA sequencing technology (Mardis 2011),

123

536

which now allow the sequencing of more than 600 gigabases per run at dramatically reduced costs ($1,000 USD
per genome). Once a causative mutation has been identiWed, transgenic systems can be generated to model the
human disease or aspects of the altered gene function.
Transgenic models provide further conWrmation of the
genetic basis for the disease and aid in identiWcation of cellular and molecular mechanisms responsible for disease
phenotypes. They also provide models in which the eYcacy
and side eVects of potential treatments can be evaluated.
Various transgenic systems of bacterial, plant, and animal
origin are currently used to model diVerent aspects of
human inherited neurodegenerative diseases. These systems vary in terms of ease of manipulation and phylogenetic relatedness to humans, but have all been useful for the
study of neurodegenerative diseases.

Bacterial transgenic systems


Because of their relatively simple genetics and the straightforward methodologies for introducing genetic modiWcations, bacteria were the Wrst organisms to be genetically
altered in the laboratory. Although bacteria are far removed
from humans phylogenetically, bacterial systems have been
useful for modeling particular aspects of human neurodegenerative conditions. One particular example is the production of pathogenic prion proteins (PrP), where attributes
of pathogenic PrP were induced in Escherichia coli
(E. coli) expressing recombinant murine PrP. The PrP generated was protease resistant and aggregated into a self-perpetuating state that induced classic prion pathology when
inoculated into brains of mice (Wang et al. 2010). Similarly, a yeast prion protein was converted into an infectious
prion protein in E. coli (Garrity et al. 2010). These systems
provide unique experimental systems for studying the spontaneous conversion of normal to pathogenic PrP and also
provide direct evidence supporting the prion hypothesis.

Plant transgenic systems


Plant somatic cells are totipotent, and can regenerate from
single cells grown in vitro. Therefore, genetic modiWcations
made in somatic cells develop into adult plants in which all
cells contain the desired alteration. DNA can be introduced
into plant cells by a variety of techniques, including treatment with polyethylene glycol (PEG), electroporation,
microinjection, biolistic transformation, and the use of
Agrobacterium, plant viruses and liposomes as DNA carriers (Ziemienowicz 2010).
Although multicellular, plants are only distantly related
to humans. Nonetheless, several insights have been derived

123

Hum Genet (2012) 131:535563

from plant studies in which genes related to human neurodegeneration have been examined. Indeed, an intersection
between the Welds of neuroscience and plant starch metabolism helped reveal the molecular defects involved in Lafora
disease (Gentry et al. 2007, 2009; Niittyla et al. 2006).
Lafora disease is an autosomal recessive disorder that produces a syndrome of progressive myoclonic epilepsy and is
pathologically characterized by accumulation of insoluble
glucan (Lafora bodies). Based on human studies, two genes
were known to be involved in the disease: EMP2A, a gene
coding for the phosphatase laforin, which is known to contain a canonical dual speciWcity phosphatase (DSP) and a
carbohydrate binding module (CBM), and EMP2B, a gene
coding for malin, which functions as an E3 ubiquitin ligase
that binds, ubiquitinates, and promotes degradation of laforin. Niittyla et al. discovered a gene in plants (Arabidopsis
thaliana) that they called starch excess 4 (SEX4), which, as
with laforin, contained DSP and CBM domains. Strikingly,
in plants, mutations in SEX4 resulted in an increase in
insoluble glucans similar to those found in Lafora disease
patients. Using A. thaliana transgenic plants, SEX4 was
shown to have the same biochemical properties as laforin
(glucan binding and phosphatase activity) and laforin was
found to be a functional equivalent of SEX4, as human
laforin could rescue the phenotype in SEX4 mutated plants
(Gentry et al. 2007, 2009; Vander Kooi et al. 2010).
Transgenic plants have also been used to study the function of genes associated with familial Alzheimers disease
(FAD) and trinucleotide repeat disorders. The presenilins
(presenilin 1 and 2) are transmembrane proteins that, when
mutated, are the most common genetic cause of FAD. Presenilins form part of the -secretase complex, which cleaves
a range of substrate proteins in animals, including notch and
the amyloid precursor protein (APP). However, presenilins
have long been suspected to have -secretase-independent
functions. Interestingly, plants produce presenilin (psn)
despite lacking homologs for notch or APP. Transgenic systems using the moss bryophyta Physcomitrella patens have
been used to investigate the -secretase-dependent and -independent functions of presenilin in a setting where there is no
interference from eVects of presenilins on notch or APP processing. Interestingly, using a catalytically inactive psn
mutant, a -secretase-independent function of P. patents
presenilin was found to be involved in proper cytoskeletal
function, including organelle transport and cell polarity, and
human presenilin 1 rescued abnormal growth and light
responses in P. patents knock-out plants (Pppsn) (Khandelwal et al. 2007). Furthermore, P. patents psn was able to
revert cell proliferation abnormalities in mouse embryonic
Wbroblasts. Recently genetic defects in plants caused by triplet repeat expansions (TTC/GAA) in the isopropyl malate
isomerase large subunit 1 gene have been studied in wild
type and transgenic Arabidopsis thaliana (Sureshkumar et al.

Hum Genet (2012) 131:535563

2009). As in humans, the triplet GAA repeat expansions


cause a reduction in gene activity that may severely impair
strain growth and development.

Animal transgenic systems


Despite the successes in bacteria and plants, transgenic
modeling of human neurodegenerative disorders has been
primarily conducted in animals. Indeed, the goal of reproducing a phenotype akin to the broader clinical manifestations of the human disorders has only been achieved in
animals. Animal systems allow for testing of potential therapeutic strategies and investigation of disease progression
in a manner not possible in humans. Transgenic models of
neurodegenerative disorders have been produced in a variety of invertebrate and vertebrate species.
Among invertebrate systems the arthropod fruit Xy Drosophila melanogaster and the nematode worm Caenorhabditis
elegans have been the most widely used (Jeibmann and Paulus
2009; Link 2001; Lu and Vogel 2009; Thompson and Marsh
2003). One central appeal of invertebrate models is the power
of the genetic tools available, including the ability to perform
forward genetic screens to identify genes that modulate disease development. Furthermore, these models enable the rapid
production and analysis of transgenic lines expressing variants
of disease-associated proteins (Link 2001). The major disadvantage of organisms like D. melanogaster or C. elegans is
that they lack factors, such as immune function and myelination that are critical components in many vertebrate pathological processes. Anatomic divergence may also preclude the
direct modeling of some processes in invertebrate models
(Jeibmann and Paulus 2009; Link 2001).
Caenorhabditis elegans The nematode C. elegans is a
transparent roundworm. One of its great appeals as a model
system is its simplicity (Muller and Grossniklaus 2010).
C. elegans contains fewer than 1,000 cells that divide in a
stereotypical manner in which the lineage of every cell can
be traced back to the egg. The worms are easy to grow
and display all the hallmarks of a multicellular organism,
including a complex organ structure and social, sexual, and
learning behaviors. Adult C. elegans hermaphrodites contain only 302 neurons, and the complete pattern of synaptic
connections has been reconstructed by serial electron
microscopy. Neuronal classes include chemosensory,
mechanosensory, and thermosensory types. Their identities
are known with such precision that seventy-Wve motor neurons are known to innervate the muscles of the body wall
(excluding the head) and of these, 56 are cholinergic and 19
are GABAergic. C. elegans larvae contain four serotonergic and eight dopaminergic neurons. Many of the proteins
involved in formation, traYcking, and release of synaptic
vesicles are highly conserved with those found in mam-

537

mals. Because the organism is transparent throughout its


life cycle, fusion proteins containing green Xuorescent protein (GFP) have been extensively used to visualize speciWc
neurons and synapses in vivo. In some instances, neuronal
death has been directly observed in living worms by the
appearance of vacuolated neurons. More generally, GFP
tagging of speciWc neuronal populations allows degeneration and neuronal cell loss to be observed throughout life
(Teschendorf and Link 2009).
Human disease-associated proteins can be expressed in
C. elegans using transgenic cassettes employing a C. elegansspeciWc promoter and transgenic cDNA. Targeted neuronal
expression can be accomplished using pan-neuronal promoters (including unc-119, snb-1, aex-3, rgef-1) or speciWc
neuronal subtype promoters (such as mec-7, mechanosensory neurons; osm-1, chemosensory neurons; dat-1, dopaminergic neurons) (Teschendorf and Link 2009). Body wall
muscle-speciWc promoters, such as unc-54 and myo-3, also
exist. These promoters often produce representative expression patterns; although, important exceptions have been
observed. Also, vector-free DNA constructs appear to
improve transgene expression (Etchberger and Hobert 2008).
Moreover, transgenic organisms have been generated by
recombining large fosmids with the galK selectable marker
(Zhang et al. 2008, 2011b). DNA is injected into either
maturing oocytes or, more commonly, into the syncytial
gonad, which targets the hermaphrodite germline. The coinjection of a selectable marker, such as rol-6, which alters
cell morphology, or GFP allows easy identiWcation of transgenic organisms. More recently, a selection system using
puromycin has been developed for rapid isolation of transgenic worms (Semple et al. 2010).
Comprehensive databases of C. elegans promoters that
are active in the nervous system can be found at http://chinook.uoregon.edu/promoters.html and http://www.wormbase.org. A detailed description of genetic screening in this
organism has also been described (Jorgensen and Mango
2002). To capture genuine spatial and temporal expression
patterns, investigators have modiWed large segments of
DNA in bacterial artiWcial chromosomes (BACs) or fosmids that have then been used to generate transgenic lines
of C. elegans (Zhang et al. 2008, 2011b; Dolphin and Hope
2006; Sarov et al. 2006). Using locus-speciWc zinc-Wnger
nucleases (ZFN), speciWc knockouts for the ben-1 and rex-1
genes have been created in C. elegans (Reyon et al. 2011;
Wood et al. 2011). Furthermore, the FLP-FRT conditional
recombination system has been used for gene activation in
this organism (Davis et al. 2008).
Drosophila melanogaster The fruit Xy has been widely
used to model both normal biological functions of genes
linked to neurodegenerative diseases and to determine
mechanisms by which mutations lead to neuronal dysfunction and cell death (Bonini and Fortini 2003; Muqit and

123

538

Hum Genet (2012) 131:535563

Fig. 1 Basic Gal4/UAS System. The Saccharomyces cerevisiae transactivator Gal4, produced from a driver regulatory cassette, binds as a
homodimer to a 17-bp (CGG-N11-CCG) upstream activating sequence
(UAS) on the target transgene cassette. Gal4 homodimers activate the

target transgene by binding to four tandemly repeated UAS sequences


(UAS4) placed upstream of a minimal promoter (TATA) (Lewandoski
2001)

Feany 2002). Drosophila develops through embryo, larval,


pupal, and adult stages. The adult Xy brain and the complex
eye have been extensively used to study neurodegenerative
diseases (Jeibmann and Paulus 2009).
The embryonic CNS in Drosophila contains both neurons and glial cells. Neurons build commissures in close
association with midline glial cells. CNS glial cells can be
classiWed as midline or lateral glia, and surface glial cells
form a continuous covering of the CNS and peripheral
nerves. Drosophila glia can be further divided into diVerent
classes, including peripheral and exit glial cells, subperineural glial cells which enclose the CNS, and channel glial
cells which line the channels in the ventral nerve cord. The
larval brain is composed of two hemispheres and the subesophageal ganglion. In the adult, association areas necessary for olfactory learning and memory (mushroom bodies)
contain about 2,500 neurons that have small densely packed
cell bodies known as Kenyon cells. Neurons in the Xy
visual system develop in a stereotypical manner. Eye development begins with the diVerentiation of the R8 photoreceptor neurons at uniformly spaced positions. These
neurons signal neighboring cells to develop into ommatidia
(unit eyes). The sequential diVerentiation of the other
seven-photoreceptor types (R1R7) then follows. Each R8
neuron recruits one of each cell type such that seven photoreceptors eventually cluster around each R8 neuron (Jeibmann and Paulus 2009).
The power of Drosophila lies in the ability to dissect cellular signaling pathways by performing large-scale unbiased genetic screens that make no assumptions about what
kind of molecules are involved. Forward genetic screens
(from mutant phenotype to causative gene) using chemical
mutagenesis, insertional mutagenesis, or RNA interference
(RNAi) have been used to identify mutations associated
with neurodegeneration. For example, Drosophila mutants
in the bubblegum, swisscheese, and drop-dead loci were
identiWed by screening for Xies with reduced life span and
then analyzing their brain pathology. Some of these genes
have mammalian homologs that also cause neurodegenera-

tion when mutated, indicating that this approach can identify conserved genes that are essential for maintaining
nervous system integrity across distant organisms (Lu and
Vogel 2009; Roman 2004). Dominant genes associated
with neurodegenerative diseases frequently code for proteins that can misfold and aggregate. One example is -synuclein, which is found in the Lewy bodies characteristic of
Parkinsons disease. Similar protein misfolding has been
observed when -synuclein was expressed in transgenic
Drosophila (Feany and Bender 2000; Auluck et al. 2002).
The use of P elements allows the number of transgene
copies to be increased in almost any genetic background.
These vectors are most commonly used to generate hypermorphs by overexpressing a gene of interest or to complement loss of function mutations. For example, the Gal4/
UAS binary system has been used to overexpress many
proteins (Fig. 1; DuVy 2002). This system involves two
transgenic cassettes with one harboring the yeast Gal4 transcription factor under the control of deWned promoter/
enhancer sequences or enhancer detector P elements and
the other containing an upstream activating sequence
(UAS) responder with the gene of interest cloned downstream of a Gal4 responsive promoter. Hormone-inducible
Gal4 systems based on the Gal4-estrogen receptor
(Gal4ER) or the Gal4-progesterone receptor fusion proteins
have also been described (Jones 2009). These systems can
activate transcription from UAS responders after feeding
Xies either -estradiol or the anti-progestin RU486.
In a related approach, the Tet-On tetracycline/doxycycline inducible system (Fig. 2) combines a modiWed version
of the inducible tetracycline-responsive transactivator
(rtTA-M2) with the Gal4 system (Stebbins et al. 2001;
Stebbins and Yin 2001). A Tet-OV system in which transcription is negatively regulated by the presence of tetracycline or doxycycline combined with the GAL/UAS system
has also been described (Stebbins et al. 2001). More
recently, a Tet-OV system using a transposon (piggyBac)based vector has been developed for gene targeting studies
(Hara et al. 2008). The use of deWned promoters, such as

123

Hum Genet (2012) 131:535563

539

Fig. 2 Schematic representation of tetracycline-regulated systems: Tet-OV and Tet-On (Stieger et al. 2009)

torso-like tsl, which targets border cells and a subset of posterior cells of the follicle, as well as reverse polarity repo
(glial), embryonal lethal abnormal vision elav (pan-neuronal), Glass Multimer Reporter (GMR, eye-speciWc) and
mhc (pan-muscular), allow spatial expression patterns to be
controlled (Jeibmann and Paulus 2009).
Conditional control of transgene expression can be
achieved through the use of the Saccharomyces cerevisiae
FLP/FRT and the P1-bacteriophage Cre/loxP recombination systems (Fig. 3; Bischof and Basler 2008). These systems rely on the capacity of site-speciWc FLP or Cre
recombinases to recognize and bind to their recognition
sequences and catalyze recombination between FRT and
LoxP sites, respectively. These systems (mainly FLP) have
been extensively used to generate genetic mosaics, chromosomal rearrangements, and site-speciWc mutagenesis (Horn
and Handler 2005; Oberstein et al. 2005; Siegal and Hartl
2000; St Johnston 2002). Streptomyces phage C31-mediated transgenic systems that produce speciWc targeted integration have also been described. These systems are based
on the site-speciWc C31 integrase, which mediates
sequence-directed, irreversible, and highly eYcient integration between a bacterial attachment site (attB) and a phage

attachment site (attP). C31-based systems include Pacman, which is a BAC transgenic platform that allows targeted insertion of large DNA fragments into speciWc
locations (Venken et al. 2006), and the FlyC31 system
(Bateman et al. 2006). Zinc-Wnger nuclease (ZFN) technology has also been used for gene targeting in Drosophila
(Beumer et al. 2006, 2008; Reyon et al. 2011), and RNAi
technology is routinely used for genetic downregulation
and genomic screens (Mohr et al. 2010). Neurodegenerative disorders associated with loss of function mutations
can be modeled by targeting endogenous Drosophila genes
through homologous recombination, transposon-mediated
mutagenesis, or transgenic RNAi. Importantly, all of these
allow for the generation of genetic knockouts (Roman
2004). A comprehensive database of Drosophila genetics
and genomics can be found at http://flybase.org.
ZebraWsh (Danio rerio) The zebraWsh Danio rerio has
experienced a dramatic rise in popularity as an experimental organism. This low-cost vertebrate system has a close
evolutionary relationship with mammals, yet combines the
power of forward and reverse genetic screens otherwise
found only in invertebrates. Despite the evolutionary divergence of teleost Wsh over more than 400 million years,

123

540

Hum Genet (2012) 131:535563

Fig. 3 Genomic engineering


using Cre/Flp-mediated DNA
recombination. Cre or Flp catalyze recombination between two
directly repeated loxP or FRT
sequences (triangles), respectively. In the example, a region
B is deleted which may lead to
gene inactivation or cause activation of a downstream gene if
region B contains sequences
that block transcription

zebraWsh are more closely related to humans than invertebrates. This organism, originally from the river Ganges in
India, is a common aquarium Wsh that is easy to maintain
and breed. Another major advantage of zebraWsh is the
ease of imaging due to its transparency and external
development.
An adult female can lay 200500 eggs every 10 days and
the eggs are fertilized externally. The larvae have adult features after 72 h, are autonomous after 4 days, and are fertile
by 3 months of age. In addition, although there are some
notable diVerences in structure and scale between the
zebraWsh and human brain, the overall organization shows
many similarities. SpeciWcally, zebraWsh have a fore-, midand hindbrain including a diencephalon, telencephalon and
cerebellum. Furthermore, the peripheral nervous system
has both motor and sensory components. ZebraWsh exhibit
many higher order behaviors including memory, conditioned responses, and social behaviors like schooling (Lieschke and Currie 2007). SpeciWc regions of zebraWsh brain
are strikingly conserved with the human counterparts, making zebraWsh an excellent organism to model human neurodegenerative conditions. Moreover, the advanced optical
imaging techniques available in zebraWsh research allow

123

sophisticated anatomic correlations to be drawn between


brain structure and the development of complex behavioral
traits (Cheng et al. 2011).
Many approaches to generating transgenic zebraWsh are
currently available, including microinjection or electroporation of plasmid DNA or transgenic cassettes into the cytoplasm of fertilized eggs. Use of the I-Sce1 meganuclease
and the Tol2 transposon has greatly increased eYciency of
transgenesis and decreased mosaicism (Kawakami et al.
2000; Sager et al. 2010; Thermes et al. 2002). Other techniques include sperm-mediated gene transfer, pseudotyped
retroviral infection, retroviral infection of in vitro-cultured
sperm, and BAC transgenesis (Gama Sosa et al. 2010a;
Sager et al. 2010). The ability to regulate gene expression
in a cell type-speciWc and temporally restricted manner has
been achieved using Gal4/UAS (Halpern et al. 2008) and
Tet-inducible systems (Huang et al. 2011c; Knopf et al.
2010). The Gal4 driver in the zebraWsh Gal4/UAS system
contains an attenuated version of the herpes simplex virus
(HSV) VP16 trans-activation domain. Using these modiWed
Gal4-Vp16 constructs, driver lines are being generated to
mirror expression patterns of endogenous genes in order to
target desired temporal and tissue-speciWc transgenic

Hum Genet (2012) 131:535563

expression (Asakawa et al. 2008; Distel et al. 2009; Sager


et al. 2010). A database of currently available zebraWsh
Gal4 lines can be found at http://www.zfin.org.
More recently, a mifepristone-inducible LexPR system
has been developed in zebraWsh, which can be induced at
any stage of the life cycle by administration of mifepristone
in the water. This system utilizes a transgene derived
recombinant transcription factor (LexPR) consisting of the
DNA-binding domain of the bacterial LexA repressor coupled to a truncated ligand-binding domain of the human
progesterone receptor with an activation domain of the
human NF-B/p65 protein. The gene of interest is placed
under the control of an operator-promoter sequence
(LexOP) containing four ColE1 operator sequences fused
to a minimal 35S promoter from the cauliXower mosaic
virus (CaMV). Upon binding of the progesterone antagonist
mifepristone (RU-486) to the progesterone receptor LDB,
the LexPR transactivator binds to the operator sequences
and induces gene transcription (Emelyanov and Parinov
2008). The Cre/loxP (Hans et al. 2009; Langenau et al.
2005; Liu et al. 2008b; Pan et al. 2005; Thummel et al.
2005) and FLP/FRT (Wong et al. 2011) site-speciWc
recombination systems have also been applied to conditional mutagenesis in zebraWsh (Deiters and Yoder 2006).
A well-established gene knockdown technology in
zebraWsh uses morpholinos, which are synthetic oligonucleotides of approximately 25 bases that hybridize speciWcally
to complementary sequences of mRNA to disrupt translation initiation or splicing (Bill et al. 2009; Deiters and
Yoder 2006). Morpholinos have standard nucleic acid bases
that are linked to morpholine rings instead of ribose or
deoxyribose. The morpholine rings are attached to nonionic phosphorodiamidate groups instead of phosphates.
Morpholinos can be microinjected into the cytoplasm of
single cell stage embryos or into the yolk of up to 16-cell
stage embryos. Morpholinos can diVuse into cells until the
16-cell stage when a membrane forms between the yolk and
the cells that will form the embryo proper. Another eVective approach for targeted gene knockdown in zebraWsh is
the use of short interfering RNAs (siRNAs) (Kelly and
Hurlstone 2011).
The lack of a gene targeting technology equivalent to
homologous recombination in embryonic stem (ES) cells
prevents the range of gene targeting in zebraWsh that can be
achieved in the mouse. However, using locus-speciWc
ZFNs, speciWc gene knockouts are routinely created in
zebraWsh (Amacher 2008; Ekker 2008; Remy et al. 2010).
Gene knockouts are produced by constructing chimeric
ZFNs consisting of a DNA-binding zinc-Wnger domain and
a cleavage domain of a restriction endonuclease (FokI).
Each DNA-binding zinc-Wnger domain is engineered to
confer speciWcity for a particular triplet of DNA base pairs.
The DNA-binding domains of individual ZFNs typically

541

contain between three and six individual zinc-Wnger


repeats. Gene knockouts are generated by injecting ZFNs
directly into the embryo (Reyon et al. 2011).
More recently, a novel technology for gene targeting in
zebraWsh has been developed based on the use of transcription activator-like (TAL) eVectors. TAL eVectors are a family of virulence factors produced by the bacteria
Xanthomonas, which bind to speciWc promoter sequences
in the host plant to regulate genes aVecting bacterial growth
(Boch and Bonas 2010; Romer et al. 2010). To create a
technology that allows genome editing, a TAL eVector
DNA-binding domain that recognizes single nucleotides is
fused to a FokI cleavage domain (TAL eVector nucleases,
TALENs) (Boch et al. 2009; Moscou and Bogdanove 2009;
Voytas and Joung 2009; Zhang et al. 2011a). The use of
customized TALENs has been successful for targeting the
tnikb and dip2a loci in zebraWsh (Huang et al. 2011b).
Mutant orthologues of human disease loci in zebraWsh
have been generated on a large scale using N-ethyl-N-nitrosourea (ENU) chemical mutagenesis as well as by insertional mutagenesis by retroviral insertion (Amsterdam et al.
2004; Gaiano et al. 1996; Golling et al. 2002; Lieschke and
Currie 2007) or by the use of transposons (Sleeping Beauty
SB, Tol2 or Ds/Ac systems) (Choo et al. 2006; Emelyanov
et al. 2006; Ivics et al. 1997; Kawakami 2005; Kawakami
et al. 1998). Mutant genes involved in human disease have
also been identiWed using a reverse genetic approach called
targeting-induced local lesions in genomes (TILLING) that
combines ENU mutagenesis in the generation of an F1
mutant zebraWsh library with heteroduplex DNA screening
of pooled DNA by Cel I endonuclease screening or direct
DNA sequencing (Draper et al. 2004; Moens et al. 2008;
Wienholds et al. 2003). Genes of interest obtained through
this process can be found at http://www.sanger.ac.uk/
Projects/D_rerio/mutres/ and http://webapps.fhcrc.org/science/tilling/index.php. Genetically mapped retroviral insertions in zebraWsh orthologues of human disease genes also
exist, and these mutant genes are available commercially
(Znomics). Information on transposon insertions can be
obtained through the International Gene Trap Consortium
at http://www.genetrap.org/.
Mouse (Mus musculus) The mouse was the Wrst organism in which transgenic technology and gene targeting
became widely available. Despite the notable advances that
have occurred in other species, the mouse remains the most
widely used system for modeling human neurodegenerative
diseases. The mouse is closely related to humans through
the evolutionary radiation of Epitheria about 75 million
years ago, which resulted in the generation of rodents and
primates. The general outline of development is similar in
mice and humans, and there are syntenic relationships
between mouse and human genes across much of the
genome. The availability of inbred strains allows studies to

123

542

be performed on genetically homogeneous backgrounds.


Other advantages of the mouse include its relatively low
cost, short generation time (18.521 days, depending on the
strain), the high environmental control that is possible, and
the availability of well-developed technologies for genome
manipulation that allow virtually any disease-associated
genetic alteration in humans to be introduced into the
mouse. In addition, techniques for forward genetic mutagenesis screens, complex behavioral analysis, and in vivo
imaging are well developed in the mouse (Muller and
Grossniklaus 2010).
Transgenic models relevant to Alzheimers disease
(Crews et al. 2010; Elder et al. 2010b; Gotz and Ittner
2008; Harvey et al. 2011; Trancikova et al. 2011), Parkinsons disease (Bezard and Przedborski 2011; Crabtree and
Zhang 2011; Harvey et al. 2011; Skaper and Giusti 2010;
Trancikova et al. 2011), Huntingtons disease (Harvey et al.
2011; Skaper and Giusti 2010; Trancikova et al. 2011),
amyotrophic lateral sclerosis (ALS) (Harvey et al. 2011;
Trancikova et al. 2011), schizophrenia (Arguello and
Gogos 2010; Jaaro-Peled 2009; Young et al. 2010), childhood-onset psychiatric disorders (Robertson and Feng
2011), lysosomal storage disorders (Haskins et al. 2006;
Suzuki and Mansson 1998), bipolar disorder (Kato et al.
2007), depression (Snyder et al. 2011; Urani and Gass
2003), and genetic predisposition to drug addiction
(Changeux 2010; Sora et al. 2010) have all been created in
the mouse. These models have been used to develop novel
therapeutic strategies. For example, pharmacological chaperone therapy by active-site-speciWc chaperones has proved
promising for the treatment of some forms of Fabrys disease (deWciency of -galactosidase A, -gal A) by treating a
transgenic mouse expressing mutant -gal A (R301Q) with
the competitive inhibitor 1-deoxygalactonojirimycin
(Khanna et al. 2010; Germain and Fan 2009). Moreover,
for some diseases, preclinical testing in the mouse is virtually regarded as a necessary step before proceeding to
human clinical trials.
Various strategies have been developed to prepare transgenic mice. The most common involves injection of a
transgenic DNA cassette into the male pronucleus of a fertilized mouse egg followed by its implantation into a pseudopregnant mother. Usually a cDNA or other cassette
containing the sequences to be expressed are positioned
downstream of a heterologous promoter that confers spatial
and temporal expression. The inclusion of upstream Kozak
sequences allows for ribosomal recognition of the initial
ATG site, while introns placed at the 5 or 3 end of the
transgene allow for splicing, which makes the mRNA more
stable and favors translocation from the nucleus to the cytoplasm. A termination signal containing a poly (A)-addition
sequence also is placed at the 3 end to improve transgene
expression.

123

Hum Genet (2012) 131:535563

Generally, restricted transgene expression in mature neurons can be achieved with pan neuronal promoters, such as
the tubulin 1, platelet-derived growth factor B-chain
(PDGF-) (Liu et al. 2004), neuron-speciWc enolase (NSE)
(Sakimura et al. 1987; Wen et al. 2004), neuroWlament L
(NFL) (Beaudet et al. 1993), or the microtubule-associated
protein 2 (MAP2). Regionally restricted neuronal expression
can be achieved with promoters like calmodulin kinase II
(CamKII), that drives forebrain speciWc neuronal expression,
while expression in interneurons can be driven with the Glutamic acid decarboxylase 1 (GAD1) promoter (Ma et al.
2006). Pan-neural transgene expression has also been
achieved with the murine thymocyte antigen promoter
(Thy1.2) (Luthi et al. 1997; van der Putten et al. 2000) and
with the prion protein (PrP) promoter (Borchelt et al. 1996;
Xu et al. 2010). Transgene expression in neural progenitor
cells can be achieved with the neural speciWc enhancer from
the second intron of the nestin gene in combination with a
minimal promoter or the native nestin promoter (Zimmerman
et al. 1994). The doublecortin promoter (Dcx) is transiently
active in diVerentiating and migrating neurons in the embryo
and adult (Walker et al. 2007; Piens et al. 2010). AstrocytespeciWc transgene expression can be driven by the glial Wbrillary acidic protein (GFAP) promoter (Miura et al. 1990;
Brenner et al. 1994; Zhuo et al. 1997). The use of the human
GFAP promoter also results in transgenic expression in
radial glial cells during development, despite the fact that the
endogenous murine GFAP promoter is not active in radial
glia. Oligodendrocyte-speciWc promoters include myelin
basic protein (MBP), proteolipid protein (PLP), and 23cyclic nucleotide 3-phosphodiesterase (CNPase). Mammalian promoter databases and software tools can be found at
http://rulai.cshl.edu.
Transgenes typically integrate randomly as concatemers
of 70100 kb in length. In some instances, local chromatin eVects may result in transgene silencing or altered
expression due to eVects from local enhancer/promoter elements (Gaszner and Felsenfeld 2006), which can be pronounced in brain (Elder et al. 1994). Local chromatin
eVects can be often avoided by use of insulator sequences
that establish genomic barriers, which block interactions
between adjacent chromatin domains and protect DNA
sequences from eVects of distal and neighboring genetic
elements (Gaszner and Felsenfeld 2006; Giraldo et al.
2003; Houdebine 2010). Local chromatin eVects can also
be largely avoided with the use of P1 bacteriophage artiWcial chromosomes (PACs), bacterial artiWcial chromosomes
(BACs), and yeast artiWcial chromosomes (YACs) (Montoliu et al. 1993) that permit the cloning of large genomic
regions usually containing all necessary regulatory elements for proper temporal and spatial expression (Giraldo
and Montoliu 2001). PACs and BACs can include up to
350-kb inserts, while YACs can accommodate more than

Hum Genet (2012) 131:535563

1 Mb. Relatively simple technologies have been developed


for introduction of genetic modiWcations into artiWcial chromosomes through recombination (recombineering or retroWtting). This technology has, for example, been used to
perform widespread expression pattern mapping of genes
expressed in the CNS using BACs retroWtted with GFP
(GENSAT project, http://www.gensat.org).
Smaller transgenes (<7.5 kb) can be cloned into recombinant retroviral or lentiviral vectors and the infectious particles can be used to infect zona pellucida-free embryos.
Alternatively, infectious viruses can be injected directly
into the perivitelline space of the embryo or used to infect
cultured ES cells. Intracytoplasmic sperm injection (ICSI)
of DNA-loaded sperm cells has also proven eVective for
production of transgenic mice, particularly when large
DNA constructs, such as sub-megabase sized artiWcial
chromosomes are used (Moreira et al. 2004, 2006). With
this approach, foreign DNA is loaded onto freeze-thawed
spermatozoa in vitro, and individual sperm heads are
injected into metaphase II oocytes. This methodology has
for example been used to prepare transgenic mice harboring
the human amyloid precursor protein (APP) by YAC transgenesis (Moreira et al. 2007).
A major step in the establishment of the mouse as the
dominant system for modeling human neurodegenerative
diseases was the development of gene targeting strategies
(Cheah and Behringer 2001; Joyner and Guillemot 1994).
Although primarily used to generate gene knockouts, gene
targeting through homologous recombination also permits
introduction of point mutations, deletions, insertions, inversions, or translocations into ES cells. After selection, chimeric mice are generated by injecting genetically modiWed
ES cells into mouse blastocysts. Historically, most ES cell
lines in widespread use were derived from the 129/Sv
strain, which contains the wild-type alleles of the albino,
pink-eyed dilution, and agouti coat-color loci. By injecting
modiWed 129 ES cells into recipient blastocysts from
C57BL/6 donors, which carry a recessive black coat color,
chimeric mice can be readily identiWed by their agouti coat
color. Upon breeding of the chimeric mice, lines of genetically modiWed mice can be established. Because strain
background may have signiWcant eVects on phenotype,
mutant lines are frequently backcrossed to other inbred
strains (usually C57BL/6). Unfortunately, even with the use
of speed congenics, backcrossing can require over a year to
place the targeted mutation on a C57BL/6 background. A
more direct approach would be to use ES cells derived from
the C57BL/6 strain. However, historically, C57BL/6 ES
cell lines have been regarded as diYcult to establish and
maintain, although in recent years their use has become
more common, and some commercial suppliers now routinely generate gene-targeted mice on the C57BL/6 background.

543

While not as widely used as gene targeting in ES cells,


gene knockouts in the mouse can also be generated using
ZFNs (Carbery et al. 2010; Cui et al. 2011; Goldberg et al.
2010; Li et al. 2011a; Meyer et al. 2010). Random generation of point mutations with ENU or random gene interruptions with gene traps through mobile DNA elements,
including class I retrotransposons like LINE1 (L1) (Ostertag et al. 2007; An et al. 2006) and class II transposons like
Sleeping Beauty (SB) (Izsvak and Ivics 2005; Yae et al.
2006; Largaespada 2009b; Takeda et al. 2008), are generating large comprehensive resources of mutant mouse strains
(Carlson and Largaespada 2005; Largaespada 2009a). More
recently, the DNA transposon piggyBac from the cabbage
looper moth Trichoplusia ni has been used as both a mutagenic tool (Chew et al. 2011; Rad et al. 2010; Wang et al.
2008) and as a method for mobilizing large pieces of DNA
(100 kb) within the mouse genome (Li et al. 2011b).
The ENU mutagenesis approach involves the selection
of a phenotypic trait of interest followed by identiWcation of
the genetic locus or loci that control that trait (Justice et al.
1999). This approach, usually coordinated in large consortiums or centers, involves phenotyping F1 mutant mice
to identify traits of interest with genetic loci that can be
subsequently determined by gene mapping approaches.
Recessive phenotypes require outcrossing and backcrossing
to recover mutations in the homozygous state. Random SB
transposon mutagenesis usually includes a transposon cassette containing a poly-A gene trap composed of an internal
promoter, a reporter gene like GFP, and a splice donor. If
the gene trap integrates into a transcription unit in proper
orientation, the reporter gene will be activated marking the
active locus. Linker-mediated PCR can be used to determine transposon integration site (Largaespada 2009a).
Because ZFN and TALEN-mediated technologies generate
speciWc gene knockouts, these approaches are largely
replacing the use of mobile DNA elements and ENU for
genome-wide mutagenesis.
Constitutive and conditional knockdown of gene expression has also been accomplished with RNAi technology
(Gao and Zhang 2007). In this technique, expression vectors are designed to contain sense and antisense regions that
are complementary to a selected mRNA target. The resulting transcripts, which have a stem-loop structure, can fold
back into short hairpin (<30 bp) RNAs (shRNAs) that are
processed by the Dicer ribonuclease into siRNAs. The
eYciency of gene knockdown can be as high as 90% or
greater. In one study, a scalable system for studying gene
function in mice was developed using a combination of
Xuorescence-coupled miR30-based shRNAs with high
eYciency ES cell targeting (Premsrirut et al. 2011).
A potential drawback of the gene knockout approach is
the embryonic lethality of many null mutations, which precludes study of gene function in the adult. To circumvent

123

544

Hum Genet (2012) 131:535563

Fig. 4 Flp recombinase-mediated cassette exchange (RMCE). Flp


recombinase catalyzes the reciprocal recombination between two identical short Flp recombinase targets (FRTs) present in the target locus
and in the exchange plasmid. The wild-type FRT recognition consists

of two 13-bp inverted repeats that Xank an 8-bp core sequence (red).
The availability of FRT mutants permits the exchange of genetic cassettes for the targeted integration of a desired transgene

these problems, conditional recombination systems, such as


the Cre-loxP (Nagy 2000; Miller 2011; Wang 2009) and
Flp-FRT systems described above, allow for the location
and timing of gene inactivation to be closely regulated. An
ever-expanding number of mouse Cre deleter lines have
been created (http://nagy.mshri.on.ca/cre_new/). Other
conditional systems, such as the D6 phage Dre-rox and
those using novel yeast site-speciWc recombinases (B3 and
KD) that allow genome manipulation in vivo, have also
been developed (Anastassiadis et al. 2009; Nern et al.
2011).
The tamoxifen (RU486)-inducible conditional system
uses a chimeric Cre protein fused to a mutated nuclear
estrogen receptor that no longer responds to natural estrogen but which can be activated with synthetic RU486
(Metzger et al. 1995; Garcia and Mills 2002; Hayashi and
McMahon 2002). This system permits temporal and tissue
or cell type-speciWc gene inactivation within a time frame
dictated by the investigator, rather than determined by time
of onset and pattern of a speciWc promoters expression.
Cre recombination has also been used to activate gene
expression in brain neurons by including strong translational stop sequences Xanked by loxP sites between a promoter and the regulated gene (De Gasperi et al. 2008). Cre
can be used to delete megabase regions and mediate trans-

locations between non-homologous mammalian chromosomes at very high eYciencies (Brault et al. 2007; Horev
et al. 2011; Wirth et al. 2007). In comparison to Cre, Flp
has been considered less eYcient partly due to an unsuitable optimum temperature in mammalian cells. However, a
thermostable Flpe variant has been identiWed (Buchholz
et al. 1998; Ringrose et al. 1998). The Flp/FRT system is
widely used for recombinase-mediated cassette exchange
(RMCE) to generate knock-in mutations in speciWc genes
in the mouse genome (Fig. 4) (Roebroek et al. 2011). Also
the Cre/loxP system has been successfully used for RMCE
(Liu et al. 2006).
Because Cre and Flp excision/recombination is an irreversible event, they are not useful when the ability to
switch a gene on and oV is desirable. Thus, a variety of
inducible/regulatable systems has been developed. The
most commonly used of these in transgenic mice are the
Tet-regulated systems (Fig. 2; Stieger et al. 2009). The TetOV system uses a chimeric tetracycline transactivator (tTA)
protein made by fusing the E. coli tetracycline repressor
(TetR) with the HSV transcriptional activator VP16 protein. tTA binds to the tet-O operator and activates a CMV
minimal promoter coupled to the tet-O operator (tetresponse element, TRE) activating the transgene of interest.
Tetracycline or its more stable analog doxycycline

123

Hum Genet (2012) 131:535563

(Dox) bind to tTA preventing it from binding to the TRE,


thus preventing transactivation of the transgene (Gossen
and Bujard 1992). To circumvent the necessity of continuous doxycycline administration to maintain genes in the
oV state, the Tet-On system was developed. The Tet-On
system is based on the observation that mutations within
the chimeric tTA activator protein (rtTA) can reverse its
behavior so that, rather than spontaneously binding to the
TRE, it does so only in the presence of tet or doxycycline
(Urlinger et al. 2000).
A variation on the Tet system known as the Kruppelassociated box (KRAB) Tet-On system also permits tight
regulation of transgene expression by doxycycline. This
system takes advantage of the repressor activity of a fusion
protein tTRKRAB made of the KRAB domain, found in
many vertebrate transcriptional regulators (such as human
Kox1 or mouse Kid-1), and the tetracycline repressor
(tetR). The tTRKRAB-mediated repression of cellular Pol
II or Pol III promoters juxtaposed to tet operator (tetO)
sequences can be reversibly controlled by doxycycline
(Deuschle et al. 1995; Szulc et al. 2006). A database containing tTA and rtTA eVector mice, Tet-O reporter strains,
and Cre-inducible rtTA strains can be found at http://
www.zmg.uni-mainz.de/tetmouse/.
A tet-inducible shRNA gene knockdown system has
been developed for use in ES cells, which uses recombinase-mediated cassette exchange (RMCE) through Flpmediated recombination in the Rosa26 locus (Seibler et al.
2007). The exchange vector carries the shRNA under the
control of the pol III H1-tetO promoter, a codon-optimized
itetR gene under the control of the chicken -actin promoter and a truncated neoR gene for positive clonal selection after successful RMCE. Transgenic animals are then
prepared from the recombinant ES cells (Seibler et al.
2007). Other inducible systems that have been successfully
used in transgenic mice include the lac-inducible system
(Cronin et al. 2001) as well as insect- and mammaliansteroid inducible systems (Albanese et al. 2002; Graham
2002).
Rat (Rattus norvegicus) Rats have a lifespan of 2.5
3.5 years and a gestation time of 1922 days. This rodent
exhibits many physiological and morphological characteristics similar to those of humans. Historically, neuroscientists have preferred rats to mice as model systems and,
indeed, many aspects of rat pharmacology and physiology
are regarded as more relevant to humans and easier to
observe than in mice (Abbott 2004; Bugos et al. 2009). Furthermore, rats generally exhibit a more robust capacity for
learning in behavioral tasks. Although, virtually all the
technologies used for generating transgenic mice should be
applicable to the rat in theory, transgenic technologies are
markedly less available in rats. For example, while core
facilities for generation of transgenic mice are common in

545

major academic research centers, these facilities have only


recently become more available for rats. In addition, rat ES
cells have been diYcult to establish and maintain although
rat ES cells for gene targeting by homologous recombination have been recently described (Tong et al. 2010, 2011).
Moreover, random mutagenesis by ENU (Zan et al. 2003),
mobile DNA elements, such as the piggyBac (Jang and
Behringer 2007) and Sleeping beauty (SB) DNA transposons (Ivics et al. 2011; Izsvak et al. 2010), ZFNs (Cui et al.
2011; Geurts et al. 2009), and TALENs (Christian et al.
2010; Tesson et al. 2011) have permitted the generation of
knock out rats that mimic human diseases (Huang et al.
2011a). A database of mutant rats generated by mobile
DNA technology can be found at http://www.knockoutrat.org/.

Other transgenic organisms


Transgenic technology has been developed in a variety of
other mammalian species. For example, human disease
models have been prepared in transgenic rabbits (Oryctolagus cuniculus) (Bosze et al. 2003; Liang et al. 2006), and
commercial entities have been established solely dedicated to the production of transgenic rabbits. Transgenic
models of Alzheimers disease have been created in pigs
(Sus domestica) using nuclear transplantation via cell
fusion (Kragh et al. 2009), and ZFNs have been used to
generate biallelic knockout pigs (Hauschild et al. 2011).
More recently, it was shown in transgenic pigs prepared
by somatic cell nuclear transplantation (SCNT) that
expression of the Huntingtons disease protein results in
neuronal apoptosis (Yang et al. 2010). A transgenic Huntingtons disease model in rhesus monkeys (Macaca mulatta) has been prepared by lentiviral injection into the
oocyte of polyglutamine-expanded Huntingtin (HTT) followed by fertilization by ICSI (Yang et al. 2008). Transgenic rhesus monkeys have also been generated using a
simian immunodeWciency virus (SIV)-based vector to
infect early-cleavage stage embryos (Niu et al. 2010). A
major obstacle to generating transgenic non-human primates is the low eYciency of assisted reproductive technologies in producing oocytes and embryos suitable for
genetic engineering as well as the technical challenges of
embryonic stem cell derivation and cloning. In addition,
the high cost of production and maintenance of nonhuman primates and other large animals prevents their
wider use in research.
EVorts to develop transgenic models for neurodegenerative disorders are being made in other species as well. For
example, human APP695 has been expressed in the ascidian
non-vertebrate chordate Ciona intestinalis, where it was
observed to be processed into A peptides in the nervous

123

546

system resulting in formation of amyloid plaques and


altered larval behavior (Virata and Zeller 2010). Clearly,
eVorts to develop models of human neurodegenerative disease can be expected to continue in traditional as well as
non-traditional species.

Applications to age-related human inherited


neurodegenerative diseases
Human neurodegenerative diseases take various forms.
Among the most common are age-related neurodegenerative disorders, such as Alzheimers disease (Selkoe
2011), Parkinsons disease (Shulman et al. 2011), Huntingtons disease (Finkbeiner 2011), the frontotemporal
dementias (Ferrari et al. 2011; Seelaar et al. 2011), and
ALS (Ferrari et al. 2011). In each of these disorders,
there is a selective vulnerability of distinct neuronal
populations to the neurodegenerative process, and the
clinical manifestations of each disorder reXect the distinctive subpopulations of neurons involved. For example, loss of dopaminergic neurons is responsible for the
extrapyramidal features characteristic of Parkinsons
disease, while loss of motor neurons causes the weakness and atrophy characteristic of ALS. Another
common feature of human neurodegenerative disorders
is the accumulation of protein aggregates. These accumulations can take various forms. For example, in
Alzheimers disease, accumulation of -amyloid results
in extracellular neuritic plaques and aggregates of
abnormally phosphorylated forms of tau protein predominate in intracellular neuroWbrillary tangles. In ALS,
characteristic intracellular accumulations of cytoskeletal
elements are also seen, but the aggregates are composed primarily of neuroWlament proteins. In contrast, the Lewy bodies characteristic of Parkinsons disease and related disorders
contain aggregates of -synuclein.
With the exception of Huntingtons disease, these disorders are largely sporadic, and although a number of
genetic risk factors have been identiWed, the disorders
can be traced to a single inherited gene in only a small
number of cases. However, despite their rarity, it is
likely that the genetic forms of these disorders target
common pathways that are also aVected in the sporadic
diseases. In addition, because transgenic modeling
requires a genetic lesion or at least a hypothesis that can
be modeled genetically, the mutations found in these
familial cases have formed the basis for most of the
transgenic modeling that has been conducted in animals.
In Table 1, selected examples are presented of transgenic models related to hereditary neurodegenerative
diseases that were created with the transgenic systems
discussed in this review.

123

Hum Genet (2012) 131:535563

Transgenic modeling and the example of Alzheimers


disease
While it is beyond the scope of this review to discuss the
relevance of all the models listed in Table 1 to their respective disorders the case of Alzheimers disease illustrates
many of the issues common across disorders. AD may in
many ways be regarded as an ideal disease for modeling in
transgenic animals. In humans it has a well-recognized
pathology consisting of senile plaques and neuroWbrillary
tangles (NFTs). The major constituents of these lesions are
well deWned. In the case of plaques being the -amyloid
(A) peptide, which is derived from processing of the
larger amyloid precursor protein (APP) that itself belongs
to the APP-related protein family. By contrast hyperphosphorylated forms of tau are the major constituents of NFTs.
AD also has other well-recognized pathological features
including neuronal and synaptic loss, dystrophic neurites,
reactive astrocytes and activated microglia. There is in
addition a well-deWned behavioral phenotype.
EVorts at modeling AD have been conducted in all the
transgenic systems described in Table 1 based in large part
on the amyloid cascade hypothesis which postulates that
shunting of APP processing toward longer more neurotoxic
A42 species sets oV a chain of pathological events (Selkoe
2001, 2011). Mutations in three genes including the amyloid precursor protein (APP) as well as two related proteins, presenlin-1 and presenlin-2, have been identiWed as
causing familial Alzheimers disease (FAD) providing the
genetic lesions needed for a transgenic model. However,
characteristics of the individual organisms have dictated
that diVerent strategies be used to model AD in diVerent
organisms.
In C. elegans there is a single APP-related gene, apl-1
(Ewald and Li 2010). Knockout of apl-1 causes lethality
during early larval development while overexpression
causes defects in brood size, movement and reduces viability. A functional domain homologous to the Fe65 binding
domain within the cytoplasmic tail of mammalian APP
exists in apl-1 and a C. elegans ortholog of Fe65 (FEH-1)
exists. Several orthologues to the mammalian - secretases
can be found in C. elegans although no -secretase activity
has been clearly identiWed. C. elegans contains an active
-secretase complex and like mammalian systems has multiple presenilins. Indeed work with the C. elegans presenilin SEL-12 provided the Wrst insights into the role of
presenilins in mediating Notch signaling (Ewald and Li
2010). However, apl-1 lacks an A sequence and thus does
not produce A peptide. As such to establish C. elegans
models of AD, investigators have introduced various forms
of transgenic human A. In muscle cells, human A42 has
been expressed using an A sequence fused to an artiWcial
signal sequence designed to allow the extracellular release

Hum Genet (2012) 131:535563

547

Table 1 Transgenic technologies used in the preparation of animal models of neurodegenerative diseases
Disease

Strategy

Pathology and other Wndings

Reference

Caenorhabditis elegans
AD

Pan neuronal expression of human A42

DeWcits in odorant preference-associated


learning behavior, serotonin-controlled
behaviors, experience-dependent
learning, and egg laying

(Dosanjh et al. 2010)

AD

Body wall muscle expression


of human A42

Accumulation of amyloid deposits with


Wbrillary structure accompanied by
progressive paralysis

(Link 1995)

AD

Inducible pan-neuronal
expression of human A42

Neuronal amyloid deposits associated with


subtle phenotypic changes

(Wu et al. 2006)

FTLD

Expression in dopaminergic
neurons of wt or A53T
mutant -synuclein

Accumulation of insoluble phosphorylated


tau, age-dependent neurodegeneration
and neuronal loss, uncoordinated
movement, and greater toxicity in worms
expressing the FTLD-17 mutant tau than
the wt form

(Kraemer et al. 2003)

FTLD/ALS

Pan neuronal expression


of wt human TDP-43

Motor neuron dysfunction, abnormal


motor neuron synapses, and
uncoordinated phenotype

(Ash et al. 2010)

PD

Expression in dopaminergic neurons


of wt or A53T mutant -synuclein

Degeneration and loss of dopaminergic


neurons including dendritic atrophy
and accumulation of -synuclein in perikarya

(Lakso et al. 2003;


Settivari et al. 2009;
Kuwahara et al. 2006;
Harrington et al. 2010;
Settivari et al. 2009)

HD

Expression of the polyglutamine repeat


HT-Q150 in sensory neurons

Age-dependent neuronal dysfunction

(Faber et al. 1999)

HD

Genome wide RNAi screen for modiWers


of YFP-polyQ aggregation

IdentiWcation of large set of genes


involved in protein folding or
degradation that increase polyQ
aggregation when their expression
is knocked down

(Nollen et al. 2004)

Fruit Fly Drosophila melanogaster


AD

gmr-GAL4/UAS-driven triple transgenic


Xies expressing human wild-type APP,
human BACE and wild-type
Drosophila presenilin (dPsn)
or dPsn with point mutations
corresponding to human FAD mutations
in photoreceptor cells of the compound eye

Elevations of both human A40 and A42


peptides; thioXavin S stained amyloid
plaques in the retina and age-dependent
retinal photoreceptor degeneration that
was enhanced by the presence of FAD
related dPsn mutations and suppressed
by genetic reduction of dPsn or by the
treatment with  or -secretase inhibitors

(Greeve et al. 2004)

AD

Pan-neural GAL4-UAS-driven
expression of human 42

Decreased life span, slower locomotion,


altered tapping reXexes and grooming
routines, and age-dependent learning
defects; amyloid deposition associated
with neurodegeneration

(Finelli et al. 2004;


Iijima et al. 2004;
Crowther et al. 2005;
Iijima et al. 2008)

FTLD

Pan-neural GAL4-UAS driven


expression of wt and R406W tau

Accumulation of abnormally
phosphorylated tau without
neuroWbrillary tangles, progressive
neurodegeneration, and early death.
Tau-induced pathology more pronounced
with the FTLD-associated mutation

(Wittmann et al. 2001)

ALS

Motor neuron and photoreceptor


GAL4-UAS driven
expression of wt and mutant hSOD1

Neither loss of motor neurons nor retinal


degeneration was observed, although
motor neuron expression of either wt or
mutant hSOD1 led to progressive
motor dysfunction. Earlier attempts at
expressing hSOD1 in motor neurons
increased longevity and could rescue
the shortened lifespan of a dSod null mutant

(Elia et al. 1999;


Parkes et al. 1998;
Watson et al. 2008)

123

548

Hum Genet (2012) 131:535563

Table 1 continued
Strategy

Pathology and other Wndings

Reference

PD

Pan-neuronal and photoreceptor


GAL4-UAS driven expression of
-synuclein (wt and mutants
A30P and A53T)

Overexpression of wt and mutant forms of


-synuclein causes dopaminergic
degeneration with progressive loss of
dopaminergic neurons and locomotor
dysfunction. Neuronal inclusions reminiscent
of Lewy bodies were found in the cytoplasm

(Feany and Bender 2000)

HD

Photoreceptor neuronal-speciWc
expression of human huntingtin,
2, 75 or 120Q repeats

Neuronal degeneration for which severity


and age of onset correlate with repeat length.
Nuclear localization of huntingtin preceding
neuronal degeneration

(Jackson et al. 1998)

HD

FRT/FLP-mediated deletion
of the huntingtin locus

Removal of the Drosophila HTT homolog, htt,


(Zhang et al. 2009)
results in reduced mobility, long-term survival,
and modulation of adult axonal terminal complexity.
Htt deletion also accelerates the neurodegenerative
phenotype associated with the HD-Q93
polyglutamine repeat associated with HD

Disease

ZebraWsh Danio rerio


AD

Antisense morpholino gene knock-down


of presenilin (psen1 and psen2)
and APP (appa and appb) orthologues

Knockdown of both psen1 and psen2 causes


hydrocephalus and reduced melanin
pigmentation. Psen1 MO also exhibit
somite defects, altered myo D expression,
a short tail, smaller head, and defective
brain development including reduced
branching of trigeminal neurons Loss
of appa and appb function resulted
in reduced body length and defective
convergent-extension movements
during gastrulation

(Campbell et al. 2006;


Groth et al. 2002;
Newman et al. 2009;
Nornes et al. 2008, 2009;
van Tijn et al. 2011)

FTLD ZebraWsh enolase-2 promoter driving


neuronal expression of human
4-repeat tau

Expression found throughout the CNS;


accumulations of tau occur in axons
and neuronal soma in the retina, brain,
and spinal cord

(Bai et al. 2007; Xi et al. 2011)

FTLD Pan-neuronal Huc2 promoter


induced GAL4-VP16
transactivation of UAS Xanked by
Tau P301L (3) and DsRed (5)
in early neuronal cells

Red Xuorescent cells can be seen expressing


tau in embryos. Mutant tau-expressing
embryos exhibit a transient motor phenotype
caused by delayed outgrowth of motor axons
as well as behavioral deWcits in escape response.
Transgenic animals have hyperphosphorylated
tau, tangle-like inclusions, and neurodegeneration
in the spinal cord

(Paquet et al. 2009, 2010)

ALS

In vitro transcribed RNAs of TDP-43


(TARDBP wt and mutants G348C,
A382T, A315T) were injected into 14
cell stage blastulae

Preferential vulnerability of motor neurons


to toxicity of TDP-43 mutants; shorter
motor neuron axons, premature and
excessive branching, and swimming deWcits

(Kabashi et al. 2010)

HD

Rhodopsin promoter driven expression


of EGFP-tagged huntingtin exon 1 with
71Q expanded polyglutamine repeat
(EGFP-HDQ71) in rod photoreceptors

Aggregates of EGFP-HDQ71
fusion protein and loss of rhodopsin expression
associated with rod photoreceptor degeneration

(Williams et al. 2008)

Age-dependent brain amyloid deposition,


dystrophic neurites, reactive astrocytes,
and activated microglia near plaques; l
earning deWcits. Pathology has
earlier-onset in the Swedish/Indiana
APP double mutant

(Calhoun et al. 1999;


Chishti et al. 2001;
Games et al. 1995;
Hsiao et al. 1996)

Mouse Mus musculus


AD

Mouse models generated by pronuclear


injection of mutant APP cDNAs
resulting in pan-neuronal
expression of mutant APPs
associated with FAD
(Indiana, Swedish and Swedish +
Indiana mutants)

123

Hum Genet (2012) 131:535563

549

Table 1 continued
Strategy

Pathology and other Wndings

AD

Pronuclear injection of PS1 FAD


mutations using platelet-derived growth
factor (PDGF), thy-1, neuron-speciWc
enolase (NSE) promoters and others

Age-dependent neuronal and synaptic loss,


(DuV et al. 1996;
increased lipid and protein peroxidation,
Elder et al. 2010b;
and increased sensitivity to excitotoxic
Wen et al. 2004;
injury; impaired hippocampal neurogenesis Wen et al. 2002)
in adult mice; A elevated in brain but
no plaque deposition

AD

Transgenesis with human BAC harboring


the wt PS1 and a retroWtted BAC
harboring the M146 V FAD mutation

Age-dependent brain vascular pathology


(Gama Sosa et al. 2010b)
associated with the expression of the
human PS1 harboring the M416 V
mutation. Transgenic expression human wt
and mutant PS1 mimics entirely
that of the endogenous murine presenilin

AD

Transgenesis with a YAC harboring the


entire 400-kb human APP gene.
The 650-kb YAC was introduced by l
ipid-mediated transfection into ES cells
that were then used to generate chimeric mice

Resulting transgenic animals express


human APP mRNA and protein at
levels comparable to endogenous
APP. No brain amyloid deposition
was observed

(Lamb et al. 1993)

AD

APP Swedish mutation associated with


FAD introduced into the mouse APP gene.
In addition, the three amino acid diVerences
between mouse and human APPs in
the A region were humanized

FAD mutation expressed at physiological


levels and mice produce human A;
increased A production but no amyloid
plaques or neuropathology

(Reaume et al. 1996)

AD

Flp-mediated RMCE harboring a


Tet-inducible GSAP RNAi transgenic cassette

Doxycycline-induction of gamma-secretase
activating protein (GSAP) RNAi in
double transgenic animals harboring
the APPswe and PS19 mutant
alleles reduced amyloid plaque load

(He et al. 2010;


Seibler et al. 2007)

AD

Tet-OV inducible system for APP695


High-level APP expression rapidly
expression in forebrain neurons. Transgenic
induces amyloid pathology. Doxycyline
mice harboring the mouse/human chimeric
administration inhibits 95% of APP
APP695 with the Swedish (KM570/571NL)
expression and reduces A production
to levels found in non-transgenic mice,
and Indiana (V617F) FAD mutations under the
halting the progression of
control of a tet-responsive (tet-O) promoter were
amyloid pathology
bred to animals expressing tTA under
control of the CamK2a promoter

Disease

Reference

(Jankowsky et al. 2005)

AD/FTLD Pronuclear injection of transgenes containing


Thy 1.2 driven APP Swe FAD and tau P301L
FTLD mutations were co-injected into oocytes
from homozygous PS1 M146 V knock-in mice
to generate 3xTg mice

Elevated A 40 and 42, age-dependent


parenchymal amyloid plaques, and
tau pathology development along
with synaptic dysfunction and
behavioral deWcits

(Oddo et al. 2003)

FTLD

Development of progressive motor


neuron disease; tangle-like
accumulations containing tau in
brain and spinal cord with gliosis
and neuronal loss including 50% loss
of anterior horn motor neurons

(Lewis et al. 2000)

Pronuclear injection of PrP-driven four repeat


human tau containing the P301L mutation
associated with FTDP-17

FTLD/ALS Pan-neuronal constitutive overexpression


of TDP-43 or mutant TDP-43 (A315T)
via pronuclear injection of transgenic
cassettes using PrP and Thy1.2 promoters

Expression of TDP-43 wt and mutant


(Gendron and Petrucelli 2011;
proteins results in variable pathological
Shan et al. 2010;
phenotypes depending on the animal model Stallings et al. 2010;
Wegorzewska and
Baloh 2011;
Wegorzewska et al. 2009;
Wils et al. 2010;
Xu et al. 2010)

123

550

Hum Genet (2012) 131:535563

Table 1 continued
Strategy

Pathology and other Wndings

Reference

FTLD/ALS

Forebrain-targeted Tet-OV conditional expression


of TDP-43. Bigenic mice were created with
cassettes harboring the Camk2a promoter
driving the expression of Tet-OV tTA and
the tetracycline-responsive promoter to
drive hTDP-43 (tetO-hTDP). hTDP-43 was
preferentially induced (in the absence of
doxycycline) in forebrain neurons

Selective degeneration of hippocampal


dentate gyrus and neocortical neurons
along with dysregulation and loss of
endogenous TDP-43 and presence of
rare phosphorylated and ubiquitinated
TDP-43 inclusions in aVected neurons.
Transgenic mice develop a motor
phenotype associated with spasticity

(Igaz et al. 2011)

ALS

Pronuclear injection of a human SOD1


transgene containing the G93A
substitution associated with familial ALS

Hemizygous transgenic mice develop


motor neuron disease associated with
progressive limb paralysis and
death by 56 months of age

(Gurney et al. 1994)

ALS/AD

CD11b-TKmt-30 mice contain a transgenic


cassette in which the HSV-1 TKmt-30
gene is expressed under the control of
the CD11b promoter, which directs high
levels of expression in macrophages.
Proliferating microglial cells in the brain
can be selectively eliminated by
ganciclovir treatment

After ganciclovir treatment, selective


ablation of proliferating microglial
cells occurs. Ablation of proliferating
microglia does not aVect motor neuron
degeneration in a mouse model of
ALS but causes increased amyloid
deposition in a mouse model of AD,
suggesting that microglia play a
critical role in restricting AD
associated senile plaque formation

(Dziennis et al. 1995;


Gowing et al. 2008;
Simard et al. 2006)

PD

Transgenesis with a BAC harboring the dopamine


transporter (Slc6a3) gene retroWtted with
a FLAG-tagged park2 truncated mutant
(Q311X) in exon 2

Mutant parkin-expressing animals exhibit


age-dependent dopaminergic neuron
degeneration and accumulation
of -synuclein in substantia
nigra (SN) neurons

(Lu et al. 2009)

PD

Transgenesis with retroWtted BAC


constructs containing the entire
FLAG-tagged wt or mutant (G2019S)
sequence of the leucine-rich repeat
kinase 2 (LRRK2) gene

Transgenic LRRK2-wt mice had elevated


striatal dopamine (DA) release with
unaltered DA uptake or tissue content,
mice were hyperactive and showed
enhanced performance in motor
function tests. LRRK2-G2019S transgenics
showed an age-dependent decrease
in striatal DA content, decreased striatal
DA release and uptake. LRRK2-G2019S
overexpression was not associated with
loss of dopaminergic neurons in
substantia nigra or degeneration of
nigrostriatal terminals by 12 months

(Li et al. 2010)

PD

Conditional knockout of the 19S RP


subunit, Psmc1 gene involved in 26S
proteasomal function in forebrain/substantia
nigra neurons using the Cre/loxP system.
Because Psmc1 null mice are embryonic
lethal, Psmc1 conditional knock-out mice
were generated by introducing loxP sites
in introns 1 and 3 in ES cells. Spatial ablation
of Psmc1 was performed by crossing the
Xoxed transgenic mice (Psmc1X/X) with a Cre
deleter strain expressing Cre recombinase
under the control of either the Camk2a
promoter or the tyrosine hydroxylase locus

Neurodegeneration, ubiquitin, and


-synuclein pathology with Lewy-like
inclusions apparent in Psmc1X/X Cre
animals. Inclusion bodies found
in mitochondria

(Bedford et al. 2008)

PD

Selective Tet-On inducible monoamine oxidase


B (MAO-B) in astrocytes. Two transgenic
cassettes were injected by pronuclear injection.
One harbored the MAO-B cDNA under the
control of a bidirectional Tet-inducible promoter
(also regulating the expression of  galactosidase)
and the other driving expression of the
tet-responsive reverse transactivator, rtTA
under the control of the mouse GFAP promoter

Dox-treatment induces hMAO-B and


-galactosidase in GFAP+ cells causing
altered spontaneous locomotor activity
associated with progressive degeneration
of TH+ SN neurons and reduced striatal DA

(Mallajosyula
et al. 2008)

Disease

123

Hum Genet (2012) 131:535563

551

Table 1 continued
Strategy

Pathology and other Wndings

PD

Human -synuclein (wild-type or mutant A53T


and A30P) cDNAs were included in
a transgenic cassette harboring the promoter,
5 intronic and 3 untranslated sequences
of the murine prion protein gene

Transgenic expression of A53T -synuclein


(Lee et al. 2002)
(but not the wild-type nor the A30P proteins)
resulted in the development of adult onset
neurodegenerative disease with a
progressive motor dysfunction together
with accumulations of -synuclein and ubiquitin

HD

Pronuclear injection of a 1.9-kb transgenic


containing 1 kb of 5 UTR
(promoter sequences), Htt exon 1
carrying expanded CAG repeats
(130 units) and the Wrst
262 bp of intron 1

Transgenic mice (R6/2) manifest a


rapidly progressive disease similar to
the juvenile form of HD in humans.
Overt behavioral symptoms apparent
by 5-6 weeks of age. Motor and learning
impairments are present. Limb
clasping, weight loss, and death occur
by 15 weeks. Nuclear inclusions present
particularly in the striatum and
CA1 region of the hippocampus

(Carter et al. 1999;


Davies et al. 1997;
Lione et al. 1999;
Mangiarini et al. 1996)

HD

YAC transgenesis with full-length normal


and mutant (72 and 128 repeat) human Htt

Mutant Htt results in abnormal behavior


and causes selective degeneration of
medium-sized spiny neurons in the lateral
striatum. An earlier and more severe
pathology found in YAC 128 transgenic
mice expressing longer CAG repeats.
Progressive cognitive dysfunction
and mood disturbance precede motor
abnormalities. Selective atrophy and
neuronal loss occurs in the striatum
and cortex of YAC128 transgenics

(Hodgson et al. 1999;


Slow et al. 2003)

HD

Knock-in mutant Htt using homologous


recombination in ES cells

Expression of mutant Htt in its native


genomic context. Presence of nuclear
staining and microaggregates early in
the course of the disease. Early overt
behavioral changes seen and strength
of the phenotype generally correlated
with the number of CAG repeats (200)

(Heng et al. 2009;


Levine et al. 2004;
Lin et al. 2001;
Raymond et al. 2011;
Shelbourne et al. 1999;
Wheeler et al. 2000;
White et al. 1997)

HD

BAC transgenesis with full-length


mutant Htt with 97 CAG repeats

Transgenic mice (BACHD) exhibit progressive


motor deWcits associated with later
neuropathology in striatum and cortex.
Pathology occurs without evidence
of aggregated mutant Htt

(Gray et al. 2008)

Disease

Reference

Rat Rattus norvegicus


AD

Neuronal expression of normal and mutant


APP under the control of either synapsin-1,
PDGF or ubiquitin C (UbC)
promoters. Pronuclear
injection of transgenic cassettes

No -amyloid plaques unless two independent


FAD mutant transgenic lines are bred
to double homozygosity. High-level
expression and the presence of an
FAD mutation necessary to induce
age-dependent plaque deposition

(Bugos et al. 2009; Clarke


et al. 2007; Echeverria
et al. 2004; Agca et al.
2008; Flood et al. 2009;
Folkesson et al. 2007;
Liu et al. 2008a;
Ruiz-Opazo et al. 2004)

AD

Pronuclear injection of a cassette harboring the


hAPP751 cDNA containing the Swedish
and Indiana mutations under the control
of the Thy1.2 promoter

Pan-neuronal expression of human


APP751 was found. Homozygous
transgenic animals produce extracellular
A deposits by 6 months of age associated
with glial activation and dystrophic neurites.
Cognitive functions are altered
as early as 3 months of age

(Leon et al. 2010)

Constitutive expression of the mutant


TDP-43 causes early postnatal death
in transgenic rats. Founders develop
weakness in the limbs and do not
survive past 29 days

(Zhou et al. 2010)

FTLD/ALS Transgenesis with a 22-kb normal and


retroWtted-mutant (M337 V) human
TDP-43 gene extracted from a BAC

123

552

Hum Genet (2012) 131:535563

Table 1 continued
Disease
FTLD/ALS

Strategy

Pathology and other Wndings

Reference

Tet-OV inducible system for TDP-43 expression.


tTA protein expression controlled by a CMV
enhancer fused to the chicken  actin promoter (CAG).
Normal and mutant TDP-43 cDNAs were expressed
under the control of a tTA-dependent promoter
constructed by the juxtaposition of multiple t
etracycline-responsive elements (TRE, tet-O)
upstream from the minimal CMV promoter

Overexpression of mutant TDP-43 but


not wt protein induce progressive
widespread neurodegeneration
aVecting predominantly the motor
system with denervation atrophy
of skeletal muscles

(Zhou et al. 2009, 2010)

of A3-42 after cleavage (Link 1995). Expression of this


human A42 minigene in muscle cells resulted in progressive paralysis and staining of muscle cells with a human
A42 speciWc antibody revealed A42 deposits that reacted
with Congo red and thioXavin S. Ultrastructural studies,
however, revealed that the deposits were intracytoplasmic
rather than extracellular as are amyloid plaques found in
AD (Ewald and Li 2010). Pan-neuronal expression of
human A42 has also been associated with amyloid deposits and deWcits in odorant preference-associated learning
behavior, serotonin-controlled behaviors, experiencedependent learning and egg laying behavior (Dosanjh et al.
2010). Transgenic C. elegans has also been used to investigate how speciWc residues within human A aVect Wbril
formation (Ewald and Li 2010).
Models such as those developed in C. elegans may be
questioned for their reliance on the isolated overexpression
of a non-native protein that is not naturally processed. Overexpression of APP in Downs syndrome is associated with
AD pathology in humans (Zigman et al. 1996) and families
have been identiWed in which a duplication of the APP gene
leads to FAD presumably due to overexpression of wild type
APP (Kasuga et al. 2009; Sleegers et al. 2006). Thus, there
is a precedent for APP overexpression leading to AD in
humans. Yet there is no evidence for APP overexpression in
the most common sporadic forms of AD. The lack of extracellular amyloid deposits can also be seen as a limitation of
these models. On the other hand current models of the amyloid hypothesis postulate that soluble forms of oligomeric
A rather than plaque amyloid are the critical toxic species
(Lublin and Gandy 2010) and indeed after only several
hours of A induction in C. elegans, paralysis occurs without detectable A deposits (Drake et al. 2003). Toxicity has
been correlated with levels of oligomeric A (Wu et al.
2006), making it plausible that the changes are physiologically relevant. Induction of oxidative stress has long been
postulated as one of the key steps leading to injury in the
amyloid cascade and transgenic worms expressing human
A42 exhibit signs of oxidative stress prior to deposition of
A (Drake et al. 2003), suggesting that soluble A42 may
induce oxidative stress in vivo in C. elegans.

123

Like C. elegans, Drosophila has a single homolog of


APP. Flies deWcient for Drosophila APP (dAPPL) exhibit
behavioral abnormalities that can be rescued with a human
APP transgene and dAPPL has also been implicated in neurite outgrowth and synapse formation (Iijima et al. 2008).
Drosophila also contains orthologs of the  and -secretases although -secretase activity in Drosophila is low. In
addition, while dAPPL shares about 30% amino acid identity with the human protein, like C. elegans apl-1 it lacks a
homologous A region although it has been suggested that
an equivalent A-peptide exists that upon cleavage can
generate a neurotoxic peptide capable of generating amyloid deposits (Carmine-Simmen et al. 2009).
In transgenic Drosophila human A peptides have been
introduced either by overexpressing full-length human APP
or human A peptides. To create a partially humanized system Greeve et al. (2004) engineered triple transgenic Xies
that express human wild type APP, human BACE and also
overexpresses wild-type Drosophila presenilin (dPsn) or
dPsn with point mutations corresponding to human FAD
mutations. The lines produced exhibited modest elevations
of A40 and A42 peptides. Triple transgenic Xies developed thioXavin S stained amyloid plaques in the retina and
exhibited an age-dependent degeneration of the photoreceptors that was enhanced in Xies expressing the FAD
related dPsn mutations. This phenotype could be suppressed by genetically reducing endogenous Drosophila
dPsn or by the treatment with  or -secretase inhibitors.
Others have taken the approach used in C. elegans of
introducing isolated human A peptides fused to an N-terminal signal peptide directly into neurons (Crowther et al.
2005; Finelli et al. 2004; Iijima et al. 2004, 2008). Both
A40 and A42 peptides accumulate during aging in the Xy
brain, but only A42 forms amyloid deposits in brain and
retina (Finelli et al. 2004; Iijima et al. 2004). Age-dependent short-term memory defects are detected in both A40
and A42 expressing Xies but at later ages, A42, but not
A40, induces locomotor deWcits, as well as causes an agedependent neurodegeneration and premature death. These
models have also been associated with intraneuronal accumulation of A42 (Iijima et al. 2004, 2008).

Hum Genet (2012) 131:535563

As with similar approaches used in C. elegans, strategies


that rely on the isolated overexpression of a non-native protein may be considered artiWcial. Indeed as discussed below
overexpression strategies even in mammalian systems may
be questioned as artiWcial. Organisms like D. melanogaster
or C. elegans also lack a mammalian like immune system
which is thought to play a role in the development of AD
pathology. Yet if the essential tenets of the amyloid hypothesis are correct then whether overexpressed or not any system that exhibits a robust amyloid dependent pathology
may be suitable to model disease pathogenesis or search for
disease modiWers. Retinal toxicity for example has been
widely used as a phenotypic readout for screens in Drosophila aimed at identifying genetic modiWers of neurodegenerative disease.
Transgenic modeling has been most aggressively pursued in mice and transgenic mice now exist that mimic
nearly the full range of AD-related pathology (Table 1;
Elder et al. 2010a). These models have suggested new
insights into the pathophysiology of AD and have been the
source of novel therapeutic approaches, most notably the
development of immunotherapy (Wisniewski and Boutajangout 2010). These models, however, raise a number of
issues as well. Firstly, like the models in C. elegans and
Drosophila it is clear that their success has depended on the
overexpression of APP transgenes containing FAD-associated mutations at levels that are not physiological. Indeed,
despite the fact that a number of APP wild transgenic lines
have been created only one transgenic mouse line overexpressing wild-type human APP has been reported as
developing amyloid plaques (Herzig et al. 2004) and for
plaque pathology to develop it seems to require the presence of an APP FAD.
In this context it is interesting to compare APP FAD
over-expression models to APP knock-in mice. For example, the APP Swedish mutation (associated with FAD) has
been introduced into the mouse APP gene (Reaume et al.
1996). The three amino acid diVerences between mouse and
human APPs in the A region were humanized such that
these mice produce human A. These mice should represent the most natural model of human FAD in the mouse.
Yet despite increased A production, no amyloid plaques
or neuropathology have been observed to develop.
The models have also been troubled by the diYculty of
producing the full spectrum of AD pathology including
neuronal loss. For example neither of the commonly studied PDAPP nor Tg2576 mice exhibit neuronal loss despite
having extensive amyloid deposition (Irizarry et al. 1997a,
b). More substantial neuronal loss has been reported in
mice expressing multiple PS1 and APP mutations (Casas
et al. 2004; Oakley et al. 2006; Schmitz et al. 2004) and
gross neuronal loss has been reported in mice expressing
three APP and two PS1 FAD mutations (Oakley et al.

553

2006). Thus, neuronal loss can be induced in the mouse but


only it would seem by combining multiple mutations that
individually are suYcient to cause disease in humans.
Another problem with the models has been the general
diYculty of recreating the characteristic cytoskeletal
pathology found in AD (Elder et al. 2010a). Various suggestions have been made for the diYculty of inducing neuroWbrillary tangle-like lesions in the mouse including
diVerences between human and mouse tau, although the
reasons remain to be fully explained. Recently mice that
exhibit NTF-like lesions and plaques have been produced
by combining FAD mutations with mutant forms of tau
associated with FTLD. In the 3xTg-AD model Oddo et al.
(2003) generated triple transgenic mice by co-injecting two
transgenes containing the FAD APP Swedish mutation and
the P301L mutations associate with FTLD into fertilized
eggs harvested from PS1M146V knock-in mice. The resulting transgenic lines thus expressed mutations in APP and
tau from exogenous transgenes combined with a PS1 FAD
mutation from the endogenous mouse gene. With aging
these mice exhibited increased A40 and 42 levels. Intraneuronal A accumulated and the mice developed amyloid
plaques and NFT-like lesions that immunostained with
antibodies to conformationally altered tau epitopes. Functionally, 3xTg-AD mice developed age-dependent synaptic
dysfunction including altered long-term potentiation and
deWcits in spatial memory (Billings et al. 2005; Oddo et al.
2003).
The 3xTg-AD model thus exhibits a broader spectrum of
AD pathology with the much-sought-after plaque and tangle pathology. However, they represent a composite of two
distinct diseases that do not naturally occur together.
Plaque development is almost certainly driven by the APP
and PS1 FAD mutations while tangle-like pathology is
driven by the tau mutations. Indeed in 3xTg-AD mice, the
pathologies arise in spatially distinct patterns with A
deposition starting in neocortex and appearing later in hippocampus, while tau pathology progresses in the opposite
direction, beginning in hippocampus and appearing later in
neocortex (Oddo et al. 2003). The two pathologies do
appear, however, to interact. Intracerebral injections of
anti-A antibodies into the hippocampus of 3xTg-AD mice
not only reduce A accumulation but cause clearance of
early stage tau pathology (Oddo et al. 2004).
Due to the lack of a plaque pathology, mice expressing
only presenlin FAD mutations have been less studied than
APP or APP/PS FAD mutant transgenic mice which
develop robust plaque pathology (Elder et al. 2010a). Why
presenilin FAD mutant transgenic mice do not develop a
more AD-like pathology is unclear given the potency of
especially the presenlin-1 FAD mutations in humans, which
typically induce disease at younger ages than APP FAD
mutations (Bertram and Tanzi 2004a, b; Elder et al. 2010b).

123

554

Why single transgenic PS1 and PS2 mice fail to develop


plaque pathology is not entirely clear, but may relate to the
generally lower levels of A42 found in single PS transgenics compared to APP overexpressing lines as well as the
lack of elevation of A40 in PS transgenics. It may also
relate to diVering aggregation properties of mouse and
human A (Jankowsky et al. 2007). Yet despite the lack of
a plaque and tangle pathology presenilin FAD mutant mice
exhibit a wide range of anatomic, electrophysiological and
biochemical changes that mimic many of the features of
AD (Elder et al. 2010b). Rat models while becoming more
available appear to suVer the same limitations as models in
the mouse requiring high-level expression of an APP FAD
mutation to induce signiWcant plaque pathology (Table 1;
Bugos et al. 2009; Clarke et al. 2007; Echeverria et al.
2004; Agca et al. 2008; Flood et al. 2009; Folkesson et al.
2007; Leon et al. 2010; Liu et al. 2008a; Ruiz-Opazo et al.
2004).

Hum Genet (2012) 131:535563

its orthologues protect against neurodegeneration in both


invertebrate and mammalian models of neurodegenerative
disease including Alzheimers disease and ALS (Gan and
Tang 2010; Kim et al. 2007; Pallas et al. 2009). In a transgenic mouse model of ALS, deletion of p66Shc ameliorates
mitochondrial dysfunction and delays disease onset as well
as improves motor performance and prolongs survival
(Pesaresi et al. 2011). Inhibition of mTOR by rapamycin
abolishes cognitive deWcits and reduces -amyloid levels in
a mouse model of Alzheimers disease (Spilman et al.
2010). Circadian rhythms generate oscillations in physiology
and behavior. Circadian clock gene expression is often
altered in human disease states and transgenic mice with
mutations in clock genes develop premature aging (Khapre
et al. 2010). Circadian dysfunctions are found in transgenic
mouse models of AD (Wisor et al. 2005; Sterniczuk et al.
2010), PD (Kudo et al. 2011a) and HD (Kudo et al. 2011b).
These and many other factors related to how organisms age
may inXuence the phenotypic expression of a neurodegenerative process in a transgenic model.

The factor of aging in transgenic models


of neurodegeneration
Conclusions
Many factors may aVect the ability of transgenic organisms
to model human diseases. These include obvious anatomical and morphological diVerences between humans and
model organisms as well as possibly diVerences in critical
proteins such as tau or APP. Another factor, however, is the
process of aging itself. All age-related neurodegenerative
diseases develop in the context of aging. Aging as a comorbid process is believed to be the result of the gradual
loss of metabolic and genetic assets necessary to preserve
the integrity and functionality of all cellular components. It
has been assumed (Minois et al. 2010) that senescence is
under the control of many genes (Ljubuncic and Reznick
2009), and is aVected by accumulation of deleterious
somatic mutations (Best 2009). The factor of pleiotrophic
antagonism has also been invoked whereby certain gene
functions that may be beneWcial in early life may be deleterious as the organism ages (Williams 1957).
Due to diVerences in cellular turnover, metabolic rates,
life span, oxidative stress, diet and other environmental factors as well as species-speciWc genetic factors, aging may
be modeled insuYciently in relation to development of neurodegenerative diseases in transgenic animal models. Nevertheless, it is clear that aging inXuences considerably the
development of neurodegenerative processes. Over activation or disruption of key lifespan determinant pathways
such as SIR2/SIRT1 (NAD-dependent class III histone
deacetylases), p66Shc (66-kDa isoform of the growth factor
adapter Shc) and mTOR (mammalian target of rapamycin)
clearly aVect the development of neurodegenerative diseases in model organisms. For example, SIR2/SIRT1 and

123

Advances in the genetic tools available across a wide range


of species have made transgenic modeling more versatile
than ever before. While the mouse remains the most widely
used model system, signiWcant eVorts have been made in a
variety of other species. In particular, recent advances in
the tools available for genetic manipulation in the rat have
lead to increased interest in this species as a model system.
These models have improved understanding of disease
pathogenesis and, in some cases, have lead to new therapeutic approaches. They will continue to play central roles
for years to come in both preclinical testing and as tools for
developing insight into the biological basis of human neurodegenerative diseases.
Acknowledgments Supported by the Alzheimers Association
(IIRG-07-57318), Department of Veterans AVairs (1 I01 BX00034201) and the National Institute on Aging (P50 AG005138).

References
Abbott A (2004) Laboratory animals: the Renaissance rat. Nature
428:464466
Agca C, Fritz JJ, Walker LC, Levey AI, Chan AW, Lah JJ, Agca Y
(2008) Development of transgenic rats producing human betaamyloid precursor protein as a model for Alzheimers disease:
transgene and endogenous APP genes are regulated tissue-speciWcally. BMC Neurosci 9:28
Albanese C, Hulit J, Sakamaki T, Pestell RG (2002) Recent advances
in inducible expression in transgenic mice. Semin Cell Dev Biol
13:129141

Hum Genet (2012) 131:535563


Amacher SL (2008) Emerging gene knockout technology in zebraWsh:
zinc-Wnger nucleases. Brief Funct Genomic Proteomic 7:460464
Amsterdam A, Nissen RM, Sun Z, Swindell EC, Farrington S, Hopkins
N (2004) IdentiWcation of 315 genes essential for early zebraWsh
development. Proc Natl Acad Sci USA 101:1279212797
An W, Han JS, Wheelan SJ, Davis ES, Coombes CE, Ye P, Triplett C,
Boeke JD (2006) Active retrotransposition by a synthetic L1 element in mice. Proc Natl Acad Sci USA 103:1866218667
Anastassiadis K, Fu J, Patsch C, Hu S, Weidlich S, Duerschke K,
Buchholz F, Edenhofer F, Stewart AF (2009) Dre recombinase,
like Cre, is a highly eYcient site-speciWc recombinase in E. coli,
mammalian cells and mice. Dis Model Mech 2:508515
Arguello PA, Gogos JA (2010) Cognition in mouse models of schizophrenia susceptibility genes. Schizophr Bull 36:289300
Asakawa K, Suster ML, Mizusawa K, Nagayoshi S, Kotani T, Urasaki
A, Kishimoto Y, Hibi M, Kawakami K (2008) Genetic dissection
of neural circuits by Tol2 transposon-mediated Gal4 gene and enhancer trapping in zebraWsh. Proc Natl Acad Sci USA 105:1255
1260
Ash PE, Zhang YJ, Roberts CM, Saldi T, Hutter H, Buratti E, Petrucelli L, Link CD (2010) Neurotoxic eVects of TDP-43 overexpression in C. elegans. Hum Mol Genet 19:32063218
Auluck PK, Chan HY, Trojanowski JQ, Lee VM, Bonini NM (2002)
Chaperone suppression of alpha-synuclein toxicity in a Drosophila model for Parkinsons disease. Science 295:865868
Bai Q, Garver JA, Hukriede NA, Burton EA (2007) Generation of a
transgenic zebraWsh model of tauopathy using a novel promoter
element derived from the zebraWsh eno2 gene. Nucleic Acids Res
35:65016516
Bateman JR, Lee AM, Wu CT (2006) Site-speciWc transformation of
Drosophila via phiC31 integrase-mediated cassette exchange.
Genetics 173:769777
Beaudet L, Cote F, Houle D, Julien JP (1993) DiVerent posttranscriptional controls for the human neuroWlament light and heavy genes
in transgenic mice. Brain Res Mol Brain Res 18:2331
Bedford L, Hay D, Devoy A, Paine S, Powe DG, Seth R, Gray T, Topham I, Fone K, Rezvani N, Mee M, Soane T, LayWeld R, Sheppard PW, Ebendal T, Usoskin D, Lowe J, Mayer RJ (2008)
Depletion of 26S proteasomes in mouse brain neurons causes
neurodegeneration and Lewy-like inclusions resembling human
pale bodies. J Neurosci 28:81898198
Bertram L, Tanzi RE (2004a) Alzheimers disease: one disorder, too
many genes? Hum Mol Genet 13 Spec No 1:R13541
Bertram L, Tanzi RE (2004b) The current status of Alzheimers disease genetics: what do we tell the patients? Pharmacol Res
50:385396
Best BP (2009) Nuclear DNA damage as a direct cause of aging. Rejuvenation Res 12:199208
Beumer K, Bhattacharyya G, Bibikova M, Trautman JK, Carroll D
(2006) EYcient gene targeting in Drosophila with zinc-Wnger
nucleases. Genetics 172:23912403
Beumer KJ, Trautman JK, Bozas A, Liu JL, Rutter J, Gall JG, Carroll
D (2008) EYcient gene targeting in Drosophila by direct embryo
injection with zinc-Wnger nucleases. Proc Natl Acad Sci USA
105:1982119826
Bezard E, Przedborski S (2011) A tale on animal models of Parkinsons disease. Mov Disord 26:9931002
Bill BR, Petzold AM, Clark KJ, Schimmenti LA, Ekker SC (2009) A
primer for morpholino use in zebraWsh. ZebraWsh 6:6977
Billings LM, Oddo S, Green KN, McGaugh JL, LaFerla FM (2005)
Intraneuronal Abeta causes the onset of early Alzheimers disease-related cognitive deWcits in transgenic mice. Neuron
45:675688
Bischof J, Basler K (2008) Recombinases and their use in gene activation, gene inactivation, and transgenesis. Methods Mol Biol
420:175195

555
Boch J, Bonas U (2010) Xanthomonas AvrBs3 family-type III eVectors: discovery and function. Annu Rev Phytopathol 48:419436
Boch J, Scholze H, Schornack S, Landgraf A, Hahn S, Kay S, Lahaye
T, Nickstadt A, Bonas U (2009) Breaking the code of DNA binding speciWcity of TAL-type III eVectors. Science 326:15091512
Bonini NM, Fortini ME (2003) Human neurodegenerative disease
modeling using Drosophila. Annu Rev Neurosci 26:627656
Borchelt DR, Davis J, Fischer M, Lee MK, Slunt HH, Ratovitsky T,
Regard J, Copeland NG, Jenkins NA, Sisodia SS, Price DL (1996)
A vector for expressing foreign genes in the brains and hearts of
transgenic mice. Genet Anal 13:159163
Bosze Z, Hiripi L, Carnwath JW, Niemann H (2003) The transgenic
rabbit as model for human diseases and as a source of biologically
active recombinant proteins. Transgenic Res 12:541553
Brault V, Besson V, Magnol L, Duchon A, Herault Y (2007) Cre/loxPmediated chromosome engineering of the mouse genome. Handb
Exp Pharmacol 178:2948
Brenner M, Kisseberth WC, Su Y, Besnard F, Messing A (1994) GFAP
promoter directs astrocyte-speciWc expression in transgenic mice.
J Neurosci 14:10301037
Buchholz F, Angrand PO, Stewart AF (1998) Improved properties of
FLP recombinase evolved by cycling mutagenesis. Nat Biotechnol 16:657662
Bugos O, Bhide M, Zilka N (2009) Beyond the rat models of human
neurodegenerative disorders. Cell Mol Neurobiol 29:859869
Calhoun ME, Burgermeister P, Phinney AL, Stalder M, Tolnay M,
Wiederhold KH, Abramowski D, Sturchler-Pierrat C, Sommer B,
Staufenbiel M, Jucker M (1999) Neuronal overexpression of mutant amyloid precursor protein results in prominent deposition of
cerebrovascular amyloid. Proc Natl Acad Sci USA 96:14088
14093
Campbell WA, Yang H, Zetterberg H, Baulac S, Sears JA, Liu T,
Wong ST, Zhong TP, Xia W (2006) ZebraWsh lacking Alzheimer
presenilin enhancer 2 (Pen-2) demonstrate excessive p53-dependent apoptosis and neuronal loss. J Neurochem 96:14231440
Carbery ID, Ji D, Harrington A, Brown V, Weinstein EJ, Liaw L, Cui
X (2010) Targeted genome modiWcation in mice using zinc-Wnger
nucleases. Genetics 186:451459
Carlson CM, Largaespada DA (2005) Insertional mutagenesis in mice:
new perspectives and tools. Nat Rev Genet 6:568580
Carmine-Simmen K, Proctor T, Tschape J, Poeck B, Triphan T, Strauss
R, Kretzschmar D (2009) Neurotoxic eVects induced by the Drosophila amyloid-beta peptide suggest a conserved toxic function.
Neurobiol Dis 33:274281
Carter RJ, Lione LA, Humby T, Mangiarini L, Mahal A, Bates GP,
Dunnett SB, Morton AJ (1999) Characterization of progressive
motor deWcits in mice transgenic for the human Huntingtons disease mutation. J Neurosci 19:32483257
Casas C, Sergeant N, Itier JM, Blanchard V, Wirths O, van der Kolk
N, Vingtdeux V, van de Steeg E, Ret G, Canton T, Drobecq H,
Clark A, Bonici B, Delacourte A, Benavides J, Schmitz C, Tremp
G, Bayer TA, Benoit P, Pradier L (2004) Massive CA1/2 neuronal
loss with intraneuronal and N-terminal truncated Abeta42 accumulation in a novel Alzheimer transgenic model. Am J Pathol
165:12891300
Changeux JP (2010) Nicotine addiction and nicotinic receptors: lessons from genetically modiWed mice. Nat Rev Neurosci 11:389
401
Cheah SS, Behringer RR (2001) Contemporary gene targeting strategies for the novice. Mol Biotechnol 19:297304
Cheng RK, Jesuthasan S, Penney TB (2011) Time for zebraWsh. Front
Integr Neurosci 5:40
Chew SK, Rad R, Futreal PA, Bradley A, Liu P (2011) Genetic screens
using the piggyBac transposon. Methods 53:366371
Chishti MA, Yang DS, Janus C, Phinney AL, Horne P, Pearson J,
Strome R, Zuker N, Loukides J, French J, Turner S, Lozza G,

123

556
Grilli M, Kunicki S, Morissette C, Paquette J, Gervais F, Bergeron C, Fraser PE, Carlson GA, George-Hyslop PS, Westaway D
(2001) Early-onset amyloid deposition and cognitive deWcits in
transgenic mice expressing a double mutant form of amyloid
precursor protein 695. J Biol Chem 276:2156221570
Choo BG, Kondrichin I, Parinov S, Emelyanov A, Go W, Toh WC,
Korzh V (2006) ZebraWsh transgenic Enhancer TRAP line database (ZETRAP). BMC Dev Biol 6:5
Christian M, Cermak T, Doyle EL, Schmidt C, Zhang F, Hummel A,
Bogdanove AJ, Voytas DF (2010) Targeting DNA double-strand
breaks with TAL eVector nucleases. Genetics 186:757761
Clarke J, Thornell A, Corbett D, Soininen H, Hiltunen M, Jolkkonen J
(2007) Overexpression of APP provides neuroprotection in the
absence of functional beneWt following middle cerebral artery
occlusion in rats. Eur J Neurosci 26:18451852
Crabtree DM, Zhang J (2011) Genetically engineered mouse models of
Parkinsons disease. Brain Res Bull. doi:10.1016/jbrainresbull.2011.07.019
Crews L, Rockenstein E, Masliah E (2010) APP transgenic modeling
of Alzheimers disease: mechanisms of neurodegeneration and
aberrant neurogenesis. Brain Struct Funct 214:111126
Cronin CA, Gluba W, Scrable H (2001) The lac operator-repressor system is functional in the mouse. Genes Dev 15:15061517
Crowther DC, Kinghorn KJ, Miranda E, Page R, Curry JA, Duthie FA,
Gubb DC, Lomas DA (2005) Intraneuronal Abeta, non-amyloid
aggregates and neurodegeneration in a Drosophila model of Alzheimers disease. Neuroscience 132:123135
Cui X, Ji D, Fisher DA, Wu Y, Briner DM, Weinstein EJ (2011) Targeted integration in rat and mouse embryos with zinc-Wnger nucleases. Nat Biotechnol 29:6467
Davies SW, Turmaine M, Cozens BA, DiFiglia M, Sharp AH, Ross
CA, Scherzinger E, Wanker EE, Mangiarini L, Bates GP (1997)
Formation of neuronal intranuclear inclusions underlies the neurological dysfunction in mice transgenic for the HD mutation.
Cell 90:537548
Davis MW, Morton JJ, Carroll D, Jorgensen EM (2008) Gene activation using FLP recombinase in C. elegans. PLoS Genet
4:e1000028
De Gasperi R, Rocher AB, Sosa MA, Wearne SL, Perez GM, Friedrich
VL Jr, Hof PR, Elder GA (2008) The IRG mouse: a two-color Xuorescent reporter for assessing Cre-mediated recombination and
imaging complex cellular relationships in situ. Genesis 46:308317
Deiters A, Yoder JA (2006) Conditional transgene and gene targeting
methodologies in zebraWsh. ZebraWsh 3:415429
Deuschle U, Meyer WK, Thiesen HJ (1995) Tetracycline-reversible
silencing of eukaryotic promoters. Mol Cell Biol 15:19071914
Distel M, Wullimann MF, Koster RW (2009) Optimized Gal4 genetics
for permanent gene expression mapping in zebraWsh. Proc Natl
Acad Sci USA 106:1336513370
Dolphin CT, Hope IA (2006) Caenorhabditis elegans reporter fusion
genes generated by seamless modiWcation of large genomic DNA
clones. Nucleic Acids Res 34:e72
Dosanjh LE, Brown MK, Rao G, Link CD, Luo Y (2010) Behavioral
phenotyping of a transgenic Caenorhabditis elegans expressing
neuronal amyloid-beta. J Alzheimers Dis 19:681690
Drake J, Link CD, ButterWeld DA (2003) Oxidative stress precedes
Wbrillar deposition of Alzheimers disease amyloid beta-peptide
(142) in a transgenic Caenorhabditis elegans model. Neurobiol
Aging 24:415420
Draper BW, McCallum CM, Stout JL, Slade AJ, Moens CB (2004) A
high-throughput method for identifying N-ethyl-N-nitrosourea
(ENU)-induced point mutations in zebraWsh. Methods Cell Biol
77:91112
DuV K, Eckman C, Zehr C, Yu X, Prada CM, Perez-tur J, Hutton M,
Buee L, Harigaya Y, Yager D, Morgan D, Gordon MN, Holcomb
L, Refolo L, Zenk B, Hardy J, Younkin S (1996) Increased

123

Hum Genet (2012) 131:535563


amyloid-beta42(43) in brains of mice expressing mutant presenilin 1. Nature 383:710713
DuVy JB (2002) GAL4 system in Drosophila: a Xy geneticists Swiss
army knife. Genesis 34:115
Dziennis S, Van Etten RA, Pahl HL, Morris DL, Rothstein TL, Blosch
CM, Perlmutter RM, Tenen DG (1995) The CD11b promoter
directs high-level expression of reporter genes in macrophages in
transgenic mice. Blood 85:319329
Echeverria V, Ducatenzeiler A, Alhonen L, Janne J, Grant SM, Wandosell F, Muro A, Baralle F, Li H, DuV K, Szyf M, Cuello AC
(2004) Rat transgenic models with a phenotype of intracellular
Abeta accumulation in hippocampus and cortex. J Alzheimers
Dis 6:209219
Ekker SC (2008) Zinc Wnger-based knockout punches for zebraWsh
genes. ZebraWsh 5:121123
Elder GA, Friedrich VL Jr, Liang Z, Li X, Lazzarini RA (1994) Enhancer trapping by a human mid-sized neuroWlament transgene
reveals unexpected patterns of neuronal enhancer activity. Brain
Res Mol Brain Res 26:177188
Elder GA, Gama Sosa MA, De Gasperi R (2010a) Transgenic mouse
models of Alzheimers disease. Mt Sinai J Med 77:6981
Elder GA, Gama Sosa MA, De Gasperi R, Dickstein DL, Hof PR
(2010b) Presenilin transgenic mice as models of Alzheimers disease. Brain Struct Funct 214:127143
Elia AJ, Parkes TL, Kirby K, St George-Hyslop P, Boulianne GL, Phillips JP, Hilliker AJ (1999) Expression of human FALS SOD in
motorneurons of Drosophila. Free Radic Biol Med 26:13321338
Emelyanov A, Parinov S (2008) Mifepristone-inducible LexPR system
to drive and control gene expression in transgenic zebraWsh. Dev
Biol 320:113121
Emelyanov A, Gao Y, Naqvi NI, Parinov S (2006) Trans-kingdom
transposition of the maize dissociation element. Genetics
174:10951104
Etchberger JF, Hobert O (2008) Vector-free DNA constructs improve
transgene expression in C. elegans. Nat Methods 5:3
Ewald CY, Li C (2010) Understanding the molecular basis of Alzheimers disease using a Caenorhabditis elegans model system.
Brain Struct Funct 214:263283
Faber PW, Alter JR, MacDonald ME, Hart AC (1999) Polyglutaminemediated dysfunction and apoptotic death of a Caenorhabditis
elegans sensory neuron. Proc Natl Acad Sci USA 96:179184
Feany MB, Bender WW (2000) A Drosophila model of Parkinsons
disease. Nature 404:394398
Ferrari R, Kapogiannis D, Huey ED, Momeni P (2011) FTD and ALS:
a tale of two diseases. Curr Alzheimer Res 8:273294
Finelli A, Kelkar A, Song HJ, Yang H, Konsolaki M (2004) A model
for studying Alzheimers Abeta42-induced toxicity in Drosophila
melanogaster. Mol Cell Neurosci 26:365375
Finkbeiner S (2011) Huntingtons disease. Cold Spring Harb Perspect
Biol 3. doi:10.1101/cshperspect.a007476
Flood DG, Lin YG, Lang DM, Trusko SP, Hirsch JD, Savage MJ, Scott
RW, Howland DS (2009) A transgenic rat model of Alzheimers
disease with extracellular Abeta deposition. Neurobiol Aging
30:10781090
Folkesson R, Malkiewicz K, Kloskowska E, Nilsson T, Popova E,
Bogdanovic N, Ganten U, Ganten D, Bader M, Winblad B, Benedikz E (2007) A transgenic rat expressing human APP with the
Swedish Alzheimers disease mutation. Biochem Biophys Res
Commun 358:777782
Gaiano N, Amsterdam A, Kawakami K, Allende M, Becker T, Hopkins N (1996) Insertional mutagenesis and rapid cloning of essential genes in zebraWsh. Nature 383:829832
Gama Sosa MA, De Gasperi R, Elder GA (2010a) Animal transgenesis: an overview. Brain Struct Funct 214:91109
Gama Sosa MA, Gasperi RD, Rocher AB, Wang AC, Janssen WG,
Flores T, Perez GM, Schmeidler J, Dickstein DL, Hof PR, Elder

Hum Genet (2012) 131:535563


GA (2010b) Age-related vascular pathology in transgenic mice
expressing presenilin 1-associated familial Alzheimers disease
mutations. Am J Pathol 176:353368
Games D, Adams D, Alessandrini R, Barbour R, Berthelette P, Blackwell C, Carr T, Clemens J, Donaldson T, Gillespie F et al (1995)
Alzheimer-type neuropathology in transgenic mice overexpressing V717F beta-amyloid precursor protein. Nature 373:523527
Gan BQ, Tang BL (2010) Sirt1s beneWcial roles in neurodegenerative
diseasesa chaperonin containing TCP-1 (CCT) connection?
Aging Cell 9:924929
Gao X, Zhang P (2007) Transgenic RNA interference in mice. Physiology (Bethesda) 22:161166
Garcia EL, Mills AA (2002) Getting around lethality with inducible
Cre-mediated excision. Semin Cell Dev Biol 13:151158
Garrity SJ, Sivanathan V, Dong J, Lindquist S, Hochschild A (2010)
Conversion of a yeast prion protein to an infectious form in bacteria. Proc Natl Acad Sci USA 107:1059610601
Gaszner M, Felsenfeld G (2006) Insulators: exploiting transcriptional
and epigenetic mechanisms. Nat Rev Genet 7:703713
Gendron TF, Petrucelli L (2011) Rodent models of TDP-43 proteinopathy: investigating the mechanisms of TDP-43-mediated neurodegeneration. J Mol Neurosci 45(3):486499
Gentry MS, Dowen RH 3rd, Worby CA, Mattoo S, Ecker JR, Dixon JE
(2007) The phosphatase laforin crosses evolutionary boundaries
and links carbohydrate metabolism to neuronal disease. J Cell
Biol 178:477488
Gentry MS, Dixon JE, Worby CA (2009) Lafora disease: insights into
neurodegeneration from plant metabolism. Trends Biochem Sci
34:628639
Germain DP, Fan JQ (2009) Pharmacological chaperone therapy by
active-site-speciWc chaperones in Fabry disease: in vitro and preclinical studies. Int J Clin Pharmacol Ther 47(Suppl 1):S111
S117
Geurts AM, Cost GJ, Freyvert Y, Zeitler B, Miller JC, Choi VM, Jenkins SS, Wood A, Cui X, Meng X, Vincent A, Lam S, Michalkiewicz M, Schilling R, Foeckler J, Kalloway S, Weiler H,
Menoret S, Anegon I, Davis GD, Zhang L, Rebar EJ, Gregory PD,
Urnov FD, Jacob HJ, Buelow R (2009) Knockout rats via embryo
microinjection of zinc-Wnger nucleases. Science 325:433
Giraldo P, Montoliu L (2001) Size matters: use of YACs, BACs and
PACs in transgenic animals. Transgenic Res 10:83103
Giraldo P, Rival-Gervier S, Houdebine LM, Montoliu L (2003) The
potential beneWts of insulators on heterologous constructs in
transgenic animals. Transgenic Res 12:751755
Goldberg AD, Banaszynski LA, Noh KM, Lewis PW, Elsaesser SJ,
Stadler S, Dewell S, Law M, Guo X, Li X, Wen D, Chapgier A,
DeKelver RC, Miller JC, Lee YL, Boydston EA, Holmes MC,
Gregory PD, Greally JM, RaWi S, Yang C, Scambler PJ, Garrick
D, Gibbons RJ, Higgs DR, Cristea IM, Urnov FD, Zheng D, Allis
CD (2010) Distinct factors control histone variant H3.3 localization at speciWc genomic regions. Cell 140:678691
Golling G, Amsterdam A, Sun Z, Antonelli M, Maldonado E, Chen W,
Burgess S, Haldi M, Artzt K, Farrington S, Lin SY, Nissen RM,
Hopkins N (2002) Insertional mutagenesis in zebraWsh rapidly
identiWes genes essential for early vertebrate development. Nat
Genet 31:135140
Gossen M, Bujard H (1992) Tight control of gene expression in mammalian cells by tetracycline-responsive promoters. Proc Natl
Acad Sci USA 89:55475551
Gotz J, Ittner LM (2008) Animal models of Alzheimers disease and
frontotemporal dementia. Nat Rev Neurosci 9:532544
Gowing G, Philips T, Van Wijmeersch B, Audet JN, Dewil M, Van
Den Bosch L, Billiau AD, Robberecht W, Julien JP (2008)
Ablation of proliferating microglia does not aVect motor neuron
degeneration in amyotrophic lateral sclerosis caused by mutant
superoxide dismutase. J Neurosci 28:1023410244

557
Graham LD (2002) Ecdysone-controlled expression of transgenes. Expert Opin Biol Ther 2:525535
Gray M, Shirasaki DI, Cepeda C, Andre VM, Wilburn B, Lu XH, Tao
J, Yamazaki I, Li SH, Sun YE, Li XJ, Levine MS, Yang XW
(2008) Full-length human mutant huntingtin with a stable polyglutamine repeat can elicit progressive and selective neuropathogenesis in BACHD mice. J Neurosci 28:61826195
Greeve I, Kretzschmar D, Tschape JA, Beyn A, Brellinger C, Schweizer M, Nitsch RM, Reifegerste R (2004) Age-dependent neurodegeneration and Alzheimer-amyloid plaque formation in
transgenic Drosophila. J Neurosci 24:38993906
Groth C, Nornes S, McCarty R, Tamme R, Lardelli M (2002) IdentiWcation of a second presenilin gene in zebraWsh with similarity to
the human Alzheimers disease gene presenilin2. Dev Genes Evol
212:486490
Gurney ME, Pu H, Chiu AY, Dal Canto MC, Polchow CY, Alexander
DD, Caliendo J, Hentati A, Kwon YW, Deng HX et al (1994)
Motor neuron degeneration in mice that express a human Cu, Zn
superoxide dismutase mutation. Science 264:17721775
Halpern ME, Rhee J, Goll MG, Akitake CM, Parsons M, Leach SD
(2008) Gal4/UAS transgenic tools and their application to zebraWsh. ZebraWsh 5:97110
Hans S, Kaslin J, Freudenreich D, Brand M (2009) Temporally-controlled site-speciWc recombination in zebraWsh. PLoS One 4:e4640
Hara K, Kuwayama H, Yaginuma T, Niimi T (2008) Establishment of
a tetracycline-oV system using a piggybac-based vector as a gene
functional analysis tool for the temporal targeting of gene expression. J Insect Biotechnol Sericol 77:3_1593_166
Harrington AJ, Hamamichi S, Caldwell GA, Caldwell KA (2010) C.
elegans as a model organism to investigate molecular pathways
involved with Parkinsons disease. Dev Dyn 239:12821295
Harvey BK, Richie CT, HoVer BJ, Airavaara M (2011) Transgenic animal models of neurodegeneration based on human genetic studies. J Neural Transm 118:2745
Haskins ME, Giger U, Patterson DF (2006) Animal models of lysosomal
storage diseases: their development and clinical relevance. In: Mehta A, Beck M, Sunder-Plassmann G (eds) Fabry disease: perspectives from 5 years of FOS, chap 6. Oxford PharmaGenesis, Oxford
Hauschild J, Petersen B, Santiago Y, Queisser AL, Carnwath JW,
Lucas-Hahn A, Zhang L, Meng X, Gregory PD, Schwinzer R,
Cost GJ, Niemann H (2011) EYcient generation of a biallelic
knockout in pigs using zinc-Wnger nucleases. Proc Natl Acad Sci
USA 108:1201312017
Hayashi S, McMahon AP (2002) EYcient recombination in diverse tissues
by a tamoxifen-inducible form of Cre: a tool for temporally regulated
gene activation/inactivation in the mouse. Dev Biol 244:305318
He G, Luo W, Li P, Remmers C, Netzer WJ, Hendrick J, Bettayeb K,
Flajolet M, Gorelick F, Wennogle LP, Greengard P (2010) Gamma-secretase activating protein is a therapeutic target for Alzheimers disease. Nature 467:9598
Heng MY, DetloV PJ, Wang PL, Tsien JZ, Albin RL (2009) In vivo
evidence for NMDA receptor-mediated excitotoxicity in a murine
genetic model of Huntington disease. J Neurosci 29:32003205
Herzig MC, Winkler DT, Burgermeister P, Pfeifer M, Kohler E,
Schmidt SD, Danner S, Abramowski D, Sturchler-Pierrat C, Burki K, van Duinen SG, Maat-Schieman ML, Staufenbiel M,
Mathews PM, Jucker M (2004) Abeta is targeted to the vasculature in a mouse model of hereditary cerebral hemorrhage with
amyloidosis. Nat Neurosci 7:954960
Hodgson JG, Agopyan N, Gutekunst CA, Leavitt BR, LePiane F,
Singaraja R, Smith DJ, Bissada N, McCutcheon K, Nasir J, Jamot
L, Li XJ, Stevens ME, Rosemond E, Roder JC, Phillips AG,
Rubin EM, Hersch SM, Hayden MR (1999) A YAC mouse model
for Huntingtons disease with full-length mutant huntingtin, cytoplasmic toxicity, and selective striatal neurodegeneration. Neuron
23:181192

123

558
Horev G, Ellegood J, Lerch JP, Son YE, Muthuswamy L, Vogel H,
Krieger AM, Buja A, Henkelman RM, Wigler M, Mills AA
(2011) Dosage-dependent phenotypes in models of 16p11.2
lesions found in autism. Proc Natl Acad Sci USA 108(41):17076
17081
Horn C, Handler AM (2005) Site-speciWc genomic targeting in Drosophila. Proc Natl Acad Sci USA 102:1248312488
Houdebine LM (2010) Design of expression cassettes for the generation of transgenic animals (including insulators). Methods Mol
Biol 597:5569
Hsiao K, Chapman P, Nilsen S, Eckman C, Harigaya Y, Younkin S,
Yang F, Cole G (1996) Correlative memory deWcits, Abeta elevation, and amyloid plaques in transgenic mice. Science 274:99
102
Huang G, Ashton C, Kumbhani DS, Ying QL (2011a) Genetic manipulations in the rat: progress and prospects. Curr Opin Nephrol Hypertens 20:391399
Huang P, Xiao A, Zhou M, Zhu Z, Lin S, Zhang B (2011b) Heritable
gene targeting in zebraWsh using customized TALENs. Nat Biotechnol 29:699700
Huang X, Nguyen AT, Li Z, Emelyanov A, Parinov S, Gong Z (2011c)
One step forward: the use of transgenic zebraWsh tumor model in
drug screens. Birth Defects Res C Embryo Today 93:173181
Igaz LM, Kwong LK, Lee EB, Chen-Plotkin A, Swanson E, Unger T,
Malunda J, Xu Y, Winton MJ, Trojanowski JQ, Lee VM (2011)
Dysregulation of the ALS-associated gene TDP-43 leads to neuronal death and degeneration in mice. J Clin Invest 121:726738
Iijima K, Liu HP, Chiang AS, Hearn SA, Konsolaki M, Zhong Y
(2004) Dissecting the pathological eVects of human Abeta40 and
Abeta42 in Drosophila: a potential model for Alzheimers disease. Proc Natl Acad Sci USA 101:66236628
Iijima K, Chiang HC, Hearn SA, Hakker I, Gatt A, Shenton C, Granger
L, Leung A, Iijima-Ando K, Zhong Y (2008) Abeta42 mutants
with diVerent aggregation proWles induce distinct pathologies in
Drosophila. PLoS One 3:e1703
Irizarry MC, McNamara M, Fedorchak K, Hsiao K, Hyman BT
(1997a) APPSw transgenic mice develop age-related A beta
deposits and neuropil abnormalities, but no neuronal loss in CA1.
J Neuropathol Exp Neurol 56:965973
Irizarry MC, Soriano F, McNamara M, Page KJ, Schenk D, Games D,
Hyman BT (1997b) Abeta deposition is associated with neuropil
changes, but not with overt neuronal loss in the human amyloid
precursor protein V717F (PDAPP) transgenic mouse. J Neurosci
17:70537059
Ivics Z, Hackett PB, Plasterk RH, Izsvak Z (1997) Molecular reconstruction of Sleeping Beauty, a Tc1-like transposon from Wsh, and
its transposition in human cells. Cell 91:501510
Ivics Z, Izsvak Z, Chapman KM, Hamra FK (2011) Sleeping Beauty
transposon mutagenesis of the rat genome in spermatogonial stem
cells. Methods 53:356365
Izsvak Z, Ivics Z (2005) Sleeping Beauty hits them all: transposonmediated saturation mutagenesis in the mouse germline. Nat
Methods 2:735736
Izsvak Z, Frohlich J, Grabundzija I, Shirley JR, Powell HM, Chapman
KM, Ivics Z, Hamra FK (2010) Generating knockout rats by
transposon mutagenesis in spermatogonial stem cells. Nat Methods 7:443445
Jaaro-Peled H (2009) Gene models of schizophrenia: DISC1 mouse
models. Prog Brain Res 179:7586
Jackson GR, Salecker I, Dong X, Yao X, Arnheim N, Faber PW, MacDonald ME, Zipursky SL (1998) Polyglutamine-expanded human
huntingtin transgenes induce degeneration of Drosophila photoreceptor neurons. Neuron 21:633642
Jang CW, Behringer RR (2007) Transposon-mediated transgenesis in
rats. CSH Protoc 2007: pdb prot4866

123

Hum Genet (2012) 131:535563


Jankowsky JL, Slunt HH, Gonzales V, Savonenko AV, Wen JC, Jenkins NA, Copeland NG, Younkin LH, Lester HA, Younkin SG,
Borchelt DR (2005) Persistent amyloidosis following suppression
of Abeta production in a transgenic model of Alzheimer disease.
PLoS Med 2:e355
Jankowsky JL, Younkin LH, Gonzales V, Fadale DJ, Slunt HH, Lester
HA, Younkin SG, Borchelt DR (2007) Rodent A beta modulates
the solubility and distribution of amyloid deposits in transgenic
mice. J Biol Chem 282:2270722720
Jeibmann A, Paulus W (2009) Drosophila melanogaster as a model
organism of brain diseases. Int J Mol Sci 10:407440
Jones WD (2009) The expanding reach of the GAL4/UAS system into
the behavioral neurobiology of Drosophila. BMB Rep 42:705712
Jorgensen EM, Mango SE (2002) The art and design of genetic
screens: caenorhabditis elegans. Nat Rev Genet 3:356369
Joyner AL, Guillemot F (1994) Gene targeting and development of the
nervous system. Curr Opin Neurobiol 4:3742
Justice MJ, Noveroske JK, Weber JS, Zheng B, Bradley A (1999)
Mouse ENU mutagenesis. Hum Mol Genet 8:19551963
Kabashi E, Lin L, Tradewell ML, Dion PA, Bercier V, Bourgouin P,
Rochefort D, Bel Hadj S, Durham HD, Vande Velde C, Rouleau
GA, Drapeau P (2010) Gain and loss of function of ALS-related
mutations of TARDBP (TDP-43) cause motor deWcits in vivo.
Hum Mol Genet 19:671683
Kasuga K, Shimohata T, Nishimura A, Shiga A, Mizuguchi T, Tokunaga J, Ohno T, Miyashita A, Kuwano R, Matsumoto N, Onodera
O, Nishizawa M, Ikeuchi T (2009) IdentiWcation of independent
APP locus duplication in Japanese patients with early-onset Alzheimer disease. J Neurol Neurosurg Psychiatry 80:10501052
Kato T, Kubota M, Kasahara T (2007) Animal models of bipolar disorder. Neurosci Biobehav Rev 31:832842
Kawakami K (2005) Transposon tools and methods in zebraWsh. Dev
Dyn 234:244254
Kawakami K, Koga A, Hori H, Shima A (1998) Excision of the tol2
transposable element of the medaka Wsh, Oryzias latipes, in zebraWsh, Danio rerio. Gene 225:1722
Kawakami K, Shima A, Kawakami N (2000) IdentiWcation of a functional transposase of the Tol2 element, an Ac-like element from
the Japanese medaka Wsh, and its transposition in the zebraWsh
germ lineage. Proc Natl Acad Sci USA 97:1140311408
Kelly A, Hurlstone AF (2011) The use of RNAi technologies for gene
knockdown in zebraWsh. Brief Funct Genomics 10:189196
Khandelwal A, Chandu D, Roe CM, Kopan R, Quatrano RS (2007)
Moonlighting activity of presenilin in plants is independent of
gamma-secretase and evolutionarily conserved. Proc Natl Acad
Sci USA 104:1333713342
Khanna R, Soska R, Lun Y, Feng J, Frascella M, Young B, Brignol N,
Pellegrino L, Sitaraman SA, Desnick RJ, Benjamin ER, Lockhart
DJ, Valenzano KJ (2010) The pharmacological chaperone 1-deoxygalactonojirimycin reduces tissue globotriaosylceramide levels in a mouse model of Fabry disease. Mol Ther 18:2333
Khapre RV, Samsa WE, Kondratov RV (2010) Circadian regulation of
cell cycle: molecular connections between aging and the circadian
clock. Ann Med 42:404415
Kim D, Nguyen MD, Dobbin MM, Fischer A, Sananbenesi F, Rodgers
JT, Delalle I, Baur JA, Sui G, Armour SM, Puigserver P, Sinclair
DA, Tsai LH (2007) SIRT1 deacetylase protects against neurodegeneration in models for Alzheimers disease and amyotrophic
lateral sclerosis. EMBO J 26:31693179
Knopf F, Schnabel K, Haase C, Pfeifer K, Anastassiadis K, Weidinger
G (2010) Dually inducible TetON systems for tissue-speciWc conditional gene expression in zebraWsh. Proc Natl Acad Sci USA
107:1993319938
Kraemer BC, Zhang B, Leverenz JB, Thomas JH, Trojanowski JQ,
Schellenberg GD (2003) Neurodegeneration and defective neuro-

Hum Genet (2012) 131:535563


transmission in a Caenorhabditis elegans model of tauopathy.
Proc Natl Acad Sci USA 100:99809985
Kragh PM, Nielsen AL, Li J, Du Y, Lin L, Schmidt M, Bogh IB, Holm
IE, Jakobsen JE, Johansen MG, Purup S, Bolund L, Vajta G, Jorgensen AL (2009) Hemizygous minipigs produced by random
gene insertion and handmade cloning express the Alzheimers
disease-causing dominant mutation APPsw. Transgenic Res
18:545558
Kudo T, Loh DH, Truong D, Wu Y, Colwell CS (2011a) Circadian
dysfunction in a mouse model of Parkinsons disease. Exp Neurol
232:6675
Kudo T, Schroeder A, Loh DH, Kuljis D, Jordan MC, Roos KP, Colwell CS (2011b) Dysfunctions in circadian behavior and physiology in mouse models of Huntingtons disease. Exp Neurol
228:8090
Kuwahara T, Koyama A, Gengyo-Ando K, Masuda M, Kowa H, Tsunoda M, Mitani S, Iwatsubo T (2006) Familial Parkinson mutant
alpha-synuclein causes dopamine neuron dysfunction in transgenic Caenorhabditis elegans. J Biol Chem 281:334340
Lakso M, Vartiainen S, Moilanen AM, Sirvio J, Thomas JH, Nass R,
Blakely RD, Wong G (2003) Dopaminergic neuronal loss and
motor deWcits in Caenorhabditis elegans overexpressing human
alpha-synuclein. J Neurochem 86:165172
Lamb BT, Sisodia SS, Lawler AM, Slunt HH, Kitt CA, Kearns WG,
Pearson PL, Price DL, Gearhart JD (1993) Introduction and
expression of the 400 kilobase amyloid precursor protein gene in
transgenic mice (corrected). Nat Genet 5:2230
Langenau DM, Feng H, Berghmans S, Kanki JP, Kutok JL, Look AT
(2005) Cre/lox-regulated transgenic zebraWsh model with conditional myc-induced T cell acute lymphoblastic leukemia. Proc
Natl Acad Sci USA 102:60686073
Largaespada DA (2009a) Transposon mutagenesis in mice. Methods
Mol Biol 530:379390
Largaespada DA (2009b) Transposon-mediated mutagenesis of somatic cells in the mouse for cancer gene identiWcation. Methods
49:282286
Lee MK, Stirling W, Xu Y, Xu X, Qui D, Mandir AS, Dawson TM,
Copeland NG, Jenkins NA, Price DL (2002) Human alpha-synuclein-harboring familial Parkinsons disease-linked Ala-53Thr
mutation causes neurodegenerative disease with alpha-synuclein
aggregation in transgenic mice. Proc Natl Acad Sci USA
99:89688973
Leon WC, Canneva F, Partridge V, Allard S, Ferretti MT, DeWilde A,
Vercauteren F, Atifeh R, Ducatenzeiler A, Klein W, Szyf M, Alhonen L, Cuello AC (2010) A novel transgenic rat model with a
full Alzheimers-like amyloid pathology displays pre-plaque
intracellular amyloid-beta-associated cognitive impairment.
J Alzheimers Dis 20:113126
Levine MS, Cepeda C, Hickey MA, Fleming SM, Chesselet MF (2004)
Genetic mouse models of Huntingtons and Parkinsons diseases:
illuminating but imperfect. Trends Neurosci 27:691697
Lewandoski M (2001) Conditional control of gene expression in the
mouse. Nat Rev Genet 2:743755
Lewis J, McGowan E, Rockwood J, Melrose H, Nacharaju P, Van
Slegtenhorst M, Gwinn-Hardy K, Paul Murphy M, Baker M, Yu
X, DuV K, Hardy J, Corral A, Lin WL, Yen SH, Dickson DW,
Davies P, Hutton M (2000) NeuroWbrillary tangles, amyotrophy
and progressive motor disturbance in mice expressing mutant
(P301L) tau protein. Nat Genet 25:402405
Li X, Patel JC, Wang J, Avshalumov MV, Nicholson C, Buxbaum JD,
Elder GA, Rice ME, Yue Z (2010) Enhanced striatal dopamine
transmission and motor performance with LRRK2 overexpression in mice is eliminated by familial Parkinsons disease mutation G2019S. J Neurosci 30:17881797
Li H, Haurigot V, Doyon Y, Li T, Wong SY, Bhagwat AS, Malani N,
Anguela XM, Sharma R, Ivanciu L, Murphy SL, Finn JD, Khazi

559
FR, Zhou S, Paschon DE, Rebar EJ, Bushman FD, Gregory PD,
Holmes MC, High KA (2011a) In vivo genome editing restores
haemostasis in a mouse model of haemophilia. Nature 475:217
221
Li MA, Turner DJ, Ning Z, Yusa K, Liang Q, Eckert S, Rad L, Fitzgerald TW, Craig NL, Bradley A (2011b) Mobilization of giant piggyBac transposons in the mouse genome. Nucleic Acids Res.
doi:10.1093/nar/gkr764
Liang J, Liu E, Yu Y, Kitajima S, Koike T, Jin Y, Morimoto M, Hatakeyama K, Asada Y, Watanabe T, Sasaguri Y, Watanabe S, Fan J
(2006) Macrophage metalloelastase accelerates the progression of
atherosclerosis in transgenic rabbits. Circulation 113:19932001
Lieschke GJ, Currie PD (2007) Animal models of human disease: zebraWsh swim into view. Nat Rev Genet 8:353367
Lin CH, Tallaksen-Greene S, Chien WM, Cearley JA, Jackson WS,
Crouse AB, Ren S, Li XJ, Albin RL, DetloV PJ (2001) Neurological abnormalities in a knock-in mouse model of Huntingtons
disease. Hum Mol Genet 10:137144
Link CD (1995) Expression of human beta-amyloid peptide in transgenic Caenorhabditis elegans. Proc Natl Acad Sci USA 92:9368
9372
Link CD (2001) Transgenic invertebrate models of age-associated neurodegenerative diseases. Mech Ageing Dev 122:16391649
Lione LA, Carter RJ, Hunt MJ, Bates GP, Morton AJ, Dunnett SB
(1999) Selective discrimination learning impairments in mice
expressing the human Huntingtons disease mutation. J Neurosci
19:1042810437
Liu BH, Wang X, Ma YX, Wang S (2004) CMV enhancer/human
PDGF-beta promoter for neuron-speciWc transgene expression.
Gene Ther 11:5260
Liu K, Hipkens S, Yang T, Abraham R, Zhang W, Chopra N, Knollmann B, Magnuson MA, Roden DM (2006) Recombinase-mediated cassette exchange to rapidly and eYciently generate mice
with human cardiac sodium channels. Genesis 44:556564
Liu L, Orozco IJ, Planel E, Wen Y, Bretteville A, Krishnamurthy P,
Wang L, Herman M, Figueroa H, Yu WH, Arancio O, DuV K
(2008a) A transgenic rat that develops Alzheimers disease-like
amyloid pathology, deWcits in synaptic plasticity and cognitive
impairment. Neurobiol Dis 31:4657
Liu X, Li Z, Emelyanov A, Parinov S, Gong Z (2008b) Generation of
oocyte-speciWcally expressed cre transgenic zebraWsh for female
germline excision of loxP-Xanked transgene. Dev Dyn 237:2955
2962
Ljubuncic P, Reznick AZ (2009) The evolutionary theories of aging
revisiteda mini-review. Gerontology 55:205216
Lu B, Vogel H (2009) Drosophila models of neurodegenerative diseases. Annu Rev Pathol 4:315342
Lu XH, Fleming SM, Meurers B, Ackerson LC, Mortazavi F, Lo V,
Hernandez D, Sulzer D, Jackson GR, Maidment NT, Chesselet
MF, Yang XW (2009) Bacterial artiWcial chromosome transgenic
mice expressing a truncated mutant parkin exhibit age-dependent
hypokinetic motor deWcits, dopaminergic neuron degeneration,
and accumulation of proteinase K-resistant alpha-synuclein.
J Neurosci 29:19621976
Lublin AL, Gandy S (2010) Amyloid-beta oligomers: possible roles as
key neurotoxins in Alzheimers disease. Mt Sinai J Med 77:4349
Luthi A, Van der Putten H, Botteri FM, Mansuy IM, Meins M, Frey U,
Sansig G, Portet C, Schmutz M, Schroder M, Nitsch C, Laurent
JP, Monard D (1997) Endogenous serine protease inhibitor modulates epileptic activity and hippocampal long-term potentiation.
J Neurosci 17:46884699
Ma Y, Hu H, Berrebi AS, Mathers PH, Agmon A (2006) Distinct subtypes of somatostatin-containing neocortical interneurons revealed in transgenic mice. J Neurosci 26:50695082
Mallajosyula JK, Kaur D, Chinta SJ, Rajagopalan S, Rane A, Nicholls
DG, Di Monte DA, Macarthur H, Andersen JK (2008) MAO-B

123

560
elevation in mouse brain astrocytes results in Parkinsons pathology. PLoS One 3:e1616
Mangiarini L, Sathasivam K, Seller M, Cozens B, Harper A, Hetherington C, Lawton M, Trottier Y, Lehrach H, Davies SW, Bates
GP (1996) Exon 1 of the HD gene with an expanded CAG repeat
is suYcient to cause a progressive neurological phenotype in
transgenic mice. Cell 87:493506
Mardis ER (2011) A decades perspective on DNA sequencing technology. Nature 470:198203
Metzger D, CliVord J, Chiba H, Chambon P (1995) Conditional sitespeciWc recombination in mammalian cells using a ligand-dependent chimeric Cre recombinase. Proc Natl Acad Sci USA
92:69916995
Meyer M, de Angelis MH, Wurst W, Kuhn R (2010) Gene targeting by
homologous recombination in mouse zygotes mediated by zincWnger nucleases. Proc Natl Acad Sci USA 107:1502215026
Miller RL (2011) Transgenic mice: beyond the knockout. Am J Physiol
Renal Physiol 300:F291F300
Minois N, Sykacek P, Godsey B, Kreil DP (2010) RNA interference in
ageing researcha mini-review. Gerontology 56:496506
Miura M, Tamura T, Mikoshiba K (1990) Cell-speciWc expression of
the mouse glial Wbrillary acidic protein gene: identiWcation of the
cis- and trans-acting promoter elements for astrocyte-speciWc
expression. J Neurochem 55:11801188
Moens CB, Donn TM, Wolf-Saxon ER, Ma TP (2008) Reverse genetics in zebraWsh by TILLING. Brief Funct Genomic Proteomic
7:454459
Mohr S, Bakal C, Perrimon N (2010) Genomic screening with RNAi:
results and challenges. Annu Rev Biochem 79:3764
Montoliu L, Schedl A, Kelsey G, Lichter P, Larin Z, Lehrach H, Schutz
G (1993) Generation of transgenic mice with yeast artiWcial chromosomes. Cold Spring Harb Symp Quant Biol 58:5562
Moreira PN, Giraldo P, Cozar P, Pozueta J, Jimenez A, Montoliu L,
Gutierrez-Adan A (2004) EYcient generation of transgenic mice
with intact yeast artiWcial chromosomes by intracytoplasmic
sperm injection. Biol Reprod 71:19431947
Moreira PN, Pozueta J, Giraldo P, Gutierrez-Adan A, Montoliu L
(2006) Generation of yeast artiWcial chromosome transgenic mice
by intracytoplasmic sperm injection. Methods Mol Biol 349:151
161
Moreira PN, Pozueta J, Perez-Crespo M, Valdivieso F, Gutierrez-Adan
A, Montoliu L (2007) Improving the generation of genomic-type
transgenic mice by ICSI. Transgenic Res 16:163168
Moscou MJ, Bogdanove AJ (2009) A simple cipher governs DNA recognition by TAL eVectors. Science 326:1501
Muller B, Grossniklaus U (2010) Model organismsa historical perspective. J Proteomics 73:20542063
Muqit MM, Feany MB (2002) Modelling neurodegenerative diseases
in Drosophila: a fruitful approach? Nat Rev Neurosci 3:237243
Nagy A (2000) Cre recombinase: the universal reagent for genome tailoring. Genesis 26:99109
Nern A, PfeiVer BD, Svoboda K, Rubin GM (2011) Multiple new sitespeciWc recombinases for use in manipulating animal genomes.
Proc Natl Acad Sci USA 108:1419814203
Newman M, Tucker B, Nornes S, Ward A, Lardelli M (2009) Altering
presenilin gene activity in zebraWsh embryos causes changes in
expression of genes with potential involvement in Alzheimers
disease pathogenesis. J Alzheimers Dis 16:133147
Niittyla T, Comparot-Moss S, Lue WL, Messerli G, Trevisan M, Seymour MD, Gatehouse JA, Villadsen D, Smith SM, Chen J, Zeeman SC, Smith AM (2006) Similar protein phosphatases control
starch metabolism in plants and glycogen metabolism in mammals. J Biol Chem 281:1181511818
Niu Y, Yu Y, Bernat A, Yang S, He X, Guo X, Chen D, Chen Y, Ji S,
Si W, Lv Y, Tan T, Wei Q, Wang H, Shi L, Guan J, Zhu X,
AfanassieV M, Savatier P, Zhang K, Zhou Q, Ji W (2010)

123

Hum Genet (2012) 131:535563


Transgenic rhesus monkeys produced by gene transfer into earlycleavage-stage embryos using a simian immunodeWciency virusbased vector. Proc Natl Acad Sci USA 107:1766317667
Nollen EA, Garcia SM, van Haaften G, Kim S, Chavez A, Morimoto
RI, Plasterk RH (2004) Genome-wide RNA interference screen
identiWes previously undescribed regulators of polyglutamine
aggregation. Proc Natl Acad Sci USA 101:64036408
Nornes S, Newman M, Verdile G, Wells S, Stoick-Cooper CL, Tucker
B, Frederich-Sleptsova I, Martins R, Lardelli M (2008) Interference with splicing of presenilin transcripts has potent dominant
negative eVects on presenilin activity. Hum Mol Genet 17:402
412
Nornes S, Newman M, Wells S, Verdile G, Martins RN, Lardelli M
(2009) Independent and cooperative action of Psen2 with Psen1
in zebraWsh embryos. Exp Cell Res 315:27912801
Oakley H, Cole SL, Logan S, Maus E, Shao P, Craft J, Guillozet-Bongaarts A, Ohno M, Disterhoft J, Van Eldik L, Berry R, Vassar R
(2006) Intraneuronal beta-amyloid aggregates, neurodegeneration, and neuron loss in transgenic mice with Wve familial Alzheimers disease mutations: potential factors in amyloid plaque
formation. J Neurosci 26:1012910140
Oberstein A, Pare A, Kaplan L, Small S (2005) Site-speciWc transgenesis by Cre-mediated recombination in Drosophila. Nat Methods
2:583585
Oddo S, Caccamo A, Shepherd JD, Murphy MP, Golde TE, Kayed R,
Metherate R, Mattson MP, Akbari Y, LaFerla FM (2003) Tripletransgenic model of Alzheimers disease with plaques and tangles: intracellular Abeta and synaptic dysfunction. Neuron
39:409421
Oddo S, Billings L, Kesslak JP, Cribbs DH, LaFerla FM (2004) Abeta
immunotherapy leads to clearance of early, but not late, hyperphosphorylated tau aggregates via the proteasome. Neuron
43:321332
Ostertag EM, Madison BB, Kano H (2007) Mutagenesis in rodents using the L1 retrotransposon. Genome Biol 8(Suppl 1):S16
Pallas M, Casadesus G, Smith MA, Coto-Montes A, Pelegri C, Vilaplana J, Camins A (2009) Resveratrol and neurodegenerative diseases: activation of SIRT1 as the potential pathway towards
neuroprotection. Curr Neurovasc Res 6:7081
Pan X, Wan H, Chia W, Tong Y, Gong Z (2005) Demonstration of sitedirected recombination in transgenic zebraWsh using the Cre/loxP
system. Transgenic Res 14:217223
Paquet D, Bhat R, Sydow A, Mandelkow EM, Berg S, Hellberg S, Falting J, Distel M, Koster RW, Schmid B, Haass C (2009) A zebraWsh model of tauopathy allows in vivo imaging of neuronal cell
death and drug evaluation. J Clin Invest 119:13821395
Paquet D, Schmid B, Haass C (2010) Transgenic zebraWsh as a novel
animal model to study tauopathies and other neurodegenerative
disorders in vivo. Neurodegener Dis 7:99102
Parkes TL, Elia AJ, Dickinson D, Hilliker AJ, Phillips JP, Boulianne
GL (1998) Extension of Drosophila lifespan by overexpression of
human SOD1 in motorneurons. Nat Genet 19:171174
Pesaresi MG, Amori I, Giorgi C, Ferri A, Fiorenzo P, Gabanella F, Salvatore AM, Giorgio M, Pelicci PG, Pinton P, Carri MT, Cozzolino M (2011) Mitochondrial redox signalling by p66Shc mediates
ALS-like disease through Rac1 inactivation. Hum Mol Genet
20:41964208
Piens M, Muller M, Bodson M, Baudouin G, Plumier JC (2010) A
short upstream promoter region mediates transcriptional regulation of the mouse doublecortin gene in diVerentiating neurons.
BMC Neurosci 11:64
Premsrirut PK, Dow LE, Kim SY, Camiolo M, Malone CD, Miething
C, Scuoppo C, Zuber J, Dickins RA, Kogan SC, Shroyer KR,
Sordella R, Hannon GJ, Lowe SW (2011) A rapid and scalable
system for studying gene function in mice using conditional RNA
interference. Cell 145:145158

Hum Genet (2012) 131:535563


Rad R, Rad L, Wang W, Cadinanos J, Vassiliou G, Rice S, Campos LS,
Yusa K, Banerjee R, Li MA, de la Rosa J, Strong A, Lu D, Ellis
P, Conte N, Yang FT, Liu P, Bradley A (2010) PiggyBac transposon mutagenesis: a tool for cancer gene discovery in mice. Science 330:11041107
Raymond LA, Andre VM, Cepeda C, Gladding CM, Milnerwood AJ,
Levine MS (2011) Pathophysiology of Huntingtons disease:
time-dependent alterations in synaptic and receptor function.
Neuroscience 198:252273
Reaume AG, Howland DS, Trusko SP, Savage MJ, Lang DM, Greenberg BD, Siman R, Scott RW (1996) Enhanced amyloidogenic
processing of the beta-amyloid precursor protein in gene-targeted
mice bearing the Swedish familial Alzheimers disease mutations
and a humanized Abeta sequence. J Biol Chem 271:23380
23388
Remy S, Tesson L, Menoret S, Usal C, Scharenberg AM, Anegon I
(2010) Zinc-Wnger nucleases: a powerful tool for genetic engineering of animals. Transgenic Res 19:363371
Reyon D, Kirkpatrick JR, Sander JD, Zhang F, Voytas DF, Joung JK,
Dobbs D, CoVman CR (2011) ZFNGenome: a comprehensive resource for locating zinc Wnger nuclease target sites in model
organisms. BMC Genomics 12:83
Ringrose L, Lounnas V, Ehrlich L, Buchholz F, Wade R, Stewart AF
(1998) Comparative kinetic analysis of FLP and cre recombinases: mathematical models for DNA binding and recombination.
J Mol Biol 284:363384
Robertson HR, Feng G (2011) Annual research review: transgenic
mouse models of childhood-onset psychiatric disorders. J Child
Psychol Psychiatry 52:442475
Roebroek AJ, Gordts PL, Reekmans S (2011) Generation of a series of
knock-in alleles using RMCE in ES cells. Methods Mol Biol
693:277281
Roman G (2004) The genetics of Drosophila transgenics. Bioessays
26:12431253
Romer P, Recht S, Strauss T, Elsaesser J, Schornack S, Boch J, Wang
S, Lahaye T (2010) Promoter elements of rice susceptibility genes
are bound and activated by speciWc TAL eVectors from the bacterial blight pathogen, Xanthomonas oryzae pv. oryzae. New Phytol
187:10481057
Ruiz-Opazo N, Kosik KS, Lopez LV, Bagamasbad P, Ponce LR, Herrera VL (2004) Attenuated hippocampus-dependent learning and
memory decline in transgenic TgAPPswe Fischer-344 rats. Mol
Med 10:3644
Sager JJ, Bai Q, Burton EA (2010) Transgenic zebraWsh models of
neurodegenerative diseases. Brain Struct Funct 214:285302
Sakimura K, Kushiya E, Takahashi Y, Suzuki Y (1987) The structure
and expression of neuron-speciWc enolase gene. Gene 60:103
113
Sarov M, Schneider S, Pozniakovski A, Roguev A, Ernst S, Zhang Y,
Hyman AA, Stewart AF (2006) A recombineering pipeline for
functional genomics applied to Caenorhabditis elegans. Nat
Methods 3:839844
Schmitz C, Rutten BP, Pielen A, Schafer S, Wirths O, Tremp G, Czech
C, Blanchard V, Multhaup G, Rezaie P, Korr H, Steinbusch HW,
Pradier L, Bayer TA (2004) Hippocampal neuron loss exceeds
amyloid plaque load in a transgenic mouse model of Alzheimers
disease. Am J Pathol 164:14951502
Seelaar H, Rohrer JD, Pijnenburg YA, Fox NC, van Swieten JC (2011)
Clinical, genetic and pathological heterogeneity of frontotemporal dementia: a review. J Neurol Neurosurg Psychiatry 82:476
486
Seibler J, Kleinridders A, Kuter-Luks B, Niehaves S, Bruning JC, Schwenk F (2007) Reversible gene knockdown in mice using a tight,
inducible shRNA expression system. Nucleic Acids Res 35:e54
Selkoe DJ (2001) Alzheimers disease: genes, proteins, and therapy.
Physiol Rev 81:741766

561
Selkoe DJ (2011) Alzheimers disease. Cold Spring Harb Perspect Biol
3. doi:10.1101/cshperspect.a004457
Semple JI, Garcia-Verdugo R, Lehner B (2010) Rapid selection of
transgenic C. elegans using antibiotic resistance. Nat Methods
7:725727
Settivari R, Levora J, Nass R (2009) The divalent metal transporter
homologues SMF-1/2 mediate dopamine neuron sensitivity in
caenorhabditis elegans models of manganism and parkinson disease. J Biol Chem 284:3575835768
Shan X, Chiang PM, Price DL, Wong PC (2010) Altered distributions
of Gemini of coiled bodies and mitochondria in motor neurons of
TDP-43 transgenic mice. Proc Natl Acad Sci USA 107:16325
16330
Shelbourne PF, Killeen N, Hevner RF, Johnston HM, Tecott L, Lewandoski M, Ennis M, Ramirez L, Li Z, Iannicola C, Littman DR,
Myers RM (1999) A Huntingtons disease CAG expansion at the
murine Hdh locus is unstable and associated with behavioural
abnormalities in mice. Hum Mol Genet 8:763774
Shulman JM, De Jager PL, Feany MB (2011) Parkinsons disease:
genetics and pathogenesis. Annu Rev Pathol 6:193222
Siegal ML, Hartl DL (2000) Application of Cre/loxP in Drosophila.
Site-speciWc recombination and transgene coplacement. Methods
Mol Biol 136:487495
Simard AR, Soulet D, Gowing G, Julien JP, Rivest S (2006) Bone marrow-derived microglia play a critical role in restricting senile
plaque formation in Alzheimers disease. Neuron 49:489502
Skaper SD, Giusti P (2010) Transgenic mouse models of Parkinsons
disease and Huntingtons disease. CNS Neurol Disord Drug Targets 9:455470
Sleegers K, Brouwers N, Gijselinck I, Theuns J, Goossens D, Wauters
J, Del-Favero J, Cruts M, van Duijn CM, Van Broeckhoven C
(2006) APP duplication is suYcient to cause early onset Alzheimers dementia with cerebral amyloid angiopathy. Brain
129:29772983
Slow EJ, van Raamsdonk J, Rogers D, Coleman SH, Graham RK,
Deng Y, Oh R, Bissada N, Hossain SM, Yang YZ, Li XJ, Simpson EM, Gutekunst CA, Leavitt BR, Hayden MR (2003) Selective
striatal neuronal loss in a YAC128 mouse model of Huntington
disease. Hum Mol Genet 12:15551567
Snyder JS, Soumier A, Brewer M, Pickel J, Cameron HA (2011) Adult
hippocampal neurogenesis buVers stress responses and depressive
behaviour. Nature 476:458461
Sora I, Li B, Igari M, Hall FS, Ikeda K (2010) Transgenic mice in the
study of drug addiction and the eVects of psychostimulant drugs.
Ann N Y Acad Sci 1187:218246
Spilman P, Podlutskaya N, Hart MJ, Debnath J, Gorostiza O, Bredesen
D, Richardson A, Strong R, Galvan V (2010) Inhibition of mTOR
by rapamycin abolishes cognitive deWcits and reduces amyloidbeta levels in a mouse model of Alzheimers disease. PLoS One
5:e9979
St Johnston D (2002) The art and design of genetic screens: Drosophila
melanogaster. Nat Rev Genet 3:176188
Stallings NR, Puttaparthi K, Luther CM, Burns DK, Elliott JL (2010)
Progressive motor weakness in transgenic mice expressing human TDP-43. Neurobiol Dis 40:404414
Stebbins MJ, Yin JC (2001) Adaptable doxycycline-regulated gene
expression systems for Drosophila. Gene 270:103111
Stebbins MJ, Urlinger S, Byrne G, Bello B, Hillen W, Yin JC (2001)
Tetracycline-inducible systems for Drosophila. Proc Natl Acad
Sci USA 98:1077510780
Sterniczuk R, Antle MC, Laferla FM, Dyck RH (2010) Characterization of the 3xTg-AD mouse model of Alzheimers disease: part 2.
Behavioral and cognitive changes. Brain Res 1348:149155
Stieger K, Belbellaa B, Le Guiner C, Moullier P, Rolling F (2009)
In vivo gene regulation using tetracycline-regulatable systems.
Adv Drug Deliv Rev 61:527541

123

562
Sureshkumar S, Todesco M, Schneeberger K, Harilal R, Balasubramanian S, Weigel D (2009) A genetic defect caused by a triplet repeat
expansion in Arabidopsis thaliana. Science 323:10601063
Suzuki K, Mansson JE (1998) Animal models of lysosomal disease: an
overview. J Inherit Metab Dis 21:540547
Szulc J, Wiznerowicz M, Sauvain MO, Trono D, Aebischer P (2006)
A versatile tool for conditional gene expression and knockdown.
Nat Methods 3:109116
Takeda J, Izsvak Z, Ivics Z (2008) Insertional mutagenesis of the
mouse germline with Sleeping Beauty transposition. Methods
Mol Biol 435:109125
Teschendorf D, Link CD (2009) What have worm models told us about
the mechanisms of neuronal dysfunction in human neurodegenerative diseases? Mol Neurodegener 4:38
Tesson L, Usal C, Menoret S, Leung E, Niles BJ, Remy S, Santiago Y,
Vincent AI, Meng X, Zhang L, Gregory PD, Anegon I, Cost GJ
(2011) Knockout rats generated by embryo microinjection of TALENs. Nat Biotechnol 29:695696
Thermes V, Grabher C, Ristoratore F, Bourrat F, Choulika A, Wittbrodt J, Joly JS (2002) I-SceI meganuclease mediates highly eYcient transgenesis in Wsh. Mech Dev 118:9198
Thompson LM, Marsh JL (2003) Invertebrate models of neurologic
disease: insights into pathogenesis and therapy. Curr Neurol Neurosci Rep 3:442448
Thummel R, Burket CT, Brewer JL, Sarras MP Jr, Li L, Perry M,
McDermott JP, Sauer B, Hyde DR, Godwin AR (2005) Cre-mediated site-speciWc recombination in zebraWsh embryos. Dev Dyn
233:13661377
Tong C, Li P, Wu NL, Yan Y, Ying QL (2010) Production of p53 gene
knockout rats by homologous recombination in embryonic stem
cells. Nature 467:211213
Tong C, Huang G, Ashton C, Li P, Ying QL (2011) Generating gene
knockout rats by homologous recombination in embryonic stem
cells. Nat Protoc 6:827844
Trancikova A, Ramonet D, Moore DJ (2011) Genetic mouse models of
neurodegenerative diseases. Prog Mol Biol Transl Sci 100:419482
Urani A, Gass P (2003) Corticosteroid receptor transgenic mice: models for depression? Ann N Y Acad Sci 1007:379393
Urlinger S, Baron U, Thellmann M, Hasan MT, Bujard H, Hillen W
(2000) Exploring the sequence space for tetracycline-dependent
transcriptional activators: novel mutations yield expanded range
and sensitivity. Proc Natl Acad Sci USA 97:79637968
van der Putten H, Wiederhold KH, Probst A, Barbieri S, Mistl C,
Danner S, KauVmann S, Hofele K, Spooren WP, Ruegg MA, Lin
S, Caroni P, Sommer B, Tolnay M, Bilbe G (2000) Neuropathology
in mice expressing human alpha-synuclein. J Neurosci 20:6021
6029
van Tijn P, Kamphuis W, Marlatt MW, Hol EM, Lucassen PJ (2011)
Presenilin mouse and zebraWsh models for dementia: focus on
neurogenesis. Prog Neurobiol 93:149164
Vander Kooi CW, Taylor AO, Pace RM, Meekins DA, Guo HF, Kim
Y, Gentry MS (2010) Structural basis for the glucan phosphatase
activity of Starch Excess4. Proc Natl Acad Sci USA 107:15379
15384
Venken KJ, He Y, Hoskins RA, Bellen HJ (2006) P[acman]: a BAC
transgenic platform for targeted insertion of large DNA fragments
in D. melanogaster. Science 314:17471751
Virata MJ, Zeller RW (2010) Ascidians: an invertebrate chordate model to study Alzheimers disease pathogenesis. Dis Model Mech
3:377385
Voytas DF, Joung JK (2009) Plant science. DNA binding made easy.
Science 326:14911492
Walker TL, Yasuda T, Adams DJ, Bartlett PF (2007) The doublecortin-expressing population in the developing and adult brain contains multipotential precursors in addition to neuronal-lineage
cells. J Neurosci 27:37343742

123

Hum Genet (2012) 131:535563


Wang X (2009) Cre transgenic mouse lines. Methods Mol Biol
561:265273
Wang W, Lin C, Lu D, Ning Z, Cox T, Melvin D, Wang X, Bradley A,
Liu P (2008) Chromosomal transposition of PiggyBac in mouse
embryonic stem cells. Proc Natl Acad Sci USA 105:92909295
Wang F, Wang X, Yuan CG, Ma J (2010) Generating a prion with bacterially expressed recombinant prion protein. Science 327:1132
1135
Watson MR, Lagow RD, Xu K, Zhang B, Bonini NM (2008) A drosophila model for amyotrophic lateral sclerosis reveals motor
neuron damage by human SOD1. J Biol Chem 283:2497224981
Wegorzewska I, Baloh RH (2011) TDP-43-based animal models of
neurodegeneration: new insights into ALS pathology and pathophysiology. Neurodegener Dis 8:262274
Wegorzewska I, Bell S, Cairns NJ, Miller TM, Baloh RH (2009) TDP43 mutant transgenic mice develop features of ALS and frontotemporal lobar degeneration. Proc Natl Acad Sci USA
106:1880918814
Wen PH, Shao X, Shao Z, Hof PR, Wisniewski T, Kelley K, Friedrich
VL Jr, Ho L, Pasinetti GM, Shioi J, Robakis NK, Elder GA (2002)
Overexpression of wild type but not an FAD mutant presenilin-1
promotes neurogenesis in the hippocampus of adult mice. Neurobiol Dis 10:819
Wen PH, Hof PR, Chen X, Gluck K, Austin G, Younkin SG, Younkin
LH, DeGasperi R, Gama Sosa MA, Robakis NK, Haroutunian V,
Elder GA (2004) The presenilin-1 familial Alzheimer disease mutant P117L impairs neurogenesis in the hippocampus of adult
mice. Exp Neurol 188:224237
Wheeler VC, White JK, Gutekunst CA, Vrbanac V, Weaver M, Li XJ,
Li SH, Yi H, Vonsattel JP, Gusella JF, Hersch S, Auerbach W,
Joyner AL, MacDonald ME (2000) Long glutamine tracts cause
nuclear localization of a novel form of huntingtin in medium
spiny striatal neurons in HdhQ92 and HdhQ111 knock-in mice.
Hum Mol Genet 9:503513
White JK, Auerbach W, Duyao MP, Vonsattel JP, Gusella JF, Joyner
AL, MacDonald ME (1997) Huntingtin is required for neurogenesis and is not impaired by the Huntingtons disease CAG expansion. Nat Genet 17:404410
Wienholds E, van Eeden F, Kosters M, Mudde J, Plasterk RH, Cuppen
E (2003) EYcient target-selected mutagenesis in zebraWsh. Genome Res 13:27002707
Williams GC (1957) Pleiotropy, natural-selection, and the evolution of
senescence. Evolution 11:398411
Williams A, Sarkar S, Cuddon P, TtoW EK, Saiki S, Siddiqi FH, Jahreiss L, Fleming A, Pask D, Goldsmith P, OKane CJ, Floto RA,
Rubinsztein DC (2008) Novel targets for Huntingtons disease in
an mTOR-independent autophagy pathway. Nat Chem Biol
4:295305
Wils H, Kleinberger G, Janssens J, Pereson S, Joris G, Cuijt I, Smits V,
Ceuterick-de Groote C, Van Broeckhoven C, Kumar-Singh S
(2010) TDP-43 transgenic mice develop spastic paralysis and
neuronal inclusions characteristic of ALS and frontotemporal lobar degeneration. Proc Natl Acad Sci USA 107:38583863
Wirth D, Gama-Norton L, Riemer P, Sandhu U, Schucht R, Hauser H
(2007) Road to precision: recombinase-based targeting technologies for genome engineering. Curr Opin Biotechnol 18:411419
Wisniewski T, Boutajangout A (2010) Immunotherapeutic approaches
for Alzheimers disease in transgenic mouse models. Brain Struct
Funct 214:201218
Wisor JP, Edgar DM, Yesavage J, Ryan HS, McCormick CM, Lapustea N, Murphy GM Jr (2005) Sleep and circadian abnormalities in
a transgenic mouse model of Alzheimers disease: a role for cholinergic transmission. Neuroscience 131:375385
Wittmann CW, Wszolek MF, Shulman JM, Salvaterra PM, Lewis J,
Hutton M, Feany MB (2001) Tauopathy in Drosophila: neurodegeneration without neuroWbrillary tangles. Science 293:711714

Hum Genet (2012) 131:535563


Wong AC, Draper BW, Van Eenennaam AL (2011) FLPe functions in
zebraWsh embryos. Transgenic Res 20:409415
Wood AJ, Lo TW, Zeitler B, Pickle CS, Ralston EJ, Lee AH, Amora
R, Miller JC, Leung E, Meng X, Zhang L, Rebar EJ, Gregory PD,
Urnov FD, Meyer BJ (2011) Targeted genome editing across species using ZFNs and TALENs. Science 333:307
Wu Y, Wu Z, Butko P, Christen Y, Lambert MP, Klein WL, Link CD,
Luo Y (2006) Amyloid-beta-induced pathological behaviors are
suppressed by Ginkgo biloba extract EGb 761 and ginkgolides in
transgenic Caenorhabditis elegans. J Neurosci 26:1310213113
Xi Y, Noble S, Ekker M (2011) Modeling neurodegeneration in zebraWsh. Curr Neurol Neurosci Rep 11:274282
Xu YF, Gendron TF, Zhang YJ, Lin WL, DAlton S, Sheng H, Casey
MC, Tong J, Knight J, Yu X, Rademakers R, Boylan K, Hutton
M, McGowan E, Dickson DW, Lewis J, Petrucelli L (2010) Wildtype human TDP-43 expression causes TDP-43 phosphorylation,
mitochondrial aggregation, motor deWcits, and early mortality in
transgenic mice. J Neurosci 30:1085110859
Yae K, Keng VW, Koike M, Yusa K, Kouno M, Uno Y, Kondoh G,
Gotow T, Uchiyama Y, Horie K, Takeda J (2006) Sleeping beauty
transposon-based phenotypic analysis of mice: lack of Arpc3
results in defective trophoblast outgrowth. Mol Cell Biol
26:61856196
Yang SH, Cheng PH, Banta H, Piotrowska-Nitsche K, Yang JJ, Cheng
EC, Snyder B, Larkin K, Liu J, Orkin J, Fang ZH, Smith Y, Bachevalier J, Zola SM, Li SH, Li XJ, Chan AW (2008) Towards a
transgenic model of Huntingtons disease in a non-human primate. Nature 453:921924
Yang D, Wang CE, Zhao B, Li W, Ouyang Z, Liu Z, Yang H, Fan P,
ONeill A, Gu W, Yi H, Li S, Lai L, Li XJ (2010) Expression of
Huntingtons disease protein results in apoptotic neurons in the
brains of cloned transgenic pigs. Hum Mol Genet 19:39833994
Young JW, Zhou X, Geyer MA (2010) Animal models of schizophrenia. Curr Top Behav Neurosci 4:391433
Zan Y, Haag JD, Chen KS, Shepel LA, Wigington D, Wang YR, Hu
R, Lopez-Guajardo CC, Brose HL, Porter KI, Leonard RA, Hitt

563
AA, Schommer SL, Elegbede AF, Gould MN (2003) Production
of knockout rats using ENU mutagenesis and a yeast-based
screening assay. Nat Biotechnol 21:645651
Zhang Y, Nash L, Fisher AL (2008) A simpliWed, robust, and streamlined procedure for the production of C. elegans transgenes via
recombineering. BMC Dev Biol 8:119
Zhang S, Feany MB, Saraswati S, Littleton JT, Perrimon N (2009)
Inactivation of Drosophila Huntingtin aVects long-term adult
functioning and the pathogenesis of a Huntingtons disease model. Dis Model Mech 2:247266
Zhang F, Cong L, Lodato S, Kosuri S, Church GM, Arlotta P (2011a)
EYcient construction of sequence-speciWc TAL eVectors for
modulating mammalian transcription. Nat Biotechnol 29:149
153
Zhang Y, Kashyap L, Ferguson AA, Fisher AL (2011b) The production of C. elegans transgenes via recombineering with the galK
selectable marker. J Vis Exp. doi:10.3791/2331
Zhou H, Huang C, Yang M, Landel CP, Xia PY, Liu YJ, Xia XG
(2009) Developing tTA transgenic rats for inducible and reversible gene expression. Int J Biol Sci 5:171181
Zhou H, Huang C, Chen H, Wang D, Landel CP, Xia PY, Bowser R,
Liu YJ, Xia XG (2010) Transgenic rat model of neurodegeneration caused by mutation in the TDP gene. PLoS Genet
6:e1000887
Zhuo L, Sun B, Zhang CL, Fine A, Chiu SY, Messing A (1997) Live
astrocytes visualized by green Xuorescent protein in transgenic
mice. Dev Biol 187:3642
Ziemienowicz A (2010) Plant transgenesis. Methods Mol Biol
631:253268
Zigman WB, Schupf N, Sersen E, Silverman W (1996) Prevalence of
dementia in adults with and without Down syndrome. Am J Ment
Retard 100:403412
Zimmerman L, Parr B, Lendahl U, Cunningham M, McKay R, Gavin
B, Mann J, Vassileva G, McMahon A (1994) Independent regulatory elements in the nestin gene direct transgene expression to
neural stem cells or muscle precursors. Neuron 12:1124

123

Das könnte Ihnen auch gefallen