You are on page 1of 13

JGG

Available online at www.sciencedirect.com

Journal of Genetics and Genomics 39 (2012) 421e433

REVIEW

Reverse Genetic Approaches in Zebrafish


Peng Huang a, Zuoyan Zhu a, Shuo Lin b,*, Bo Zhang a,*
a
Key Laboratory of Cell Proliferation and Differentiation of Ministry of Education, College of Life Sciences, Peking University, Beijing 100871, China
b
Department of Molecular, Cell & Developmental Biology, University of California, Los Angeles, CA 90095, USA

Received 10 June 2012; revised 3 July 2012; accepted 7 July 2012


Available online 11 August 2012

ABSTRACT

Zebrafish (Danio rerio) is a well-established vertebrate animal model. A comprehensive collection of reverse genetics tools has been
developed for studying gene function in this useful organism. Morpholino is the most widely used reagent to knock down target gene
expression post-transcriptionally. For a long time, targeted genome modification has been heavily relied on large-scale traditional forward
genetic screens, such as ENU (N-ethyl-N-nitrosourea) mutagenesis derived TILLING (Targeting Induced Local Lesions IN Genomes)
strategy and pseudo-typed retrovirus mediated insertional mutagenesis. Recently, engineered endonucleases, including ZFNs (zinc finger
nucleases) and TALENs (transcription activator-like effector nucleases), provide new and efficient strategies to directly generate site-
specific indel mutations by inducing double strand breaks in target genes. Here we summarize the major reverse genetic approaches
for loss-of-function studies used and emerging in zebrafish, including strategies based on genome-wide mutagenesis and methods for site-
specific gene targeting. Future directions and expectations will also be discussed.

KEYWORDS: Zebrafish; Reverse genetics; Morpholino; TILLING; Retrovirus; ZFN; TALEN; Gene targeting

1. INTRODUCTION identify genes and pathways involved in different develop-


mental processes. These forward genetic studies have been
Zebrafish (Danio rerio) has become a powerful model conducted using irradiation, murine leukemia virus (MLV) and
organism not only for the study of vertebrate development, but chemical mutagens such as the DNA alkylating agent N-ethyl-
also for human disease modeling as well as for drug discovery N-nitrosourea (ENU) (Fig. 1) (Chakrabarti et al., 1983; Walker
through small molecule screening at individual level. It is also and Streisinger, 1983; Lin et al., 1994; Mullins et al., 1994;
well established that genetic studies in zebrafish can provide Solnica-Krezel et al., 1994; Driever et al., 1996; Haffter et al.,
insights into the function of vertebrate genes and lead to better 1996). Several forward genetic screens have been carried out,
understanding of human diseases. During early period in through which large numbers of mutants have been isolated
zebrafish research, mutagenesis methods relied primarily on (Driever et al., 1996; Haffter et al., 1996; Amsterdam et al.,
forward genetics tools, which is an unbiased approach to 2004). The mutated genes are then identified by positional
cloning. Applications of forward genetic tools in zebrafish
Abbreviations: DSB, double strand break; ENU, N-ethyl-N-nitrosourea; have been summarized in several excellent reviews (Patton
HR, homologous recombination; MLV, murine leukemia virus; MO, mor- and Zon, 2001; Lieschke and Currie, 2007; Lawson and
pholino; TALE, transcription activator-like effector; TALEN, TALE nuclease; Wolfe, 2011). However, forward genetic screens are intrinsi-
TILLING, Targeting Induced Local Lesions IN Genomes; ZFA, zinc finger cally limited in their effectiveness to isolate mutations of every
array; ZFN, zinc finger nuclease.
single gene due to functional redundancy between different
* Corresponding authors. Tel/fax: þ86 10 6275 9072 (B. Zhang); Tel: þ1
310 267 4970, fax: þ1 310 267 4971 (S. Lin). genes and the need to have measurable phenotypes. This is
E-mail addresses: shuolin@ucla.edu (S. Lin); bzhang@pku.edu.cn (B. Zhang). especially noteworthy for zebrafish since it has undergone an
1673-8527/$ - see front matter Copyright Ó 2012, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, and Genetics Society of China.
Published by Elsevier Limited and Science Press. All rights reserved.
http://dx.doi.org/10.1016/j.jgg.2012.07.004
422 P. Huang et al. / Journal of Genetics and Genomics 39 (2012) 421e433

additional genome duplication event during teleost evolution. activator-like effector nuclease (TALEN), are emerging as
In addition, it is also quite time-consuming but without a promising and efficient new method to achieve directly site-
guarantees to identify the responsible mutated gene. It is specific genome modification (Fig. 1) (Doyon et al., 2008;
therefore also desirable to first identify genes of interest and Meng et al., 2008; Huang et al., 2011; Sander et al., 2011a). In
subsequently obtain their mutations followed by the evaluation this review, we summarize the major reverse genetics tools
of the phenotypic effects of these mutations (i.e., “reverse” applicable for loss-of-function studies in zebrafish and
genetics). particularly the recent developments and improvements of
Genome sequencing projects of model organisms such as each strategy (Fig. 2). We also anticipate the future directions
zebrafish could document each transcript with protein-coding and open questions towards reverse genetic approaches, which
and non-coding genes within genomes. With zebrafish are still unavailable but crucial for zebrafish research.
genome sequencing project nearly complete, it is now theo-
retically possible to generate mutations in all zebrafish genes
to systematically evaluate their functions, which requires 2. HUNTING FOR TARGET GENES
powerful reverse genetics tools. Unlike mice and Drosophila,
targeted gene knock-out through homologous recombination The first step towards reverse genetic study is to find
(HR) is still unavailable in zebrafish. The existence of a target gene or a group of target genes for mutagenesis. There
embryonic stem (ES) cells in zebrafish is still an open ques- are several ways to achieve this purpose, including searching
tion. Instead, several alternative reverse genetics strategies for for novel and functional unknown genes through bio-
loss-of-function studies have been developed, some of which informatics analysis and data mining from available cDNA
were derived from forward genetics approaches, such as and genomic information, picking up potentially important
mutagenesis screens based on retrovirus or transposons, Tar- genes whose homologs have only been studied in lower
geting Induced Local Lesions IN Genomes (TILLING) (Lin organisms such as Drosophila and Caenorhabditis elegans, or
et al., 1994; Kawakami et al., 2000; Wienholds et al., 2002). screening for tissue-specifically expressed genes by in situ
In addition, antisense morpholino (MO) oligonucleotide hybridization or various genome-wide insertional traps medi-
analogs can be used to disrupt target gene expression without ated by transposons or retroviruses, such as enhancer trap,
changing its genetic information (Nasevicius and Ekker, gene trap, poly A trap and promoter trap, etc (Fig. 2). Here we
2000). Recently, engineered endonucleases (EENs), only briefly summarize the gene hunting strategies based on
including zinc finger nuclease (ZFN) and transcription Tol2 transposon as examples.

Fig. 1. A historical view of the development of some mutagenesis tools in zebrafish.


Note that the whole issue for Volume 123 of Development (1996) was dedicated to reporting the results from two major large-scale chemical mutagenesis screens
based on ENU, therefore, no author names were mentioned in the corresponding box in this figure. EMS, ethyl methanesulfonate; ENU, N-ethyl-N-nitrosourea;
TALEN, transcription activator-like effector nuclease; TILLING, Targeting Induced Local Lesions IN Genomes; ZFN, zinc finger nuclease.
P. Huang et al. / Journal of Genetics and Genomics 39 (2012) 421e433 423

Fig. 2. Strategies for loss-of-function studies by gene targeting in zebrafish.


Target genes (i.e., genes of interest) could be identified through bioinformatics analysis, searching for homologous genes from other species, screening for genes
with tissue specific expression patterns by in situ hybridization or various genome-wide insertional traps including enhancer trap, gene trap, etc. For functional
study of target genes, several mutagens could be used to create loss-of-function mutations in zebrafish, including retrovirus, transposons, ENU and EENs (ZFNs
and TALENs), while morpholino could be used as phenocopy agents to knock down gene expression post-transcriptionally. Researchers can screen for point
mutations using TILLING in a defined gene from the mutant archives generated through ENU mutagenesis. One can also look up insertional mutations for
a specific gene in the mutant archives generated through retrovirus insertional mutagenesis. Conversely, one can also search for their genes of interest from genetic
screens mediated by retrovirus or transposons. Loss-of-function indel mutations of target genes can be achieved with ZFNs, or more efficiently, TALENs. Large
genomic deletions might be achieved through imprecise excision of Tol2 transposon or cleavage of two pairs of EENs. Another potential application of transposons
and EENs is to stimulate HR by inducing DSBs. The solid line arrows indicate that the corresponding methods are already in practice, while the dotted arrows
indicate that the corresponding methods are only hypothetical and have not been established at present. ENU, N-ethyl-N-nitrosourea; HR, homologous recom-
bination; KI, knock-in; KO, knock-out; NHEJ, non-homologous end joining; TALEN, transcription activator-like effector nuclease; TILLING, Targeting Induced
Local Lesions IN Genomes; ZFN, zinc finger nuclease.

Transposons could integrate into genome sequence well as enhancer trap screens, which is similar to the pseudo-
randomly and efficiently, and thus are widely used to generate typed retrovirus used for creating insertional mutations in
transgenic fish lines. In addition to transgenesis, transposons zebrafish (Kondrychyn et al., 2009).
containing various sequence cassettes have also been used for Recently, three groups reported strategies to achieve
genetic screens of different purposes. Several transposons have conditional gene inactivation with various gene-trap cassettes
been shown to be functional in zebrafish, including Tol1, Tol2, using transposons, where one can simultaneously obtain
Sleeping Beauty and Ac/Ds elements, while Tol2 transposon is a conditional allele while identifying the genes of interest
more frequently used due to its high efficiency and ease to (Clark et al., 2011; Maddison et al., 2011; Trinh et al., 2011).
manipulate (Kawakami et al., 2000; Davidson et al., 2003; Clark et al. (2011) applied a gene-breaking mutagenic cassette
Emelyanov et al., 2006; Koga et al., 2008). Several large-scale with a terminator that can disrupt the trapped protein. Since
genetic screens based on Tol2 transposon, which are mainly the mutagenic cassette contains splicing acceptor elements
enhancer trap and gene trap, have been carried out and flanked by two loxP sites, the trapped genes can be rescued by
a collection of transgenic fish lines with known insertion sites Cre recombinase or MOs targeting the splicing sites.
are currently available (Kawakami et al., 2004; Parinov et al., Maddison et al. (2011) used a Tol2 transposon based Flip and
2004; Zhao et al., 2008; Kondrychyn et al., 2009; Tian et al., Excision (FlEx) module which contains a mCherry-EGFP dual
2009; Xue et al., 2010). One can identify their genes of interest reporter and loxP and FRT elements. By combination with
through the analysis of the expression pattern of the reporter tissue and temporal specific expressed Cre or Flp transgenic
gene and genomic sequences flanking the insertion sites. fish lines, FLEx module could invert and conditionally inac-
Enhancer trap lines usually do not disrupt endogenous genes tivate target gene expression (Maddison et al., 2011). Similar
while the genes identified from gene trap are generally already to protein-trap strategy used in Drosophila, Trinh et al. (2011)
mutation alleles. Strictly speaking, since gene trap could performed another screening using Tol2 transposon and Ac/Ds
reveal genes and their mutations at the same time, it is more elements with a “FlipTrap” cassette, which contains a Citrine
accurate to be considered as a mixed strategy of forward reporter flanked by a splicing acceptor and donor. In combi-
genetics and reverse genetics approaches. Curiously, our and nation with transgenic fish lines carrying Cre recombinase, the
others’ results suggest that Tol2 transposon tends to insert into Citrine reporter and splicing donor elements can be flipped-out
transcribed regions in the genome revealed by gene trap as in a tissue and temporal manner, which could generate
424 P. Huang et al. / Journal of Genetics and Genomics 39 (2012) 421e433

a truncated protein of the trapped gene (Trinh et al., 2011). combination with other genetic manipulations, such as injec-
Although alleles generated with these strategies are not tion into a transgenic fish line labeled with certain fluores-
complete knock-outs and genes of interest cannot be directly cence reporters/markers. Since the additional duplication
targeted, they provide useful resources to investigate gene event of zebrafish genome occurs during teleost evolution,
function in a conditional manner, especially when HR is many genes might show functional redundancy with their
currently unavailable in zebrafish. paralogs. In this case, another advantage of MOs is that
researchers can knock-down a number of genes at the same
3. GENE KNOCK-DOWN VIA PHENOCOPY time by injection of a mixture of corresponding MOs, which is
rather complicated and time-consuming to achieve through the
Zebrafish is originally discovered as an ideal embryological breeding between genetic mutants.
model to study embryogenesis due to its transparency from MO knocking-down is very convenient but also has obvious
very beginning, and soon also considered to be a good limitations. With the increase of cell numbers, the amount of
organism for genetic screens due to its capability of producing MOs per cell is gradually diluted along with embryo devel-
large quantity of offspring. However, it had been a big chal- opment. Needless to say, phenocopy by MOs is not heritable,
lenge to explore reverse genetic study in zebrafish, and it is and it has been shown that MOs are only sufficiently effective
only until recently that bona fide direct site-specific gene within the first 5 days of development when injected into one
targeting methods are gradually established. Before that, most cell stage embryos (Eisen and Smith, 2008). In addition, MOs
loss-of-function studies have heavily relied on knocking-down tend to activate p53 pathway and therefore lead to non-specific
target gene expression via MOs. MOs are a kind of artificially phenotypes of embryonic defects (Robu et al., 2007).
synthesized oligonucleotide analogs which use morpholine Although it can be overcome by co-injection of anti-p53 MOs
rings to replace the ribose backbone of nucleic acids or performing all the experiments at p53 mutant background,
(Summerton and Weller, 1997). The replacement with analo- sometimes it is still unconvincing, especially when the target
gous structures enables MOs to be resistant to nuclease genes have functional interactions with p53 pathway. Last but
digestion in addition to its enhanced binding activity to their not least, sometimes it is difficult to evaluate the true effect
complementary RNA sequences in cells (Summerton, 1999). and specificity of MO knocking-down; therefore, properly and
By this way, MOs can exert their effects post-transcriptionally carefully designed control experiments and rescue experi-
by preventing target genes from expression either by poising/ ments are crucial before reaching any conclusion.
attenuating protein translation or by disrupting normal RNA interference (RNAi) is another powerful technique for
splicing. Using an antisense MO complementary to the 50 - post-transcriptional gene silencing studies, which has been
UTR (untranslated region) sequence, the translation of mature successfully used in many species. Unfortunately, this tech-
mRNA could be blocked in the cytoplasm, while by binding to nique is still not well established in zebrafish though certain
a region spanning splicing donor or splicing accepter site, successes have been reported (Zhao et al., 2001; Su et al.,
MOs could disrupt normal splicing of pre-mRNA in the 2008a, 2008b; Dong et al., 2009). Recently, a microRNA-
nucleus. MOs could also be used to block microRNA func- based shRNA (mir-shRNA) strategy has been shown to be
tions by binding to the mature microRNA or primary micro- able to down regulate endogenous gene expression in a defined
RNA (Eisen and Smith, 2008). In the narrow sense, MO is spatial and temporal manner, which opened up the possibility
more accurate to be classified as phenocopy rather than to do conditional gene knock-down studies in zebrafish (Dong
a fundamental reverse genetics approach. et al., 2009).
To study gene functions during early development, MOs are
usually injected directly into zebrafish embryos at 1e2 cell 4. REVERSE GENETICS TOOLS BASED ON
stage. However, other delivery methods have also been GENOME-WIDE LARGE-SCALE SCREENING
developed and used for different purposes, such as injection of
MOs into the yolk at mid-blastula stages to specifically knock- 4.1. Retrovirus mediated insertional mutagenesis
down genes in the dorsal forerunner cells, and delivering MOs
by electroporation to the fin or retina in adult fish to study the A pseudo-typed retrovirus, which was derived from MLV
mechanisms of regeneration (Amack and Yost, 2004; with its envelope protein replaced by the glycoprotein G from
Thummel et al., 2006, 2011). The applications of MOs have vesicular stomatitis virus (VSV), has been shown to be able to
also been extended to extra modifications. Fluorescein labeled infect zebrafish embryos and transmit efficiently through
MOs allow examinations of the cell populations whose target germline following insertion into the genomes of zebrafish
gene expression has been inhibited (Hyde et al., 2012). Caged (Lin et al., 1994). As in human cells, this modified MLV
MOs or photo-MOs can be activated or inactivated under prefers to integrate into the genomic regions close to tran-
ultraviolet (UV) light and used for conditional gene knock- scription start sites in zebrafish genome, which makes it an
down in a defined tissue and temporal manner (Shestopalov ideal alternative mutagen besides ENU (Wu et al., 2003; Wang
et al., 2012; Tallafuss et al., 2012). et al., 2007). A unique advantage of retroviral mutagenesis
Unlike genetic mutants, embryos injected with MOs do not over chemical mutagens is that it allows rapid identification of
need to be analyzed by applying Mendel’s law of segregation the mutated genes. By using the retroviral sequence as
nor confirmation by genotyping. MOs can be used in a molecular “tag” for mapping, the genomic sequence flanking
P. Huang et al. / Journal of Genetics and Genomics 39 (2012) 421e433 425

Fig. 3. Strategies of retrovirus based insertional mutagenesis.


Zebrafish embryos are injected at 1000e2000 cell stage with the retrovirus preparation and raised to adulthood as founders. F1 families are collected by inbreeding or
outbreeding of founders. For the first generation strategy of mutagenesis, the number of proviral insertions is estimated in each F1. F2 pools are collected through
inbreeding F1 families with high insertion numbers. Phenotype analysis is carried out in F3 embryos. In the strategies for the second and third generations, sperms
from each F1 male are collected and archived, and the corresponding genomic DNA was extracted from fin clips and the proviral insertion sites were identified
through LM-PCR and sequencing. Desired insertions can be restored by index to the cryopreserved sperms and in vitro fertilization. LM-PCR, linker-mediated PCR.

the insertion site can be easily identified through inverse PCR successfully identified until 2004 (Amsterdam et al., 1999,
or linker-mediated PCR (LM-PCR). Since the original muta- 2004; Golling et al., 2002).
genesis strategy using retrovirus in zebrafish is a phenotype- However, there are certain limitations for this phenotype-
based screen, it is more accurate to be classified as a forward based mutagenesis screen. For example, genes that share
genetics strategy (Amsterdam, 2003). Essentially, 1000e2000 functional redundancy or do not create visible phenotypes
cell stage zebrafish embryos are infected with purified and during early development are usually ignored. Also, this
highly concentrated retroviruses through micro-injection. strategy is not practical for saturation mutagenesis screen due to
These embryos are raised to adulthood as founders and its demanding for intensive labor and large space to maintain
incrossed or outcrossed to get F1 families. Individual F1 fish numerous fish. With the development of zebrafish genome
with high proviral insertions is selected by Southern blot. sequence project, the genomic sequence is largely known.
Different F2 pools are generated by F1 incross. Multiple pairs Based on this information, researchers designed a new strategy
are mated in each F2 pool. If a visible phenotype is observed in to achieve high throughout retroviral insertional mutagenesis
F3 embryos (usually within the first 5 days of development by employing cryopreservation of sperms from F1 fish in
after fertilization), the retroviral insertion site and the corre- combination with identification of all the proviral insertions in
sponding disrupted gene can be identified and mapped by PCR the corresponding male fish instead of phenotype screening in
(Fig. 3) (Amsterdam et al., 1999). Using this approach, 315 their offspring (Wang et al., 2007). In this so-called second
genes essential for zebrafish development have been generation retroviral insertional mutagenesis strategy, sperms
426 P. Huang et al. / Journal of Genetics and Genomics 39 (2012) 421e433

of each F1 male are stored as frozen stocks and the genomic Since TILLNG requires large mutant libraries as well as
DNA is extracted from tail tissue in order to identify the cor- significant DNA sequencing capacity in order to identify
responding proviral insertions. By this way, a proviral insertion a desired mutation, it is labor-intensive and generally not
library can be generated and indexed, irrespective of any feasible for a regular laboratory. Therefore, a zebrafish
phenotypic effect. Any desired insertion (i.e., potential muta- mutation project has been organized by Sanger Institute using
tion) could readily be recovered through in vitro fertilization of TILLING method (http://www.sanger.ac.uk/Projects/D_rerio/
the frozen sperm sample carrying the integrations of interest zmp/). Researchers could submit an inquiry for desired
(Fig. 3) (Wang et al., 2007). Another obvious advantage for this mutations by simply providing required information of their
strategy is the release from overload of fish tanks and costs both genes of interest. To date, mutations for 6092 genes (about
for the screening process and for the subsequent maintenance of 23% of all zebrafish genes) have been identified by this
fish lines carrying proviral insertions. As a proof of theory, project. All the mutation lines can be searched and requested
a pilot experiment has been carried out since 2005 and more by researchers. However, since the mutation frequency of
than 500 gene hits (i.e., proviral insertions within introns or ENU is about 1 per 105 bp, genes consisting of only small
exons) have been identified in less than 5 years (Wang et al., exons are difficult to be mutated using ENU based TILLING
2007). Since researchers can select their genes of interest method (Moens et al., 2008; de Bruijn et al., 2009).
from the list of insertion sites before going for phenotype
analysis, this strategy in a way should be considered as a hybrid
between forward and reverse genetic approaches. Since this 5. GENE TARGETING BY SITE-SPECIFIC GENOME
strategy does not produce any bias towards insertion site MODIFICATION
identification, one can estimate for the first time the pattern of
retroviral insertions at organism level. Interestingly, about 40% 5.1. Gene targeting via ZFNs
of the proviral insertions landed in putative gene loci (introns
and exons), with w50% of intron insertions located in the 1st Transcription factors such as those from zinc finger protein
introns, which is consistent with the observation in mammalian family could recognize and bind to specific DNA sequences.
in vitro cultured cells (Wu et al., 2003; Wang et al., 2007). Researchers have tried hard to adopt these properties in site-
Inbreeding fish carrying same proviral integrations in different specific genome modifications since the discovery of tran-
genes demonstrated that the mRNA transcript levels were scription factors. ZFN has become one of the first successful
reduced by 70% or more in approximately 50% of the gene examples for such a purpose. ZFN is a chimeric protein con-
“hits”, with the highest probability for mutation occurring if the taining a DNA binding domain comprising a C2H2 type zinc
integration was in an exon or the 1st intron. This strategy finger array (ZFA) mainly based on the backbone of ZIF268,
therefore produces one mutagenic event from every five a human or mouse zinc finger protein, and a cleavage domain
proviral integrations. In addition, the mapping occurs in the F1 derived from Fok I, a bacterial non-specific endonuclease
generation, thus making recovery of the mutations compara- (Urnov et al., 2010). The alpha helix domain of each zinc
tively simple and more reliable. Again, this strategy also has its finger recognizes and binds to a specific 3 bp DNA sequence.
intrinsic disadvantages, among which mapping insertion sites Each ZFA usually contains three to six zinc finger motifs,
becomes the most labor-intensive step. Recently, in order to which could recognize a total of 9e18 bp target DNA
improve the mapping efficiency of proviral insertion sites as sequence. Since Fok I has to dimerize to become fully func-
well as reduce the sequencing costs, a third generation strategy tional, ZFNs usually function in pairs, where they could cleave
is conceived, which incorporates the next-generation high- the spacer sequence between the two ZFN binding sites in the
throughput sequencing technology by pooling the genomic chromosomal DNA and induce double strand breaks (DSBs) at
DNA samples from several hundred F1 fish into one sequencing its target locus. The DSB could be repaired through error-
reaction with “barcode” labeling (Fig. 3). prone non-homologous end joining (NHEJ) or homologous
template dependent HR pathway, leading to either indel
4.2. Gene knock-out via TILLING mutations or DNA replacement (Urnov et al., 2010). Another
application for genome modification with ZFNs is the induc-
TILLING is a method that is used to identify individual tion of large genomic deletions, where two pairs of ZFNs were
organisms harboring point mutations in a defined gene from introduced into the same cell or organism and the two DSBs
a library of potential mutants generated through chemical generated by the two ZFN pairs could join together, resulting
mutagenesis. First established in Arabidopsis and soon adopted in the deletion of the genomic region between the two
into zebrafish, TILLING has been demonstrated to be an effec- distantly separated ZFN target sites (Lee et al., 2010). So far,
tive approach to identify mutations within a sufficiently big ZFN technology has been successfully applied in in vitro
population of ENU-mutagenized zebrafish (Wienholds et al., cultured cells as well as many model organisms including
2002). At first, CEL I enzyme is used to identify point muta- zebrafish (Bibikova et al., 2003; Porteus and Baltimore, 2003;
tions induced by ENU (Wienholds et al., 2002). Later on, the low Lombardo et al., 2007; Doyon et al., 2008; Meng et al., 2008;
cost next-generation sequencing and whole exome enrichment Foley et al., 2009; Geurts et al., 2009; Shukla et al., 2009;
technologies were incorporated to facilitate TILLING method to Townsend et al., 2009; Dong et al., 2011; Yang et al., 2011; Yu
identify large numbers of alleles rapidly from mutant libraries. et al., 2011).
P. Huang et al. / Journal of Genetics and Genomics 39 (2012) 421e433 427

In zebrafish, target genes are mainly disrupted by intro- ZFNs represent the first available direct site-specific gene
ducing indels generated through NHEJ pathway, while ZFN targeting method in zebrafish. It brought out the possibility to
mediated HR or large genomic deletion has not been reported knock-out any desired zebrafish gene. However, selection of
(Fig. 4). To date, dozens of zebrafish mutations have been ZFAs with highly efficient and specific DNA binding activity
obtained through ZFNs since the first reports on successful remains a big challenge and becomes the limiting step in ZFN
gene targeting through ZFNs in zebrafish in 2008 (Doyon applications. Commercial service for ZFN screening and
et al., 2008; Meng et al., 2008; Foley et al., 2009; Siekmann assembly is available but the cost is considerably high. To get
et al., 2009; Ben et al., 2011; Gupta et al., 2011, 2012; Zhu ZFAs with high activities, several methods have been devel-
et al., 2011; Sander et al., 2011a, 2011b). oped, either via direct assembly of known zinc fingers or using

Fig. 4. Gene targeting via engineered endonuclease ZFNs or TALENs.


A pair of mRNAs encoding ZFNs or TALENs is injected into zebrafish embryos. After evaluation of the efficiency by one of the four assays (restriction enzyme
digestion, CEL I or T7 endonuclease digestion, sequencing analysis, melting curve analysis), the mosaic embryos are raised to adulthood as founders. Individual
mutations are identified from F1 embryos obtained by outcross of founders with wild type fish. F1 embryos are raised to adulthood and carriers are identified by fin clip
genotyping. Phenotype is analyzed in F2 through incrossing heterozygous F1 fish. Each TALEN is usually composed of more than 12 TALE repeat units; here we only
show 9 repeats on each side for simplicity. TALE, transcription activator-like effector; TALEN, TALE nuclease; ZFA, zinc finger array; ZFN, zinc finger nuclease.
* represents bands of DNA fragments carrying potential mutations.
428 P. Huang et al. / Journal of Genetics and Genomics 39 (2012) 421e433

screening strategies. Modular assembly (MA) is one of the a promising alternative sequence-specific DNA binding domain
earliest methods used to generate functional ZFAs, which to ZFA (Boch et al., 2009; Moscou and Bogdanove, 2009).
directly ligates identified zinc fingers that recognize different To achieve better specificity, one usually needs to construct
triplet DNA combinations. Since MA does not consider TALENs each targeting more than 12 bp, and theoretically the
context-dependent effects of the nucleotide sequence for the longer the better. However, due to its highly repetitive feature,
target site recognition by zinc fingers, it has a relatively high it is difficult to construct long TALE repeats. Several methods
failure rate, especially when the target sites are devoid of or have been developed to resolve this issue and most are based
poor in GNN (N represents any nucleotide) nucleotide triplets on Golden Gate cloning strategy, which employs type IIs
(Ramirez et al., 2008). Oligomerized pool engineering restriction enzymes and theoretically can ligate up to dozens
(OPEN) method employs a selection system based on bacterial of repeats through a single ligation reaction (Fig. 5) (Christian
two-hybridization (Maeder et al., 2008), while Meng et al. et al., 2010; Morbitzer et al., 2010, 2011; Cermak et al., 2011;
(2008) used a bacterial one-hybrid selection system, which Geibetaler et al., 2011; Li et al., 2011a, 2011b, 2012a;
was first developed for the identification of target sites for Mahfouz et al., 2011; Sander et al., 2011a; Reyon et al., 2012).
transcription factors. Numerous functional ZFAs have been We developed a simple alternative method called “Unit
generated using these two selection systems. Although the Assembly”, which took advantages of a pair of isocaudomers
ZFNs obtained by these two methods show higher success Nhe I and Spe I. Using four single-unit vectors as the starting
rates than MA, both strategies are labor-intensive and require material, which contain the coding sequence of alternative
constructions of big libraries as well as expertise. Several units recognizing A, T, C or G, TALE repeats can be easily
databases have collected identified ZFAs (http://bindr. assembled by standard molecular cloning method of restriction
gdcb.iastate.edu/ZiFDB/, http://web.iitd.ac.in/wsundar/zifbase/ enzyme digestion and ligation with any and unlimited number
and http://eendb.zfgenetics.org/). Utilizing the verified ZFA of repeats at any desired order (Fig. 5) (Huang et al., 2011).
archives, an improved screening-independent method called More recently, two high-throughput methods for TALE repeats
Context-dependent assembly (CoDA) was reported (Sander assembly called FLASH (fast ligation-based automatable
et al., 2011b). Comparing with MA, CoDA resolves the solid-phase high-throughput) and ICA (iterative capped
context-dependent effects by selecting N- and C-terminal zinc assembly) were reported, which bypass the time-consuming
fingers from identified ZFAs that contain a common middle intermediate steps of bacterial culture and amplification by
zinc finger. Fifty percent ZFNs generated with CoDA method using solid-phase magnetic beads for enzyme digestion and
showed sufficient in vivo activities (Sander et al., 2011b). ligation and allow researchers to construct large numbers of
Nevertheless, targeting any desired locus in a genome is still TALENs rapidly (Fig. 5) (Briggs et al., 2012; Reyon et al.,
difficult using ZFNs, which limits its broad application. 2012). However, the advantages and disadvantages of all
these different TALEN construction methods still need further
5.2. Gene targeting via TALENsdthe most promising investigation.
reverse genetic tool Although TALEN targeting as a technique was reported
only less than 2 years ago, it is soon broadly applied in in vitro
TALE proteins are mainly found in Xanthomonas, a plant cultured cells as well as living organisms including zebrafish
pathogen. The DNA binding domain of TALE proteins is (Hockemeyer et al., 2011; Huang et al., 2011; Miller et al.,
composed of several tandem repeat units, while each repeat is 2011; Sander et al., 2011a; Tesson et al., 2011; Wood et al.,
responsible to recognize and bind to a single nucleotide in the 2011; Cade et al., 2012; Liu et al., 2012; Moore et al., 2012;
target site (Boch et al., 2009; Moscou and Bogdanove, 2009). Tong et al., 2012; Li et al., 2012b). TALENs function in
The major and classical TALE repeat unit contains 34 amino a similar way as ZFNs, and they could also induce indel
acid residues where the two at positions 12 and 13 are called mutations to disrupt the sequence in the spacer of TALEN
RVD (repeat variable di-residue), which determines the speci- target sites (i.e., the region between the two TALEN binding
ficity of the repeat unit to recognize and bind to its target sites), but usually with higher targeting efficiency and lower
nucleotide. The most representative RVDs, i.e., the most off-target effects (Hockemeyer et al., 2011; Huang et al., 2011;
frequently occurred RVDs in naturally existing TALE proteins, Li et al., 2011b). Therefore, a unique restriction enzyme site in
which correspond for the recognition of each of the four the spacer region is very convenient for the detection of
nucleotides A, T, C and G, are NI (Asn Ile), NG (Asn Gly), HD TALEN efficiency. After PCR amplification of the sequence
(His Asp), NN (Asn Asn)/NK (Asn Lys), respectively. Two flanking the target site and digestion with the corresponding
most recent publications report that NH (Asn His) has enzyme, the efficiency of TALENs can be estimated by
a comparable activity but higher specificity than NN to recog- measuring and comparing the density and ratio of the undi-
nize nucleotide G (Cong et al., 2012; Streubel et al., 2012). In gested band (Fig. 4). In addition, TALEN efficiency could be
addition, an interesting and mysterious feature for the target site revealed directly by sequencing after PCR amplification or
recognition of TALE proteins is that they require a T as the evaluated through other assays such as CEL I or T7 endonu-
nucleotide immediately upstream of the 50 end of its binding clease digestion and melting curve analysis (Fig. 4). So far,
site. Nevertheless, comparing with zinc finger proteins, the nearly eighty TALEN pairs targeting different genomic loci in
principle for the target nucleotide recognition and binding for zebrafish have been constructed in our laboratory and the
TALE is much simpler and predictable, which makes it overall success rate is above 70%, which is similar to a recent
P. Huang et al. / Journal of Genetics and Genomics 39 (2012) 421e433 429

Fig. 5. Strategies for the construction of TALE repeats.


One-step assembly strategy for TALE repeats construction is based on Golden Gate cloning derived methods, which take advantages of type IIs enzymes and can
theoretically construct TALE repeats by a one-step digestion and ligation reaction, though it can also divide into more than one serial steps. Unit Assembly and
REAL represent the strategy of serial assembly, which can construct TALEs with any and unlimited number of repeat units through repeated cycles of standard
restriction enzyme digestion and ligation steps. Using a strategy involving solid-phase ligation, FLASH and ICA methods bypass the time-consuming trans-
formation and growing of bacteria during intermediate cloning steps and can construct TALE repeats in a high-throughput manner. FLASH, fast ligation-based
automatable solid-phase high-throughput; ICA, iterative capped assembly; REAL, restriction enzyme and ligation; TALE, transcription activator-like effector.

publication (Cade et al., 2012). Among all the TALEN pairs for zebrafish, and perhaps, for other organisms as well. This
tested, more than half of them showed >30% mutagenizing situation reminds us of the discovery of RNA interference
activity in founder embryos, some of which even reached the (RNAi) about 14 years ago, which has achieved a strong
efficiency to nearly 100%, as detected by restriction enzyme impact to the field of study on gene functions shortly after. We
digestion assay (unpublished data), suggesting that TALEN is believe TALEN has already started to bring a similar revolu-
a promisingly efficient and easy technology for gene targeting tion to the field of reverse genetic studies.
in zebrafish. However, about 30% TALEN pairs did not show
detectable mutagenizing activity in zebrafish and the reason is 6. CONCLUDING REMARKS
still unclear. In addition to the unpredictable and variable
efficiency, another limitation for the applications of TALENs Genome manipulations using powerful reverse genetics
in zebrafish is that they can only induce indel mutations but tools will greatly enhance the advantages and utility of
not precise modification of the target site at present, though we zebrafish as a vertebrate model organism. With the expansion
believe this limitation is only temporary and will soon be of zebrafish genetic toolkit, ingenious reverse genetics
overcome in the near future. Nevertheless, the ease in finding methods will certainly turn zebrafish mutagenesis into
suitable target sites, the high success rate, high efficiency and a convenient and important process. Apart from point muta-
specificity of TALEN targeting confer this relatively new tions isolated from TILLING, insertional mutations identified
reverse genetic strategy to be the most promising technique to from retrovirus or Tol2 transposon mutagenesis screening and
be able to target virtually any genes of interest in any species. indel mutations induced by EENs (ZFNs or TALENs), there
It will not be surprising if TALEN eventually becomes the are other types of mutations and mutagenesis methods which
most powerful and popular site-specific mutagenesis method are of equal or even more importance comparing with the
430 P. Huang et al. / Journal of Genetics and Genomics 39 (2012) 421e433

currently available ones. Large genomic fragment deletion and ACKNOWLEDGEMENTS


HR based gene knock-in and conditional knock-out are among
the most interesting ones. We thank X. Tong, Y. Shen, A. Xiao, L. Xu, Z. Wang, Y.
There are at least two potential approaches to achieve large Zu, Y. Hu, Z. Luo and Q. Wu for helpful discussions; Y. Gao,
genomic deletions. Although only indel mutations have been J. Zhang, Y. Jia, J. Chen, X. Yang and H. Cui for technical
reported for TALEN targeting in zebrafish so far, our support. This work was partially supported by the grants from
preliminary data showed that TALENs also hold promise to National Program on Key Basic Research Project (973
induce large genomic deletions when using two pairs of program) (Nos. 2012CB945101 and 2011CBA01000) and
TALENs separated within a reasonable distance (Fig. 2 and National Natural Science Foundation of China (NSFC) (Nos.
unpublished data). In addition to disrupting genes by inser- 31110103904 and 30730056).
tions, transposon can also be used for mutagenesis upon
excision. Deletion of large genomic sequence by inducing P
element imprecise excision at a defined locus has been widely
REFERENCES
used in Drosophila as a powerful, and during certain period of
time, the only method for the study of gene function and Amack, J.D., Yost, H.J., 2004. The T box transcription factor no tail in ciliated
regulation through loss-of-function mutations. We found that cells controls zebrafish left-right asymmetry. Curr. Biol. 14, 685e690.
Tol2 transposon excision can also induce large chromosomal Amsterdam, A., 2003. Insertional mutagenesis in zebrafish. Dev. Dyn. 228,
deletions in zebrafish (Fig. 2 and unpublished data). Consid- 523e534.
ering the availability of large and still increasing numbers of Amsterdam, A., Burgess, S., Golling, G., Chen, W., Sun, Z., Townsend, K.,
Farrington, S., Haldi, M., Hopkins, N., 1999. A large-scale insertional
transgenic zebrafish carrying Tol2 insertions, this strategy mutagenesis screen in zebrafish. Genes Dev. 13, 2713e2724.
could become an alternative gene targeting method and could Amsterdam, A., Nissen, R.M., Sun, Z., Swindell, E.C., Farrington, S.,
be used for the complete disruption/removal of a single gene Hopkins, N., 2004. Identification of 315 genes essential for early zebrafish
or a cluster of genes simultaneously. This is especially useful development. Proc. Natl. Acad. Sci. USA 101, 12792e12797.
Ben, J., Elworthy, S., Ng, A.S., van Eeden, F., Ingham, P.W., 2011. Targeted
for loss-of-function studies of non-coding RNA genes since
mutation of the talpid3 gene in zebrafish reveals its conserved requirement
point mutations or small indel mutations might not be enough for ciliogenesis and Hedgehog signalling across the vertebrates. Devel-
to disrupt the function of these non-coding RNAs which do not opment 138, 4969e4978.
have a predictable open reading frame. Bibikova, M., Beumer, K., Trautman, J.K., Carroll, D., 2003. Enhancing gene
Other advanced genetic tools such as conditional knock- targeting with designed zinc finger nucleases. Science 300, 764.
out and precise gene knock-in/replacement are widely used Briggs, A.W., Rios, X., Chari, R., Yang, L., Zhang, F., Mali, P., Church, G.M.,
2012. Iterative capped assembly: rapid and scalable synthesis of repeat-
in mouse and Drosophila and proved to be very useful. module DNA such as TAL effectors from individual monomers. Nucleic
Although some preliminary work has been performed, gene Acids Res. 40, e117.
targeting through HR is still not established in zebrafish Boch, J., Scholze, H., Schornack, S., Landgraf, A., Hahn, S., Kay, S.,
(Hagmann et al., 1998; Cui et al., 2003; Wu et al., 2006). The Lahaye, T., Nickstadt, A., Bonas, U., 2009. Breaking the code of DNA
next, and most likely also the last and the biggest, challenge binding specificity of TAL-type III effectors. Science 326, 1509e1512.
Cade, L., Reyon, D., Hwang, W.Y., Tsai, S.Q., Patel, S., Khayter, C.,
for zebrafish reverse genetics studies is therefore to achieve Joung, J.K., Sander, J.D., Peterson, R.T., Yeh, J.R., 2012. Highly efficient
HR mediated gene targeting, which is crucial for gene knock- generation of heritable zebrafish gene mutations using homo- and heter-
in as well as conditional mutagenesis studies. Several strat- odimeric TALENs. Nucleic Acids Res. doi:10.1093/nar/gks518.
egies have been developed to increase the efficiency of HR in Cermak, T., Doyle, E.L., Christian, M., Wang, L., Zhang, Y., Schmidt, C.,
Baller, J.A., Somia, N.V., Bogdanove, A.J., Voytas, D.F., 2011. Efficient
cultured cells or at organism level. By applying ZFNs to
design and assembly of custom TALEN and other TAL effector-based
induce DSBs, gene targeting through HR has been achieved constructs for DNA targeting. Nucleic Acids Res. 39, e82.
in Drosophila, mouse and rat (Bibikova et al., 2003; Cui Chakrabarti, S., Streisinger, G., Singer, F., Walker, C., 1983. Frequency of
et al., 2011). Similarly, TALENs have been shown to be gamma-ray induced specific locus and recessive lethal mutations in mature
able to induce HR in human iPSC and ES cells (Hockemeyer germ cells of the zebrafish, BRACHYDANIO RERIO. Genetics 103,
109e123.
et al., 2011). Single strand oligonucleotide template has been
Chen, F., Pruett-Miller, S.M., Huang, Y., Gjoka, M., Duda, K., Taunton, J.,
shown to be more efficient than traditional double-stranded Collingwood, T.N., Frodin, M., Davis, G.D., 2011. High-frequency
template as donors (Chen et al., 2011). Blocking DNA genome editing using ssDNA oligonucleotides with zinc-finger nucleases.
ligase IV activity may facilitate HR by inhibiting NHEJ Nat. Methods 8, 753e755.
(Ochiai et al., 2012). Instead of DSB, modified ZFNs con- Christian, M., Cermak, T., Doyle, E.L., Schmidt, C., Zhang, F., Hummel, A.,
Bogdanove, A.J., Voytas, D.F., 2010. Targeting DNA double-strand breaks
taining an inactive Fok I cleavage domain could be used to
with TAL effector nucleases. Genetics 186, 757e761.
create nicks at the target site. These modified ZFNs could Clark, K.J., Balciunas, D., Pogoda, H.M., Ding, Y., Westcot, S.E.,
increase the relative frequency of HR to NHEJ, though the Bedell, V.M., Greenwood, T.M., Urban, M.D., Skuster, K.J.,
overall frequency of HR is lower than traditional ZFN pairs Petzold, A.M., Ni, J., Nielsen, A.L., Patowary, A., Scaria, V.,
(Kim et al., 2012; Ramirez et al., 2012; Wang et al., 2012). Sivasubbu, S., Xu, X., Hammerschmidt, M., Ekker, S.C., 2011. In vivo
protein trapping produces a functional expression codex of the vertebrate
With the help of these experience and TALEN technique, we
proteome. Nat. Methods 8, 506e512.
believe that gene targeting through HR, the last missing Cong, L., Zhou, R., Kuo, Y.C., Cunniff, M., Zhang, F., 2012. Comprehensive
reverse genetics tool in zebrafish, will be ultimately achieved interrogation of natural TALE DNA-binding modules and transcriptional
in the near future. repressor domains. Nat. Commun. 3, 968.
P. Huang et al. / Journal of Genetics and Genomics 39 (2012) 421e433 431

Cui, X., Ji, D., Fisher, D.A., Wu, Y., Briner, D.M., Weinstein, E.J., 2011. Jaenisch, R., 2011. Genetic engineering of human pluripotent cells using
Targeted integration in rat and mouse embryos with zinc-finger nucleases. TALE nucleases. Nat. Biotechnol. 29, 731e734.
Nat. Biotechnol. 29, 64e67. Huang, P., Xiao, A., Zhou, M., Zhu, Z., Lin, S., Zhang, B., 2011. Heritable
Cui, Z., Yang, Y., Kaufman, C.D., Agalliu, D., Hackett, P.B., 2003. RecA- gene targeting in zebrafish using customized TALENs. Nat. Biotechnol.
mediated, targeted mutagenesis in zebrafish. Mar. Biotechnol. (NY) 5, 29, 699e700.
174e184. Hyde, D.R., Godwin, A.R., Thummel, R., 2012. In vivo electroporation of
Davidson, A.E., Balciunas, D., Mohn, D., Shaffer, J., Hermanson, S., morpholinos into the regenerating adult zebrafish tail fin. J. Vis. Exp.,
Sivasubbu, S., Cliff, M.P., Hackett, P.B., Ekker, S.C., 2003. Efficient gene e3632.
delivery and gene expression in zebrafish using the Sleeping Beauty Kawakami, K., Shima, A., Kawakami, N., 2000. Identification of a functional
transposon. Dev. Biol. 263, 191e202. transposase of the Tol2 element, an Ac-like element from the Japanese
de Bruijn, E., Cuppen, E., Feitsma, H., 2009. Highly efficient ENU muta- medaka fish, and its transposition in the zebrafish germs lineage. Proc.
genesis in zebrafish. Methods Mol. Biol. 546, 3e12. Natl. Acad. Sci. USA 97, 11403e11408.
Dong, M., Fu, Y.F., Du, T.T., Jing, C.B., Fu, C.T., Chen, Y., Jin, Y., Deng, M., Kawakami, K., Takeda, H., Kawakami, N., Kobayashi, M., Matsuda, N.,
Liu, T.X., 2009. Heritable and lineage-specific gene knockdown in Mishina, M., 2004. A transposon-mediated gene trap approach identifies
zebrafish embryo. PLoS ONE 4, e6125. developmentally regulated genes in zebrafish. Dev. Cell 7, 133e144.
Dong, Z., Ge, J., Li, K., Xu, Z., Liang, D., Li, J., Jia, W., Li, Y., Dong, X., Kim, E., Kim, S., Kim, D.H., Choi, B.S., Choi, I.Y., Kim, J.S., 2012. Precision
Cao, S., Wang, X., Pan, J., Zhao, Q., 2011. Heritable targeted inactivation genome engineering with programmable DNA-nicking enzymes. Genome
of myostatin gene in yellow catfish (Pelteobagrus fulvidraco) using engi- Res. 22, 1327e1333.
neered zinc finger nucleases. PLoS ONE 6, e28897. Koga, A., Cheah, F.S., Hamaguchi, S., Yeo, G.H., Chong, S.S., 2008. Germline
Doyon, Y., McCammon, J.M., Miller, J.C., Faraji, F., Ngo, C., Katibah, G.E., transgenesis of zebrafish using the medaka Tol1 transposon system. Dev.
Amora, R., Hocking, T.D., Zhang, L., Rebar, E.J., Gregory, P.D., Dyn. 237, 2466e2474.
Urnov, F.D., Amacher, S.L., 2008. Heritable targeted gene disruption in Kondrychyn, I., Garcia-Lecea, M., Emelyanov, A., Parinov, S., Korzh, V.,
zebrafish using designed zinc-finger nucleases. Nat. Biotechnol. 26, 2009. Genome-wide analysis of Tol2 transposon reintegration in zebrafish.
702e708. BMC Genomics 10, 418.
Driever, W., Solnica-Krezel, L., Schier, A.F., Neuhauss, S.C., Malicki, J., Lawson, N.D., Wolfe, S.A., 2011. Forward and reverse genetic approaches for
Stemple, D.L., Stainier, D.Y., Zwartkruis, F., Abdelilah, S., Rangini, Z., the analysis of vertebrate development in the zebrafish. Dev. Cell 21,
Belak, J., Boggs, C., 1996. A genetic screen for mutations affecting 48e64.
embryogenesis in zebrafish. Development 123, 37e46. Lee, H.J., Kim, E., Kim, J.S., 2010. Targeted chromosomal deletions in human
Eisen, J.S., Smith, J.C., 2008. Controlling morpholino experiments: don’t stop cells using zinc finger nucleases. Genome Res. 20, 81e89.
making antisense. Development 135, 1735e1743. Li, L., Piatek, M.J., Atef, A., Piatek, A., Wibowo, A., Fang, X., Sabir, J.S.,
Emelyanov, A., Gao, Y., Naqvi, N.I., Parinov, S., 2006. Trans-kingdom Zhu, J.K., Mahfouz, M.M., 2012a. Rapid and highly efficient construction
transposition of the maize dissociation element. Genetics 174, 1095e1104. of TALE-based transcriptional regulators and nucleases for genome
Foley, J.E., Yeh, J.-R.J., Maeder, M.L., Reyon, D., Sander, J.D., Peterson, R.T., modification. Plant Mol. Biol. 78, 407e416.
Joung, J.K., 2009. Rapid mutation of endogenous zebrafish genes using Li, T., Huang, S., Jiang, W.Z., Wright, D., Spalding, M.H., Weeks, D.P.,
zinc finger nucleases made by Oligomerized Pool ENgineering (OPEN). Yang, B., 2011a. TAL nucleases (TALNs): hybrid proteins composed of
PLoS ONE 4, e4348. TAL effectors and Fok I DNA-cleavage domain. Nucleic Acids Res. 39,
Geibetaler, R., Scholze, H., Hahn, S., Streubel, J., Bonas, U., Behrens, S.E., 359e372.
Boch, J., 2011. Transcriptional activators of human genes with program- Li, T., Huang, S., Zhao, X., Wright, D.A., Carpenter, S., Spalding, M.H.,
mable DNA-specificity. PLoS ONE 6, e19509. Weeks, D.P., Yang, B., 2011b. Modularly assembled designer TAL effector
Geurts, A.M., Cost, G.J., Freyvert, Y., Zeitler, B., Miller, J.C., Choi, V.M., nucleases for targeted gene knockout and gene replacement in eukaryotes.
Jenkins, S.S., Wood, A., Cui, X., Meng, X., Vincent, A., Lam, S., Nucleic Acids Res. 39, 6315e6325.
Michalkiewicz, M., Schilling, R., Foeckler, J., Kalloway, S., Weiler, H., Li, T., Liu, B., Spalding, M.H., Weeks, D.P., Yang, B., 2012b. High-efficiency
Menoret, S., Anegon, I., Davis, G.D., Zhang, L., Rebar, E.J., Gregory, P.D., TALEN-based gene editing produces disease-resistant rice. Nat. Bio-
Urnov, F.D., Jacob, H.J., Buelow, R., 2009. Knockout rats via embryo technol. 30, 390e392.
microinjection of zinc-finger nucleases. Science 325, 433. Lieschke, G.J., Currie, P.D., 2007. Animal models of human disease: zebrafish
Golling, G., Amsterdam, A., Sun, Z., Antonelli, M., Maldonado, E., Chen, W., swim into view. Nat. Rev. Genet. 8, 353e367.
Burgess, S., Haldi, M., Artzt, K., Farrington, S., Lin, S.Y., Nissen, R.M., Lin, S., Gaiano, N., Culp, P., Burns, J.C., Friedmann, T., Yee, J.K.,
Hopkins, N., 2002. Insertional mutagenesis in zebrafish rapidly identifies Hopkins, N., 1994. Integration and germ-line transmission of a pseudo-
genes essential for early vertebrate development. Nat. Genet. 31, 135e140. typed retroviral vector in zebrafish. Science 265, 666e669.
Gupta, A., Christensen, R.G., Rayla, A.L., Lakshmanan, A., Stormo, G.D., Liu, J., Li, C., Yu, Z., Huang, P., Wu, H., Wei, C., Zhu, N., Shen, Y., Chen, Y.,
Wolfe, S.A., 2012. An optimized two-finger archive for ZFN-mediated Zhang, B., Deng, W.M., Jiao, R., 2012. Efficient and specific modifications
gene targeting. Nat. Methods 9, 588e590. of the Drosophila genome by means of an easy TALEN strategy. J. Genet.
Gupta, A., Meng, X., Zhu, L.J., Lawson, N.D., Wolfe, S.A., 2011. Zinc finger Genomics 39, 209e215.
protein-dependent and -independent contributions to the in vivo off-target Lombardo, A., Genovese, P., Beausejour, C.M., Colleoni, S., Lee, Y.L.,
activity of zinc finger nucleases. Nucleic Acids Res. 39, 381e392. Kim, K.A., Ando, D., Urnov, F.D., Galli, C., Gregory, P.D., Holmes, M.C.,
Haffter, P., Granato, M., Brand, M., Mullins, M.C., Hammerschmidt, M., Naldini, L., 2007. Gene editing in human stem cells using zinc finger
Kane, D.A., Odenthal, J., van Eeden, F.J., Jiang, Y.J., Heisenberg, C.P., nucleases and integrase-defective lentiviral vector delivery. Nat. Bio-
Kelsh, R.N., Furutani-Seiki, M., Vogelsang, E., Beuchle, D., Schach, U., technol. 25, 1298e1306.
Fabian, C., Nüsslein-Volhard, C., 1996. The identification of genes with Maddison, L.A., Lu, J., Chen, W., 2011. Generating conditional mutations in
unique and essential functions in the development of the zebrafish, Danio zebrafish using gene-trap mutagenesis. Methods Cell Biol. 104, 1e22.
rerio. Development 123, 1e36. Maeder, M.L., Thibodeau-Beganny, S., Osiak, A., Wright, D.A.,
Hagmann, M., Bruggmann, R., Xue, L., Georgiev, O., Schaffner, W., Anthony, R.M., Eichtinger, M., Jiang, T., Foley, J.E., Winfrey, R.J.,
Rungger, D., Spaniol, P., Gerster, T., 1998. Homologous recombination Townsend, J.A., Unger-Wallace, E., Sander, J.D., Muller-Lerch, F., Fu, F.,
and DNA-end joining reactions in zygotes and early embryos of zebrafish Pearlberg, J., Gobel, C., Dassie, J.P., Pruett-Miller, S.M., Porteus, M.H.,
(Danio rerio) and Drosophila melanogaster. Biol. Chem. 379, 673e681. Sgroi, D.C., Iafrate, A.J., Dobbs, D., McCray Jr., P.B., Cathomen, T.,
Hockemeyer, D., Wang, H., Kiani, S., Lai, C.S., Gao, Q., Cassady, J.P., Voytas, D.F., Joung, J.K., 2008. Rapid “open-source” engineering of
Cost, G.J., Zhang, L., Santiago, Y., Miller, J.C., Zeitler, B., Cherone, J.M., customized zinc-finger nucleases for highly efficient gene modification.
Meng, X., Hinkley, S.J., Rebar, E.J., Gregory, P.D., Urnov, F.D., Mol. Cell 31, 294e301.
432 P. Huang et al. / Journal of Genetics and Genomics 39 (2012) 421e433

Mahfouz, M.M., Li, L., Shamimuzzaman, M., Wibowo, A., Fang, X., Shestopalov, I.A., Pitt, C.L., Chen, J.K., 2012. Spatiotemporal resolution of the
Zhu, J.K., 2011. De novo-engineered transcription activator-like effector Ntla transcriptome in axial mesoderm development. Nat. Chem. Biol. 8,
(TALE) hybrid nuclease with novel DNA binding specificity creates 270e276.
double-strand breaks. Proc. Natl. Acad. Sci. USA 108, 2623e2628. Shukla, V.K., Doyon, Y., Miller, J.C., DeKelver, R.C., Moehle, E.A.,
Meng, X., Noyes, M.B., Zhu, L.J., Lawson, N.D., Wolfe, S.A., 2008. Targeted Worden, S.E., Mitchell, J.C., Arnold, N.L., Gopalan, S., Meng, X.,
gene inactivation in zebrafish using engineered zinc-finger nucleases. Nat. Choi, V.M., Rock, J.M., Wu, Y.Y., Katibah, G.E., Zhifang, G.,
Biotechnol. 26, 695e701. McCaskill, D., Simpson, M.A., Blakeslee, B., Greenwalt, S.A.,
Miller, J.C., Tan, S., Qiao, G., Barlow, K.A., Wang, J., Xia, D.F., Meng, X., Butler, H.J., Hinkley, S.J., Zhang, L., Rebar, E.J., Gregory, P.D.,
Paschon, D.E., Leung, E., Hinkley, S.J., Dulay, G.P., Hua, K.L., Urnov, F.D., 2009. Precise genome modification in the crop species Zea
Ankoudinova, I., Cost, G.J., Urnov, F.D., Zhang, H.S., Holmes, M.C., mays using zinc-finger nucleases. Nature 459, 437e441.
Zhang, L., Gregory, P.D., Rebar, E.J., 2011. A TALE nuclease architecture Siekmann, A.F., Standley, C., Fogarty, K.E., Wolfe, S.A., Lawson, N.D., 2009.
for efficient genome editing. Nat. Biotechnol. 29, 143e148. Chemokine signaling guides regional patterning of the first embryonic
Moens, C.B., Donn, T.M., Wolf-Saxon, E.R., Ma, T.P., 2008. Reverse genetics artery. Genes Dev. 23, 2272e2277.
in zebrafish by TILLING. Brief Funct. Genomic. Proteomic. 7, 454e459. Solnica-Krezel, L., Schier, A.F., Driever, W., 1994. Efficient recovery of ENU-
Moore, F.E., Reyon, D., Sander, J.D., Martinez, S.A., Blackburn, J.S., induced mutations from the zebrafish germline. Genetics 136, 1401e1420.
Khayter, C., Ramirez, C.L., Joung, J.K., Langenau, D.M., 2012. Improved Streubel, J., Blucher, C., Landgraf, A., Boch, J., 2012. TAL effector RVD
somatic mutagenesis in zebrafish using transcription activator-like effector specificities and efficiencies. Nat. Biotechnol. 30, 593e595.
nucleases (TALENs). PLoS ONE 7, e37877. Su, J., Zhu, Z., Wang, Y., Xiong, F., Zou, J., 2008a. The cytomegalovirus
Morbitzer, R., Elsaesser, J., Hausner, J., Lahaye, T., 2011. Assembly of custom promoter-driven short hairpin RNA constructs mediate effective RNA
TALE-type DNA binding domains by modular cloning. Nucleic Acids Res. interference in zebrafish in vivo. Mar. Biotechnol. (NY) 10, 262e269.
39, 5790e5799. Su, J., Zhu, Z., Xiong, F., Wang, Y., 2008b. Hybrid cytomegalovirus-U6
Morbitzer, R., Romer, P., Boch, J., Lahaye, T., 2010. Regulation of selected promoter-based plasmid vectors improve efficiency of RNA interference
genome loci using de novo-engineered transcription activator-like effector in zebrafish. Mar. Biotechnol. (NY) 10, 511e517.
(TALE)-type transcription factors. Proc. Natl. Acad. Sci. USA 107, Summerton, J., 1999. Morpholino antisense oligomers: the case for an RNase
21617e21622. H-independent structural type. Biochim. Biophys. Acta 1489, 141e158.
Moscou, M.J., Bogdanove, A.J., 2009. A simple cipher governs DNA recog- Summerton, J., Weller, D., 1997. Morpholino antisense oligomers: design,
nition by TAL effectors. Science 326, 1501. preparation, and properties. Antisense Nucleic Acid Drug Dev. 7, 187e195.
Mullins, M.C., Hammerschmidt, M., Haffter, P., Nüsslein-Volhard, C., 1994. Tallafuss, A., Gibson, D., Morcos, P., Li, Y., Seredick, S., Eisen, J.,
Large-scale mutagenesis in the zebrafish: in search of genes controlling Washbourne, P., 2012. Turning gene function ON and OFF using sense and
development in a vertebrate. Curr. Biol. 4, 189e202. antisense photo-morpholinos in zebrafish. Development 139, 1691e1699.
Nasevicius, A., Ekker, S.C., 2000. Effective targeted gene ‘knockdown’ in Tesson, L., Usal, C., Menoret, S., Leung, E., Niles, B.J., Remy, S.,
zebrafish. Nat. Genet. 26, 216e220. Santiago, Y., Vincent, A.I., Meng, X., Zhang, L., Gregory, P.D., Anegon, I.,
Ochiai, H., Sakamoto, N., Fujita, K., Nishikawa, M., Suzuki, K., Matsuura, S., Cost, G.J., 2011. Knockout rats generated by embryo microinjection of
Miyamoto, T., Sakuma, T., Shibata, T., Yamamoto, T., 2012. Zinc-finger TALENs. Nat. Biotechnol. 29, 695e696.
nuclease-mediated targeted insertion of reporter genes for quantitative Thummel, R., Bai, S., Sarras Jr., M.P., Song, P., McDermott, J., Brewer, J.,
imaging of gene expression in sea urchin embryos. Proc. Natl. Acad. Sci. Perry, M., Zhang, X., Hyde, D.R., Godwin, A.R., 2006. Inhibition of
USA 109, 10915e10920. zebrafish fin regeneration using in vivo electroporation of morpholinos
Parinov, S., Kondrichin, I., Korzh, V., Emelyanov, A., 2004. Tol2 transposon- against fgfr1 and msxb. Dev. Dyn. 235, 336e346.
mediated enhancer trap to identify developmentally regulated zebrafish Thummel, R., Bailey, T.J., Hyde, D.R., 2011. In vivo electroporation of
genes in vivo. Dev. Dyn. 231, 449e459. morpholinos into the adult zebrafish retina. J. Vis. Exp., e3603.
Patton, E.E., Zon, L.I., 2001. The art and design of genetic screens: zebrafish. Tian, T., Zhao, L., Zhao, X.Y., Zhang, M., Meng, A.M., 2009. A zebrafish gene
Nat. Rev. Genet. 2, 956e966. trap line expresses GFP recapturing expression pattern of foxj1b. J. Genet.
Porteus, M.H., Baltimore, D., 2003. Chimeric nucleases stimulate gene tar- Genomics 36, 581e589.
geting in human cells. Science 300, 763. Tong, C., Huang, G., Ashton, C., Wu, H., Yan, H., Ying, Q.L., 2012. Rapid and
Ramirez, C.L., Certo, M.T., Mussolino, C., Goodwin, M.J., Cradick, T.J., cost-effective gene targeting in rat embryonic stem cells by TALENs. J.
McCaffrey, A.P., Cathomen, T., Scharenberg, A.M., Joung, J.K., 2012. Genet. Genomics 39, 275e280.
Engineered zinc finger nickases induce homology-directed repair with Townsend, J.A., Wright, D.A., Winfrey, R.J., Fu, F., Maeder, M.L.,
reduced mutagenic effects. Nucleic Acids Res. 40, 5560e5568. Joung, J.K., Voytas, D.F., 2009. High-frequency modification of plant
Ramirez, C.L., Foley, J.E., Wright, D.A., Muller-Lerch, F., Rahman, S.H., genes using engineered zinc-finger nucleases. Nature 459, 442e445.
Cornu, T.I., Winfrey, R.J., Sander, J.D., Fu, F., Townsend, J.A., Trinh, L.A., Hochgreb, T., Graham, M., Wu, D., Ruf-Zamojski, F.,
Cathomen, T., Voytas, D.F., Joung, J.K., 2008. Unexpected failure rates for Jayasena, C.S., Saxena, A., Hawk, R., Gonzalez-Serricchio, A.,
modular assembly of engineered zinc fingers. Nat. Methods 5, 374e375. Dixson, A., Chow, E., Gonzales, C., Leung, H.Y., Solomon, I., Bronner-
Reyon, D., Tsai, S.Q., Khayter, C., Foden, J.A., Sander, J.D., Joung, J.K., Fraser, M., Megason, S.G., Fraser, S.E., 2011. A versatile gene trap to
2012. FLASH assembly of TALENs for high-throughput genome editing. visualize and interrogate the function of the vertebrate proteome. Genes
Nat. Biotechnol. 30, 460e465. Dev. 25, 2306e2320.
Robu, M.E., Larson, J.D., Nasevicius, A., Beiraghi, S., Brenner, C., Urnov, F.D., Rebar, E.J., Holmes, M.C., Zhang, H.S., Gregory, P.D., 2010.
Farber, S.A., Ekker, S.C., 2007. p53 activation by knockdown technolo- Genome editing with engineered zinc finger nucleases. Nat. Rev. Genet.
gies. PLoS Genet. 3, e78. 11, 636e646.
Sander, J.D., Cade, L., Khayter, C., Reyon, D., Peterson, R.T., Joung, J.K., Walker, C., Streisinger, G., 1983. Induction of mutations by gamma-rays in
Yeh, J.R., 2011a. Targeted gene disruption in somatic zebrafish cells using pregonial germ cells of zebrafish embryos. Genetics 103, 125e136.
engineered TALENs. Nat. Biotechnol. 29, 697e698. Wang, D., Jao, L.-E., Zheng, N., Dolan, K., Ivey, J., Zonies, S., Wu, X.,
Sander, J.D., Dahlborg, E.J., Goodwin, M.J., Cade, L., Zhang, F., Wu, K., Yang, H., Meng, Q., Zhu, Z., Zhang, B., Lin, S., Burgess, S.M.,
Cifuentes, D., Curtin, S.J., Blackburn, J.S., Thibodeau-Beganny, S., Qi, Y., 2007. Efficient genome-wide mutagenesis of zebrafish genes by retroviral
Pierick, C.J., Hoffman, E., Maeder, M.L., Khayter, C., Reyon, D., insertions. Proc. Natl. Acad. Sci. USA 104, 12428e12433.
Dobbs, D., Langenau, D.M., Stupar, R.M., Giraldez, A.J., Voytas, D.F., Wang, J., Friedman, G., Doyon, Y., Wang, N.S., Li, C.J., Miller, J.C.,
Peterson, R.T., Yeh, J.R., Joung, J.K., 2011b. Selection-free zinc-finger- Hua, K.L., Yan, J.J., Babiarz, J.E., Gregory, P.D., Holmes, M.C., 2012.
nuclease engineering by context-dependent assembly (CoDA). Nat. Targeted gene addition to a predetermined site in the human genome using
Methods 8, 67e69. a ZFN-based nicking enzyme. Genome Res. 22, 1316e1326.
P. Huang et al. / Journal of Genetics and Genomics 39 (2012) 421e433 433

Wienholds, E., Schulte-Merker, S., Walderich, B., Plasterk, R.H., 2002. Yang, D., Yang, H., Li, W., Zhao, B., Ouyang, Z., Liu, Z., Zhao, Y., Fan, N.,
Target-selected inactivation of the zebrafish rag1 gene. Science 297, Song, J., Tian, J., Li, F., Zhang, J., Chang, L., Pei, D., Chen, Y.E., Lai, L.,
99e102. 2011. Generation of PPARgamma mono-allelic knockout pigs via zinc-
Wood, A.J., Lo, T.W., Zeitler, B., Pickle, C.S., Ralston, E.J., Lee, A.H., finger nucleases and nuclear transfer cloning. Cell Res. 21, 979e982.
Amora, R., Miller, J.C., Leung, E., Meng, X., Zhang, L., Rebar, E.J., Yu, S., Luo, J., Song, Z., Ding, F., Dai, Y., Li, N., 2011. Highly efficient
Gregory, P.D., Urnov, F.D., Meyer, B.J., 2011. Targeted genome editing modification of beta-lactoglobulin (BLG) gene via zinc-finger nucleases in
across species using ZFNs and TALENs. Science 333, 307. cattle. Cell Res. 21, 1638e1640.
Wu, X., Li, Y., Crise, B., Burgess, S.M., 2003. Transcription start regions in Zhao, L., Zhao, X., Tian, T., Lu, Q., Skrbo-Larssen, N., Wu, D., Kuang, Z.,
the human genome are favored targets for MLV integration. Science 300, Zheng, X., Han, Y., Yang, S., Zhang, C., Meng, A., 2008. Heart-specific
1749e1751. isoform of tropomyosin4 is essential for heartbeat in zebrafish embryos.
Wu, Y., Zhang, G., Xiong, Q., Luo, F., Cui, C., Hu, W., Yu, Y., Su, J., Xu, A., Cardiovasc. Res. 80, 200e208.
Zhu, Z., 2006. Integration of double-fluorescence expression vectors into Zhao, Z., Cao, Y., Li, M., Meng, A., 2001. Double-stranded RNA injection
zebrafish genome for the selection of site-directed knockout/knockin. Mar. produces nonspecific defects in zebrafish. Dev. Biol. 229, 215e223.
Biotechnol. (NY) 8, 304e311. Zhu, C., Smith, T., McNulty, J., Rayla, A.L., Lakshmanan, A., Siekmann, A.F.,
Xue, Y.L., Xiao, A., Wen, L., Jia, Y., Gao, Y., Zhu, Z.Y., Lin, S., Zhang, B., Buffardi, M., Meng, X., Shin, J., Padmanabhan, A., Cifuentes, D.,
2010. Generation and characterization of blood vessel specific EGFP Giraldez, A.J., Look, A.T., Epstein, J.A., Lawson, N.D., Wolfe, S.A., 2011.
transgenic zebrafish via To12 transposon mediated enhancer trap screen. Evaluation and application of modularly assembled zinc-finger nucleases
Prog. Biochem. Biophys. 37, 720e727. in zebrafish. Development 138, 4555e4564.

You might also like