Genome Editing
Genome Editing
Genome Editing
org/ on November 15, 2020 - Published by Cold Spring Harbor Laboratory Press
Targeted nucleases have provided researchers with the ability to manipulate virtually any
genomic sequence, enabling the facile creation of isogenic cell lines and animal models for
the study of human disease, and promoting exciting new possibilities for human gene
therapy. Here we review three foundational technologies—clustered regularly interspaced
short palindromic repeats (CRISPR)-CRISPR-associated protein 9 (Cas9), transcription acti-
vator-like effector nucleases (TALENs), and zinc-finger nucleases (ZFNs). We discuss the
engineering advances that facilitated their development and highlight several achievements
in genome engineering that were made possible by these tools. We also consider artificial
transcription factors, illustrating how this technology can complement targeted nucleases for
synthetic biology and gene therapy.
n recent years, the emergence of highly versa- nize new genomic sequences has driven a revo-
I tile genome-editing technologies has pro-
vided investigators with the ability to rapidly
lution in genome editing that has accelerated
scientific breakthroughs and discoveries in dis-
and economically introduce sequence-specific ciplines as diverse as synthetic biology, human
modifications into the genomes of a broad spec- gene therapy, disease modeling, drug discovery,
trum of cell types and organisms. The core tech- neuroscience, and the agricultural sciences.
nologies now most commonly used to facilitate The diverse array of genetic outcomes made
genome editing, shown in Figure 1, are (1) clus- possible by these technologies is the result, in
tered regularly interspaced short palindromic large part, of their ability to efficiently induce
repeats (CRISPR)-CRISPR-associated protein targeted DNA double-strand breaks (DSBs).
9 (Cas9), (2) transcription activator-like effec- These DNA breaks then drive activation of cel-
tor nucleases (TALENs), (3) zinc-finger nucle- lular DNA repair pathways and facilitate the in-
ases (ZFNs), and (4) homing endonucleases or troduction of site-specific genomic modifica-
meganucleases. tions (Rouet et al. 1994; Choulika et al. 1995).
In particular, the ease with which CRISPR- This process is most often used to achieve gene
Cas9 and TALENs can be configured to recog- knockout via random base insertions and/or
4
These authors contributed equally to this work.
Editors: Daniel G. Gibson, Clyde A. Hutchison III, Hamilton O. Smith, and J. Craig Venter
Additional Perspectives on Synthetic Biology available at www.cshperspectives.org
Copyright # 2016 Cold Spring Harbor Laboratory Press; all rights reserved; doi: 10.1101/cshperspect.a023754
Cite this article as Cold Spring Harb Perspect Biol 2016;8:a023754
1
Downloaded from http://cshperspectives.cshlp.org/ on November 15, 2020 - Published by Cold Spring Harbor Laboratory Press
T. Gaj et al.
5′- -3′
Homing endonuclease
3′- -5′
Zinc-finger
DNA-binding domain 5′-ANN-3′
1F 2F 3F 4F 5′-GNN-3′
Fokl
5′- -3′
ZFN
3′- Fokl -5′
4F 3F 2F 1F
5′-GNN-GNN-3′
TAL effector
Repeat variable DNA-binding domain TALEN
diresidues (RVDs)
NG: T Fokl
NI: A 5′- -3′
HD: C 3′- Fokl -5′
NH: G Amino-terminal
domain
Carboxy-terminal
domain
Cas9
gRNA
-5′
PAM
5′- -3′
3′- -5′
Target
Figure 1. Genome-editing technologies. Cartoons illustrating the mechanisms of targeted nucleases. From top to
bottom: homing endonucleases, zinc-finger nucleases (ZFNs), transcription activator-like effector (TALE) nu-
cleases (TALENs), and clustered regularly interspaced short palindromic repeats (CRISPR)-CRISPR-associated
protein 9 (Cas9). Homing endonucleases generally cleave their DNA substrates as dimers, and do not have
distinct binding and cleavage domains. ZFNs recognize target sites that consist of two zinc-finger binding sites
that flank a 5- to 7-base pair (bp) spacer sequence recognized by the FokI cleavage domain. TALENs recognize
target sites that consist of two TALE DNA-binding sites that flank a 12- to 20-bp spacer sequence recognized by
the FokI cleavage domain. The Cas9 nuclease is targeted to DNA sequences complementary to the targeting
sequence within the single guide RNA (gRNA) located immediately upstream of a compatible protospacer
adjacent motif (PAM). DNA and protein are not drawn to scale.
deletions that can be introduced by nonhomol- comes) (Bibikova et al. 2001, 2003; Porteus and
ogous end joining (NHEJ) (Fig. 2A) (Bibikova Baltimore 2003; Urnov et al. 2005). Indeed, the
et al. 2002). Alternatively, in the presence of a broad versatility of these genome-modifying
donor template with homology to the targeted enzymes is evidenced by the fact that they also
chromosomal site, gene integration, or base serve as the foundation for artificial transcrip-
correction via homology-directed repair (HDR) tion factors, a class of tools capable of modulat-
can occur (HDR) (Fig. 2B) (see Fig. 2 for an ing the expression of nearly any gene within a
overview of other possible genome-editing out- genome.
Genome-Editing Technologies
Genomic
target
Figure 2. Genome-editing outcomes. Targeted nucleases induce DNA double-strand breaks (DSBs) that are
repaired by nonhomologous end joining (NHEJ) or, in the presence of donor template, homology-directed
repair (HDR). (A) In the absence of a donor template, NHEJ introduces small base insertions or deletions that
can result in gene disruption. When two DSBs are induced simultaneously, the intervening genomic sequence
can be deleted or inverted. (B) In the presence of donor DNA ( plasmid or single-stranded oligonucleotide),
recombination between homologous DNA sequences present on the donor template and a specific chromo-
somal site can facilitate targeted integration. Lightning bolts indicate DSBs.
Here we review key principles of genome and Carroll 2005; Urnov et al. 2010). ZFNs
editing, emphasizing many of the engineering function as dimers, with each monomer recog-
advances that have laid the groundwork for the nizing a specific “half site” sequence—typically
creation, refinement, and implementation of nine to 18 base pairs (bps) of DNA—via the
the current suite of genome-modifying tools. zinc-finger DNA-binding domain (Fig. 1). Di-
We also provide an overview of the achieve- merization of the ZFN proteins is mediated by
ments made possible by genome editing, illus- the FokI cleavage domain, which cuts DNA
trating how this technology can enable advances within a five- to seven-bp spacer sequence that
throughout the life sciences. separates two flanking zinc-finger binding sites
(Smith et al. 2000). Each ZFN is typically com-
posed of three or four zinc-finger domains, with
TARGETED NUCLEASES each individual domain composed of 30 ami-
no acid residues that are organized in a bba
Zinc-Finger Nucleases
motif (Pavletich and Pabo 1991). The residues
ZFNs, which are fusions between a custom- that facilitate DNA recognition are located
designed Cys2-His2 zinc-finger protein and the within the a-helical domain and typically inter-
cleavage domain of the FokI restriction endo- act with three bps of DNA, with occasional over-
nuclease (Kim et al. 1996), were the first target- lap from an adjacent domain (Wolfe et al. 2000).
ed nuclease to achieve widespread use (Porteus Using methods such as phage display (Choo
T. Gaj et al.
and Klug 1994; Jamieson et al. 1994; Wu et al. to when expressed within cells from nucleic ac-
1995), a large number of zinc-finger domains ids (Gaj et al. 2012). Modified ZFN proteins
recognizing distinct DNA triplets have been endowed with improved cell-penetrating activ-
identified (Segal et al. 1999; Dreier et al. 2001, ity have since been described (Liu et al. 2015a).
2005; Bae et al. 2003). These domains can be ZFNickases can also facilitate gene correction in
fused together in tandem using a canonical the absence of a DSB (Kim et al. 2012; Ramirez
linker peptide (Liu et al. 1997) to generate poly- et al. 2012; Wang et al. 2012). These enzymes,
dactyl zinc-finger proteins that can target a wide which consist of one catalytically inactivated
range of possible DNA sequences (Beerli et al. ZFN monomer in combination with a second
1998, 2000a; Kim et al. 2009). In addition to this native ZFN monomer, can stimulate HDR by
“modular assembly” approach to zinc-finger nicking or cleaving one strand of DNA and are
construction, selection-based methods for con- derived from a concept first illustrated by Stod-
structing zinc-finger proteins have also been re- dard and colleagues using homing endonucle-
ported (Greisman and Pabo 1997; Isalan et al. ases (McConnell Smith et al. 2009).
2001; Hurt et al. 2003; Magnenat et al. 2004), Unlike TALENs and CRISPR-Cas9, the dif-
including those that consider context-depen- ficulty associated with constructing zinc-finger
dent interactions between adjacent zinc-finger arrays has hindered their widespread adoption
domains, such as oligomerized pool engineer- in unspecialized laboratories. In particular, it
ing (OPEN) (Maeder et al. 2008). In addition, remains challenging to create zinc-finger do-
specialized sets of validated two-finger, zinc-fin- mains that can effectively recognize all DNA
ger modules have been used to assemble zinc- triplets, especially those of the 50 -CNN-30 and
finger arrays (Kim et al. 2009; Bhakta et al. 50 -TNN-30 variety. As a result, ZFNs lack the
2013), including those that take context-depen- target flexibility inherent to more recent ge-
dent effects into account (Sander et al. 2011b; nome-editing platforms. Nevertheless, the po-
Gupta et al. 2012). tential for ZFNs to mediate specific and efficient
One major concern associated with the use genome editing is evidenced by ongoing clinical
of ZFNs for genome editing (in addition to all trials based on ZFN-mediated knockout of the
targeted nucleases) is off-target mutations (Ga- human immunodeficiency virus (HIV)-1 co-
briel et al. 2011; Pattanayak et al. 2011). As a receptor CCR5 for treatment of HIV/acquired
result, several approaches have been undertaken immune deficiency syndrome (AIDS) (Tebas
to enhance their specificity. Among the most et al. 2014) and a planned clinical trial based
successful of these has been the creation of on site-specific integration of the factor IX
obligate heterodimeric ZFN architectures that gene into the albumin locus to treat hemophilia
rely on charge – charge repulsion to prevent un- B (Clinical Trial ID: NCT02695160) (Sharma
wanted homodimerization of the FokI cleavage et al. 2015).
domain (Miller et al. 2007; Doyon et al. 2011),
thereby minimizing the potential for ZFNs to
TALE Nucleases
dimerize at off-target sites. Additionally, pro-
tein-engineering methods have been used to TALE proteins are bacterial effectors. In 2009,
enhance the cleavage efficiency of the FokI the code used by TALE proteins to recognize
cleavage domain (Guo et al. 2010). One par- DNA was uncovered (Boch et al. 2009; Moscou
ticularly promising approach for improving and Bogdanove 2009). In a matter of months,
ZFN specificity is to deliver them into cells as this discovery enabled the creation of custom
protein. Because of the intrinsic cell-penetrat- TALENs capable of modifying nearly any gene.
ing activity of zinc-finger domains (Gaj et al. Like ZFNs, TALENs are modular in form and
2014a), ZFN proteins themselves are inherently function, comprised of an amino-terminal
cell-permeable and can facilitate gene editing TALE DNA-binding domain fused to a car-
with fewer off-target effects when applied di- boxy-terminal FokI cleavage domain (Christian
rectly onto cells as purified protein compared et al. 2010; Miller et al. 2011). Also like ZFNs,
Genome-Editing Technologies
dimerization of TALEN proteins is mediated by amount of time and experience needed to as-
the FokI cleavage domain, which cuts within a semble a functional nuclease. Second, TALENs
12- to 19-bp spacer sequence that separates each have been reported to show improved specificity
TALE binding site (Fig. 1) (Miller et al. 2011). and reduced toxicity compared to some ZFNs
TALEs are typically assembled to recognize (Mussolino et al. 2014), potentially because of
between 12- to 20-bps of DNA, with more bases their increased affinity for target DNA (Meckler
typically leading to higher genome-editing et al. 2013) or perhaps a greater energetic pen-
specificity (Guilinger et al. 2014a). The TALE- alty for associating with base mismatches.
binding domain consists of a series of repeat However, TALENs are substantially larger than
domains, each 34 residues in length. Each re- ZFNs, and have a highly repetitive structure,
peat contacts DNA via the amino acid residues making their efficient delivery into cells through
at positions 12 and 13, known as the repeat the use of lentivirus (Holkers et al. 2013) or a
variable diresidues (RVDs) (Boch et al. 2009; single adeno-associated virus (AAV) particle
Moscou and Bogdanove 2009). Unlike zinc fin- challenging. Methods for overcoming these lim-
gers, which recognize DNA triplets, each TALE itations have emerged as TALENs can be readily
repeat recognizes only a single bp, with little to delivered into cells as mRNA (Mahiny et al.
no target site overlap from adjacent domains 2015; Mock et al. 2015) and even protein (Cai
(Deng et al. 2012; Mak et al. 2012). The most et al. 2014; Liu et al. 2014a), although alterna-
commonly used RVDs for assembling synthetic tive codon usage and amino acid degeneracy
TALE arrays are: NI for adenine, HD for cyto- can also be leveraged to express RVD arrays
sine, NG for thymine, and NN or HN for gua- that might be less susceptible to recombina-
nine or adenine (Boch et al. 2009; Moscou and tion (Kim et al. 2013a). In addition, adenoviral
Bogdanove 2009; Cong et al. 2012; Streubel vectors have also proven particularly useful for
et al. 2012). TALE DNA-binding domains can mediating TALEN delivery to hard-to-transfect
be constructed using a variety of methods, cell types (Holkers et al. 2014; Maggio et al.
with the most straightforward approach being 2016).
Golden Gate assembly (Cermak et al. 2011).
However, high-throughput TALE assembly
CRISPR-Cas9
methods have also been developed, including
FLASH assembly (Reyon et al. 2012), iterative The CRISPR-Cas9 system, which has a role in
capped assembly (Briggs et al. 2012), and liga- adaptive immunity in bacteria (Horvath and
tion independent cloning (Schmid-Burgk et al. Barrangou 2010; Marraffini and Sontheimer
2013), among others. More recent advances 2010), is the most recent addition to the ge-
in TALEN assembly, though, have focused on nome-editing toolbox. In bacteria, the type-II
the development of methods that can enhance CRISPR system provides protection against
their performance, including specificity profil- DNA from invading viruses and plasmids via
ing to uncover nonconventional RVDs that RNA-guided DNA cleavage by Cas proteins
improve TALEN activity (Guilinger et al. 2014a; (Wiedenheft et al. 2012; Sorek et al. 2013). Short
Yang et al. 2014; Juillerat et al. 2015; Miller segments of foreign DNA are integrated within
et al. 2015), directed evolution as means to re- the CRISPR locus and transcribed into CRISPR
fine TALE specificity (Hubbard et al. 2015), and RNA (crRNA), which then anneal to trans-ac-
even fusing TALE domains to homing endonu- tivating crRNA (tracrRNA) to direct sequence-
clease variants to generate chimeric nucleases specific degradation of pathogenic DNA by the
with extended targeting specificity (discussed Cas9 protein (Jinek et al. 2012). In 2012, Char-
in more detail below) (Boissel et al. 2014). pentier, Doudna, and co-workers reported that
Compared to ZFNs, TALENs offer two dis- target recognition by the Cas9 protein only re-
tinct advantages for genome editing. First, no quires a seed sequence within the crRNA and a
selection or directed evolution is necessary to conserved protospacer-adjacent motif (PAM)
engineer TALE arrays, dramatically reducing the upstream of the crRNA binding site (Jinek
T. Gaj et al.
et al. 2012). This system has since been simpli- cificity of this system, including using paired
fied for genome engineering (Cho et al. 2013; Cas9 nickases (Mali et al. 2013a; Ran et al.
Cong et al. 2013; Jinek et al. 2013; Mali et al. 2013), which increase gene-editing specificity
2013b) and now consists of only the Cas9 nu- by requiring the induction of two sequential
clease and a single guide RNA (gRNA) contain- and adjacent nicking events for DSB formation,
ing the essential crRNA and tracrRNA elements or truncated gRNA that are more sensitive to
(Fig. 1). Because target site recognition is me- mismatches at the genomic target site than a
diated entirely by the gRNA, CRISPR-Cas9 has full-length gRNA (Fu et al. 2014). Off-target
emerged as the most flexible and user-friendly cleavage has also been reduced by controlling
platform for genome editing, eliminating the the dosage of either the Cas9 protein or gRNA
need for engineering new proteins to recognize within the cell (Hsu et al. 2013), or even by
each new target site. The only major restriction using Cas9 variants configured to enable con-
for Cas9 target site recognition is that the PAM ditional genome editing, such as a rapamycin-
motif—which is recognized by the Cas9 nucle- inducible split-Cas9 architecture (Zetsche et al.
ase and is essential for DNA cleavage—be locat- 2015b) or a Cas9 variant that contains a strate-
ed immediately downstream of the gRNA target gically placed small-molecule-responsive intein
site. The PAM sequence for the Streptococcus domain (Davis et al. 2015). Nucleofection (Kim
pyogenes Cas9, for example, is 50 -NGG-30 (al- et al. 2014) or transient transfection (Zuris et al.
though in some cases 50 -NAG-30 can be tolerat- 2015) of a preformed Cas9 ribonucleoprotein
ed) (Hsu et al. 2013; Jiang et al. 2013; Mali et al. complex has also been shown to reduce off-
2013a). Several studies have now shed light on target effects, enabling DNA-free gene editing
the structural basis of DNA recognition by Cas9, in primary human T cells (Schumann et al.
revealing that the heteroduplex formed by the 2015), embryonic stem cells (Liu et al. 2015b),
gRNA and its complementary strand of DNA is Caenorhabditis elegans gonads (Paix et al. 2015),
housed in a positively charged groove between mouse (Menoret et al. 2015; Wang et al. 2015a)
the two nuclease domains (RuvC and HNH) and zebrafish embryos (Sung et al. 2014), and
within the Cas9 protein (Nishimasu et al. even plant protoplasts (Woo et al. 2015). The
2014), and that PAM recognition is mediated incorporation of specific chemical modifica-
by an arginine-rich motif present in Cas9 (An- tions known to protect RNA from nuclease deg-
ders et al. 2014). Doudna and colleagues have radation and stabilize secondary structure can
since proposed that DNA strand displacement further enhance Cas9 ribonucleoprotein activi-
induces a structural rearrangement within the ty (Hendel et al. 2015; Rahdar et al. 2015). In a
Cas9 protein that directs the nontarget DNA clever marriage of genome-editing platforms,
strand into the RuvC active site, which then the FokI cleavage domain has even been fused
positions the HNH domain near target DNA to an inactivated Cas9 variant to generate hy-
(Jiang et al. 2016), enabling Cas9-mediated brid nucleases that require protein dimerization
cleavage of both DNA strands. for DNA cleavage (Guilinger et al. 2014b; Tsai
The Cas9 nuclease and its gRNA can be de- et al. 2014), theoretically increasing CRISPR-
livered into cells for genome editing on the same Cas9 specificity. Similarly, fusing Cas9 to
or separate plasmids, and numerous resources DNA-binding domains has also proven effective
have been developed to facilitate target site at improving its specificity (Bolukbasi et al.
selection and gRNA construction, including 2015). Finally, several studies have recently
E-CRISP (Heigwer et al. 2014), among others. showed that protein engineering can broadly
Although Cas9 boasts the highest ease of use enhance Cas9 specificity (Kleinstiver et al.
among the targeted nuclease platforms, several 2016; Slaymaker et al. 2016) and even alter its
reports have indicated that it could be prone to PAM requirements (Kleinstiver et al. 2015), the
inducing off-target mutations (Cradick et al. latter having the potential to enable creation of
2013; Fu et al. 2013). To this end, considerable customized variants of Cas9 for allele-specific
effort has been devoted to improving the spe- gene editing, although Cas9 orthologs (Cong
Genome-Editing Technologies
et al. 2013; Esvelt et al. 2013; Hou et al. 2013; lines carrying nearly any genomic modification
Ran et al. 2015) or alternative CRISPR systems can now be generated in simply a matter of
(Zetsche et al. 2015a) with unique PAM speci- weeks. Examples of some of the outcomes that
ficities have been uncovered in nature. have become routine because of the emergence
of targeted nucleases include gene knockout
Homing Endonucleases (Santiago et al. 2008; Mali et al. 2013b), gene
deletion (Lee et al. 2010), gene inversion (Xiao
Homing endonucleases, also known as mega- et al. 2013), gene correction (Urnov et al. 2005;
nucleases, represent the final member of the Ran et al. 2013), gene addition (Moehle et al.
targeted nuclease family. These enzymes have 2007; Hockemeyer et al. 2011; Hou et al. 2013),
been reviewed at length elsewhere (Silva et al. and even chromosomal translocation (Fig. 2)
2011; Stoddard 2014) but, briefly, members of (Torres et al. 2014). In addition to cell line en-
the LAGLIDADG family of endonucleases— gineering, targeted nucleases have also expedit-
so named for the conserved amino acid motif ed the generation of genetically modified or-
present within these enzymes that interacts with ganisms, facilitating the rapid creation of
DNA—are a collection of naturally occurring transgenic zebrafish (Doyon et al. 2008; Sander
enzymes that recognize and cleave long DNA et al. 2011a; Hwang et al. 2013), mice (Cui et al.
sequences (14 – 40 bps) (Fig. 1). These enzymes 2011; Wang et al. 2013; Wu et al. 2013), rats
make extensive sequence-specific contacts with (Geurts et al. 2009; Tesson et al. 2011; Li et al.
their DNA substrate (Stoddard 2011), and thus 2013), monkeys (Liu et al. 2014c), and livestock
typically show exquisite specificity. However, (Hauschild et al. 2011; Carlson et al. 2012),
unlike ZFNs and TALENs, the binding and which together have the capacity to accelerate
cleavage domains in homing endonucleases human disease modeling and the discovery of
are not modular. This overlap in form and func- new therapeutics.
tion make their repurposing challenging, and Targeted nucleases have also emerged as
limits their utility for more routine applications powerful tools for plant engineering (Baltes
of genome editing. More recently megaTALs— and Voytas 2015). Both TALENs and CRISPR-
fusions of a rare-cleaving homing endonuclease Cas9 have been used to modify multiple alleles
to a TALE-binding domain—have been report- within hexaploid bread wheat to confer herita-
ed to induce highly specific gene modifications ble resistance to powdery mildew (Wang et al.
(Boissel et al. 2014; Lin et al. 2015a). These en- 2014b). In another study, TALENs were used to
zymes have enabled integration of antitumor knock out nonessential genes in the fatty acid
and anti-HIV factors into the human CCR5 metabolic pathway in soybean to generate a sim-
gene in both primary T cells and hematopoietic plified plant cell with reduced metabolic com-
stem/progenitor cells (Sather et al. 2015), as ponents (Haun et al. 2014). Of special note, two
well as disruption of endogenous T-cell receptor recent reports showed that purified nuclease
elements in T cells (Osborn et al. 2016), indi- proteins can be introduced directly into plant
cating their potential for enabling and enhanc- protoplasts, enabling the introduction of germ-
ing immunotherapies. line-transmissible modifications that are virtu-
ally indistinguishable from naturally occurring
GENOME-EDITING APPLICATIONS (Luo et al. 2015; Woo et al. 2015). This technical
advance could help to overcome certain regula-
Engineering Cell Lines and Organisms tory hurdles associated with the use of transgen-
Before the emergence of engineered nucleases, ic crops. Finally, targeted nucleases have also
genetically modifying mammalian cell lines was been used to inactivate pathogenic genes to pre-
labor intensive, costly, and often times limited vent viral (Lin et al. 2014) or parasitic (Ghorbal
to laboratories with specialized expertise. How- et al. 2014) infection, as well as to introduce
ever, with the advent of cost-effective and user- knockin-specific factors capable of imparting
friendly gene-editing technologies, custom cell pathogen resistance (Wu et al. 2015).
T. Gaj et al.
Intriguingly, targeted nucleases could also for functional genomics (Hilton and Gersbach
serve as conduits for curbing mosquito- or in- 2015), having facilitated the discovery of geno-
sect-borne diseases through a technique known mic loci that confer drug resistance to cells
as gene drive (Burt 2003; Sinkins and Gould (Koike-Yusa et al. 2014; Shalem et al. 2014;
2006), which harnesses genome editing to facil- Wang et al. 2014a; Zhou et al. 2014), uncovered
itate the introduction of a specific gene or mu- how cells can control induction of the host
tation that can then confer a particular pheno- immune response (Parnas et al. 2015), provided
type into a host and also be transmitted to its new insights into the genetic basis of cellular
progeny (Windbichler et al. 2011). Gene drives fitness (Hart et al. 2015; Wang et al. 2015b),
have now been tested in the malaria vector mos- and even shed light on how certain viruses in-
quitos Anopheles stephensi (Gantz et al. 2015) duce cell death (Ma et al. 2015). Genome-wide
and Anopheles gambiae (Hammond et al. 2016) CRISPR screens can also facilitate the discovery
as a means for achieving population control of functional noncoding elements (Kim et al.
and to prevent disease transmission, respective- 2013b; Korkmaz et al. 2016), and provide a
ly. However, owing to the ease with which means for studying the structure and evolution
CRISPR-Cas9 can be programmed (Gantz and of the human genome. In a remarkable example
Bier 2015), debate has ignited on the potential of the latter, Shendure and colleagues used Cas9
societal and environmental impact of this tech- to mediate integration of short randomized
nology (Esvelt et al. 2014; Akbari et al. 2015), DNA sequences into the BRCA1 and DBR1
spurring the development of safeguard ele- genes (Findlay et al. 2014). They then measured
ments that could help to minimize the risk of the functional consequences of these mutations
gene-edited organisms escaping from the labo- on fitness, achieving an unprecedented look at
ratory (DiCarlo et al. 2015). some of the factors driving genome and disease
evolution. Finally, CRISPR screens have even
proven effective in vivo, enabling the identifica-
Synthetic Biology and Genome-Scale
tion of factors involved in zebrafish develop-
Engineering
ment (Shah et al. 2015) and disease progression
Targeted nucleases also offer a facile means for in mice (Chen et al. 2015).
generating modified bacterial and yeast strains
for synthetic biology, including metabolic path-
Therapeutic Genome Editing
way engineering. Bacterial species of the order
Actinomycetales, for instance, are one of the Genome editing itself also holds tremendous
most important sources of industrially relevant potential for treating the underlying genetic
secondary metabolites. However, many Actino- causes of certain diseases (Cox et al. 2015; Por-
mycetales species are recalcitrant to genetic teus 2015; Maeder and Gersbach 2016). In one
manipulation, a fact that has severely hampered of the most successful examples of this to date,
their use for metabolic engineering. CRISPR- ZFN-mediated disruption of the HIV corecep-
Cas9 has been used to inactivate multiple genes tor CCR5 was used to engineer HIV resistance
in actinomycetes (Tong et al. 2015), indicating into both CD4þ T cells (Perez et al. 2008) and
its ability to enable the creation of designer CD34þ hematopoietic stem/progenitor cells
bacterial strains with enhanced metabolite (HSPCs) (Holt et al. 2010), proving safe and
production capabilities. CRISPR has also facil- well-tolerated in a phase I clinical trial that in-
itated multiplexed metabolic pathway engi- fused these gene-modified T cells into individ-
neering in yeast at high efficiencies (Jakociunas uals with HIV/AIDS (Tebas et al. 2014). In ad-
et al. 2015a,b), as well as random mutagenesis dition to enabling the introduction of gene
of yeast chromosomal DNA for phenotypic modification that can enhance autologous cell
screening of desired mutants (Ryan et al. therapies, targeted nucleases can also be com-
2014). Indeed, genome-wide CRISPR-based bined with viral vectors—including AAV—to
knockout screens hold tremendous potential mediate genome editing in situ (Gaj et al.
Genome-Editing Technologies
2016). For instance, delivery of an AAV vector domains. Among the first fully synthetic tran-
encoding a ZFN pair designed to target a defec- scriptional effector proteins to be generated
tive copy of the factor IX gene, along with its (Beerli et al. 1998) were those based on the fu-
repair template, led to efficient gene correction sion of engineered zinc-finger proteins with
in mouse liver, increasing factor IX protein pro- either the Herpes simplex– derived transactiva-
duction in both neonatal (Li et al. 2011) and tion domain (Sadowski et al. 1988) or the Krüp-
adult (Anguela et al. 2013) models of the dis- pel-associated box (KRAB) repression protein
ease. In vivo genome editing also recently en- (Margolin et al. 1994). Over the course of the
abled the restoration of dystrophin gene expres- next 15 years, zinc-finger-based transcriptional
sion and the rescue of muscle function in mouse modulators were expanded and featured sever-
models of Duchenne muscular dystrophy (Long al other types of effector domains (Beerli and
et al. 2015; Nelson et al. 2015; Tabebordbar et al. Barbas 2002), including, for example, the
2015). Therapeutic gene editing in a mouse Dnmt3a methyltransferase domain (Rivenbark
model of human hereditary tyrosinemia has et al. 2012; Siddique et al. 2013) and the ten-elev-
also been reported using both hydrodynamic en translocation methylcytosine dioxygenase 1
injection of plasmid DNA encoding CRISPR- (TET1) (Chen et al. 2014), which can modulate
Cas9 (Yin et al. 2014) and by combining nano- transcription via targeted methylation or de-
particle-mediated delivery of Cas9-encoding methylation, respectively. As a natural extension
mRNA with AAV-mediated delivery of the of zinc-finger transcription factors, and further
DNA template for gene correction (Yin et al. drawing on the parallels with zinc-finger pro-
2016). More recently, a dual particle AAV sys- teins, TALE transcription factors have also
tem, wherein one AAV vector carried the Cas9 emerged as an especially effective platform for
nuclease and a second harbored the gRNA and achieving targeted transcriptional modulation
donor repair template, was able to mediate cor- (Miller et al. 2011; Zhang et al. 2011). Effector
rection of a disease-causing mutation in the or- domains are generally fused to the carboxyl ter-
nithine transcarbamylase gene in the liver of a minus of the synthetic TALE array and, contrary
neonatal model of the disease (Yang et al. 2016). to the longer sequence typically required for
This work, in particular, showed that therapeu- efficient modulation by zinc-finger transcrip-
tic levels of gene correction could be achieved in tion factors, TALEs have been reported to regu-
a regenerating tissue even when using multiple late gene expression with as few as 10.5 repeats
AAV particles. Although highly promising, nu- (Boch et al. 2009). Like zinc fingers, TALEs are
merous hurdles still need to be overcome for in also compatible with numerous epigenetic
vivo applications of genome editing to reach its modifiers, including the TET1 hydroxylase cat-
full potential. Chief among these are methods alytic domain (Maeder et al. 2013b) and the
that can facilitate nuclease delivery or expression lysine-specific histone demethylase 1 (LSD1)
to only diseased cells or tissues, and the devel- (Mendenhall et al. 2013) domains, which have
opment of new strategies that can enhance HDR been used for targeted CpG demethylation and
in disease-associated postmitotic cells in vivo. histone demethylation, respectively. In particu-
lar, the ease with which a large number of TALEs
TARGETED TRANSCRIPTION FACTORS can be constructed has enabled the discovery
that tiling a promoter sequence with combina-
Tools for Modulating Gene Expression tions of synthetic transcription factors can lead
The modular qualities of zinc-finger and TALE to a synergistic increase in gene expression
proteins, in addition to the highly flexible DNA (Maeder et al. 2013b; Perez-Pinera et al. 2013).
recognition ability of CRISPR-Cas9, also pro- And, like zinc fingers (Beerli et al. 2000b; Pol-
vide investigators with the ability to modulate lock et al. 2002; Magnenat et al. 2008; Polstein
the expression of nearly any gene from its pro- and Gersbach 2012), TALE activators have also
moter or enhancer sequences via their fusion to been successfully engineered to regulate gene
transcriptional activator and repressor protein expression in response to external (Mercer
T. Gaj et al.
et al. 2014) or endogenous (Li et al. 2012) chem- velopment of second-generation CRISPR acti-
ical stimuli, optical signals (Konermann et al. vators quickly emerged as a highly active area of
2013), and even proteolytic cues (Copeland research. One particularly elegant approach for
et al. 2016; Lonzaric et al. 2016). overcoming the low activation thresholds inher-
Because of the exquisite ease with which it ent within first-generation systems was by stra-
can be programmed, the CRISPR-Cas9 system tegically inserting an RNA aptamer within a
has also been adapted for transcriptional mod- functionally inert region of the gRNA. This ap-
ulation through fusion of specific effector do- tamer recruits specific activation helper pro-
mains to a catalytically inactivated variant of the teins that work in concert with a dCas9 activator
Cas9 protein (Qi et al. 2013). Deactivation is to enhance transcription (Konermann et al.
achieved by introducing two amino acid substi- 2015; Zalatan et al. 2015). Other strategies based
tutions, D10A and H840A, into the RuvC and on directly fusing additional helper activation
NHN endonuclease domains of Cas9, respec- domains to dCas9 have also been shown to en-
tively. Although unable to cleave DNA, this mu- hance transcription (Chavez et al. 2015). Target-
tant, referred to as dCas9, retains its ability to ed acetylation of histone proteins within a pro-
bind DNA in an RNA-directed manner. Effector moter or enhancer sequence via epigenome
domains are fused to the carboxyl terminus of editing using the catalytic core of the human
the dCas9 protein and can modulate gene ex- acetyltransferase p300 fused to dCas9 can also
pression from either strand of the targeted DNA lead to robust levels of gene activation (Hilton
sequence (Farzadfard et al. 2013; Maeder et al. et al. 2015). Similarly, dCas9 repressor proteins
2013a; Perez-Pinera et al. 2013). Genome-scale targeted to distal regulatory elements have been
activation studies have indicated that the most found to facilitate chromatin remodeling and
robust levels of activation are generally observed gene repression via epigenomic modification
when dCas9 activators are targeted to -400 to (Thakore et al. 2015). Finally, by simply reduc-
-50 bp upstream from the transcriptional start ing the length of the gRNA, catalytically active
site (Gilbert et al. 2014; Hu et al. 2014). Addi- variants of Cas9 can stimulate transcription
tionally, dCas9 can inhibit gene expression by without inducing DNA breaks (Dahlman et al.
simply blocking transcriptional initiation or 2015; Kiani et al. 2015), enabling orthogonal
elongation through a process known as CRISPR gene knockout and activation with the same
interference (Qi et al. 2013), although fusing Cas9 variant in a single cell.
dCas9 to transcriptional repressor domains
can also lead to efficient silencing from the pro-
Applications of Targeted Transcriptional
moter (Gilbert et al. 2013; Zalatan et al. 2015).
Regulation
Much like zinc fingers and TALEs, methods for
achieving conditional gene modulation using Early work on the use of engineered zinc-finger
dCas9 have also been reported, including the transcription factors revealed that synthetic
fusion of a dihydrofolate reductase destabiliza- transcriptional modulators are effective tools
tion domain to dCas9, which can provide for a broad range of applications, enabling
chemical control over activation, enabling cel- such tasks as inhibiting viral replication (Pap-
lular reprogramming or differentiation (Balboa worth et al. 2003; Reynolds et al. 2003; Segal
et al. 2015). Light-inducible dCas9-based sys- et al. 2004; Eberhardy et al. 2006), modulating
tems capable of providing optical control of the expression of disease-associated loci (Gras-
gene expression provide another means for lund et al. 2005; Wilber et al. 2010), inducing
achieving conditional control of gene expres- angiogenesis for accelerated wound healing (Re-
sion (Nihongaki et al. 2015; Polstein and Gers- bar et al. 2002), and genomic screening of cel-
bach 2015). lular targets for cancer progression and drug
Although flexible, first-generation dCas9 resistance (Park et al. 2003; Blancafort et al.
activators were routinely found to display sub- 2005, 2008). Facilitated by many of the insights
optimal levels of activation. As a result, the de- gained from zinc-finger transcription factor
Genome-Editing Technologies
technology, both TALEs and CRISPR-Cas9 have DNA sequences (Gaj et al. 2013; Sirk et al. 2014;
now further expanded the possibilities of engi- Wallen et al. 2015) and even integrate therapeu-
neered transcriptional activators and repressors. tic factors into the human genome (Gaj et al.
For example, TALEs and CRISPR-Cas9 have 2014b), are one such option. More recent work
enabled rapid construction of custom genetic has indicated that single-base editing without
circuits and logic gates (Gaber et al. 2014; Lebar DNA breaks can be achieved using an engi-
et al. 2014; Liu et al. 2014b), complex gene neered Cas9 nickase complex (Komor et al.
regulation networks (Nielsen and Voigt 2014; 2016), although it remains unknown how effec-
Nissim et al. 2014), and even facilitated cellular tive this technology is in therapeutically rele-
reprogramming (Gao et al. 2013) and the dif- vant settings. By linking genomic modifications
ferentiation of mouse embryonic fibroblasts to induced by targeted nucleases to their own self-
skeletal myocytes (Chakraborty et al. 2014). degradation, self-inactivating vectors are also
dCas9 transcriptional effectors have even been poised to improve the specificity of genome
used to efficiently mediate repression and acti- editing, especially because the frequency of
vation of endogenous genes in Drosophila (Lin off-target modifications can be directly propor-
et al. 2015b) and in plant cells (Piatek et al. tional to the duration of cellular exposure to a
2015). Both TALE and Cas9 activators have nuclease (Pruett-Miller et al. 2009). In addition,
also been configured to stimulate transcription much of the knowledge behind genome engi-
of latent HIV (Zhang et al. 2015; Ji et al. 2016; neering has been obtained in immortalized cell
Limsirichai et al. 2016; Perdigao et al. 2016; lines. However, in the case of regenerative med-
Saayman et al. 2016), indicating their potential icine, it is highly desirable to genetically manip-
to work in concert with antiretroviral therapy ulate progenitor or stem-cell populations, both
for eradicating HIV infection. Importantly, of which can differ markedly from transformed
because of the ease with which the CRISPR- cell lines with respect to their epigenome or
Cas9 system can be used, genome-wide screens three-dimensional organization of their geno-
using Cas9 transcriptional activators (Gilbert mic DNA. These differences can have profound
et al. 2014; Konermann et al. 2015) and re- effects on the ability of genome-editing tools to
pressors (Gilbert et al. 2014) can be easily either modify a specific sequence or regulate
implemented to discover genes involved in a gene expression. Although the union between
number of diverse processes, including drug genome engineering and regenerative medicine
resistance and cancer metastasis. In particular, is still in its infancy, realizing the full potential of
CRISPR-based genome-scale screening meth- these technologies in stem/progenitor cells re-
ods have the potential to overcome many of quires that their functional landscape be fully
the technical hurdles associated with other explored in these genetic backgrounds. Only
contemporary screening technologies, such as then will genome editing technologies truly be
cDNA libraries and RNAi, indicating its poten- able to reprogram cell fate and behavior for the
tial for facilitating drug discovery and basic next generation of advances in synthetic biology
biological research. and gene therapy.
CONCLUSIONS ACKNOWLEDGMENTS
Despite the successes already achieved, many We gratefully acknowledge the support and
challenges remain before the full potential of mentorship of the late Carlos F. Barbas, III
genome editing can be realized. First and fore- (1964– 2014). This work is supported by the
most are the development of new tools cap- National Institutes of Health (F32GM113446
able of introducing genomic modifications in to T.G.) and ShanghaiTech University (to J.L.).
the absence of DNA breaks. Targeted recombi- We apologize to those whose important contri-
nases (Akopian et al. 2003; Mercer et al. 2012), butions were not cited because of space con-
which can be programmed to recognize specific straints.
T. Gaj et al.
Genome-Editing Technologies
somes by using the I-SceI system of Saccharomyces cere- tificial transcription factors. J Biol Chem 280: 35588–
visiae. Mol Cell Biol 15: 1968–1973. 35597.
Christian M, Cermak T, Doyle EL, Schmidt C, Zhang F, Eberhardy SR, Goncalves J, Coelho S, Segal DJ, Berkhout B,
Hummel A, Bogdanove AJ, Voytas DF. 2010. Targeting Barbas CF III. 2006. Inhibition of human immunodefi-
DNA double-strand breaks with TAL effector nucleases. ciency virus type 1 replication with artificial transcription
Genetics 186: 757– 761. factors targeting the highly conserved primer-binding
Cong L, Zhou R, Kuo YC, Cunniff M, Zhang F. 2012. Com- site. J Virol 80: 2873–2883.
prehensive interrogation of natural TALE DNA-binding Esvelt KM, Mali P, Braff JL, Moosburner M, Yaung SJ,
modules and transcriptional repressor domains. Nat Church GM. 2013. Orthogonal Cas9 proteins for RNA-
Commun 3: 968. guided gene regulation and editing. Nat Methods 10:
Cong L, Ran FA, Cox D, Lin S, Barretto R, Habib N, Hsu PD, 1116–1121.
Wu X, Jiang W, Marraffini LA, et al. 2013. Multiplex Esvelt KM, Smidler AL, Catteruccia F, Church GM. 2014.
genome engineering using CRISPR/Cas systems. Science Concerning RNA-guided gene drives for the alteration of
339: 819– 823. wild populations. eLife doi: 10.7554/eLife.03401.
Copeland MF, Politz MC, Johnson CB, Markley AL, Pfleger Farzadfard F, Perli SD, Lu TK. 2013. Tunable and multifunc-
BF. 2016. A transcription activator-like effector (TALE) tional eukaryotic transcription factors based on
induction system mediated by proteolysis. Nat Chem Biol CRISPR/Cas. ACS Synth Biol 2: 604– 613.
12: 254 –260. Findlay GM, Boyle EA, Hause RJ, Klein JC, Shendure J. 2014.
Cox DB, Platt RJ, Zhang F. 2015. Therapeutic genome edit- Saturation editing of genomic regions by multiplex ho-
ing: Prospects and challenges. Nat Med 21: 121– 131. mology-directed repair. Nature 513: 120– 123.
Cradick TJ, Fine EJ, Antico CJ, Bao G. 2013. CRISPR/Cas9 Fu Y, Foden JA, Khayter C, Maeder ML, Reyon D, Joung JK,
systems targeting b-globin and CCR5 genes have sub- Sander JD. 2013. High-frequency off-target mutagenesis
induced by CRISPR-Cas nucleases in human cells. Nat
stantial off-target activity. Nucleic Acids Res 41: 9584–
Biotechnol 31: 822– 826.
9592.
Fu Y, Sander JD, Reyon D, Cascio VM, Joung JK. 2014.
Cui X, Ji D, Fisher DA, Wu Y, Briner DM, Weinstein EJ. 2011.
Improving CRISPR-Cas nuclease specificity using trun-
Targeted integration in rat and mouse embryos with zinc-
cated guide RNAs. Nat Biotechnol 32: 279 –284.
finger nucleases. Nat Biotechnol 29: 64– 67.
Gaber R, Lebar T, Majerle A, Ster B, Dobnikar A, Bencina M,
Dahlman JE, Abudayyeh OO, Joung J, Gootenberg JS,
Jerala R. 2014. Designable DNA-binding domains enable
Zhang F, Konermann S. 2015. Orthogonal gene knockout construction of logic circuits in mammalian cells. Nat
and activation with a catalytically active Cas9 nuclease. Chem Biol 10: 203 –208.
Nat Biotechnol 33: 1159– 1161.
Gabriel R, Lombardo A, Arens A, Miller JC, Genovese P,
Davis KM, Pattanayak V, Thompson DB, Zuris JA, Liu DR. Kaeppel C, Nowrouzi A, Bartholomae CC, Wang J, Fried-
2015. Small molecule-triggered Cas9 protein with im- man G, et al. 2011. An unbiased genome-wide analysis of
proved genome-editing specificity. Nat Chem Biol 11: zinc-finger nuclease specificity. Nat Biotechnol 29: 816–
316–318. 823.
Deng D, Yan C, Pan X, Mahfouz M, Wang J, Zhu JK, Shi Y, Gaj T, Guo J, Kato Y, Sirk SJ, Barbas CF III. 2012. Targeted
Yan N. 2012. Structural basis for sequence-specific rec- gene knockout by direct delivery of zinc-finger nuclease
ognition of DNA by TAL effectors. Science 335: 720 –723. proteins. Nat Methods 9: 805 –807.
DiCarlo JE, Chavez A, Dietz SL, Esvelt KM, Church GM. Gaj T, Mercer AC, Sirk SJ, Smith HL, Barbas CF III. 2013. A
2015. Safeguarding CRISPR-Cas9 gene drives in yeast. comprehensive approach to zinc-finger recombinase cus-
Nat Biotechnol 33: 1250– 1255. tomization enables genomic targeting in human cells.
Doyon Y, McCammon JM, Miller JC, Faraji F, Ngo C, Kati- Nucleic Acids Res 41: 3937–3946.
bah GE, Amora R, Hocking TD, Zhang L, Rebar EJ, et al. Gaj T, Liu J, Anderson KE, Sirk SJ, Barbas CF III. 2014a.
2008. Heritable targeted gene disruption in zebrafish us- Protein delivery using Cys2-His2 zinc-finger domains.
ing designed zinc-finger nucleases. Nat Biotechnol 26: ACS Chem Biol 9: 1662– 1667.
702–708. Gaj T, Sirk SJ, Tingle RD, Mercer AC, Wallen MC, Barbas CF
Doyon Y, Vo TD, Mendel MC, Greenberg SG, Wang J, Xia III. 2014b. Enhancing the specificity of recombinase-me-
DF, Miller JC, Urnov FD, Gregory PD, Holmes MC. 2011. diated genome engineering through dimer interface re-
Enhancing zinc-finger-nuclease activity with improved design. J Am Chem Soc 136: 5047–5056.
obligate heterodimeric architectures. Nat Methods 8: Gaj T, Epstein BE, Schaffer DV. 2016. Genome engineering
74–79. using adeno-associated virus: Basic and clinical research
Dreier B, Beerli RR, Segal DJ, Flippin JD, Barbas CF III. applications. Mol Ther 24: 458–464.
2001. Development of zinc finger domains for recogni- Gantz VM, Bier E. 2015. The mutagenic chain reaction: A
tion of the 50 -ANN-30 family of DNA sequences and their method for converting heterozygous to homozygous mu-
use in the construction of artificial transcription factors. tations. Science 348: 442– 444.
J Biol Chem 276: 29466–29478. Gantz VM, Jasinskiene N, Tatarenkova O, Fazekas A, Macias
Dreier B, Fuller RP, Segal DJ, Lund CV, Blancafort P, Huber VM, Bier E, James AA. 2015. Highly efficient Cas9-me-
A, Koksch B, Barbas CF III. 2005. Development of zinc diated gene drive for population modification of the ma-
finger domains for recognition of the 50 -CNN-30 family laria vector mosquito Anopheles stephensi. Proc Natl Acad
DNA sequences and their use in the construction of ar- Sci 112: E6736–E6743.
T. Gaj et al.
Gao X, Yang J, Tsang JC, Ooi J, Wu D, Liu P. 2013. Repro- Hauschild J, Petersen B, Santiago Y, Queisser AL, Carnwath
gramming to pluripotency using designer TALE tran- JW, Lucas-Hahn A, Zhang L, Meng X, Gregory PD,
scription factors targeting enhancers. Stem Cell Rep 1: Schwinzer R, et al. 2011. Efficient generation of a biallelic
183–197. knockout in pigs using zinc-finger nucleases. Proc Natl
Geurts AM, Cost GJ, Freyvert Y, Zeitler B, Miller JC, Choi Acad Sci 108: 12013–12017.
VM, Jenkins SS, Wood A, Cui X, Meng X, et al. 2009. Heigwer F, Kerr G, Boutros M. 2014. E-CRISP: Fast CRISPR
Knockout rats via embryo microinjection of zinc-finger target site identification. Nat Methods 11: 122 –123.
nucleases. Science 325: 433. Hendel A, Bak RO, Clark JT, Kennedy AB, Ryan DE, Roy
Ghorbal M, Gorman M, Macpherson CR, Martins RM, S, Steinfeld I, Lunstad BD, Kaiser RJ, Wilkens AB, et
Scherf A, Lopez-Rubio JJ. 2014. Genome editing in the al. 2015. Chemically modified guide RNAs enhance
human malaria parasite Plasmodium falciparum using CRISPR-Cas genome editing in human primary cells.
the CRISPR-Cas9 system. Nat Biotechnol 32: 819– 821. Nat Biotechnol 33: 985 –989.
Gilbert LA, Larson MH, Morsut L, Liu Z, Brar GA, Torres Hilton IB, Gersbach CA. 2015. Enabling functional geno-
SE, Stern-Ginossar N, Brandman O, Whitehead EH, mics with genome engineering. Genome Res 25: 1442–
Doudna JA, et al. 2013. CRISPR-mediated modular 1455.
RNA-guided regulation of transcription in eukaryotes. Hilton IB, D’Ippolito AM, Vockley CM, Thakore PI, Craw-
Cell 154: 442– 451. ford GE, Reddy TE. 2015. Epigenome editing by a
Gilbert LA, Horlbeck MA, Adamson B, Villalta JE, Chen Y, CRISPR-Cas9-based acetyltransferase activates genes
Whitehead EH, Guimaraes C, Panning B, Ploegh HL, from promoters and enhancers. Nat Biotechnol 33:
Bassik MC, et al. 2014. Genome-scale CRISPR-mediated 510 –517.
control of gene repression and activation. Cell 159: 647 – Hockemeyer D, Wang H, Kiani S, Lai CS, Gao Q, Cassady JP,
661. Cost GJ, Zhang L, Santiago Y, Miller JC, et al. 2011. Ge-
Graslund T, Li X, Magnenat L, Popkov M, Barbas CF III. netic engineering of human pluripotent cells using TALE
2005. Exploring strategies for the design of artificial tran- nucleases. Nat Biotechnol 29: 731 –734.
scription factors: Targeting sites proximal to known reg- Holkers M, Maggio I, Liu J, Janssen JM, Miselli F, Mussolino
ulatory regions for the induction of g-globin expression C, Recchia A, Cathomen T, Goncalves MA. 2013. Differ-
and the treatment of sickle cell disease. J Biol Chem 280: ential integrity of TALE nuclease genes following adeno-
3707– 3714. viral and lentiviral vector gene transfer into human cells.
Greisman HA, Pabo CO. 1997. A general strategy for select- Nucleic Acids Res 41: e63.
ing high-affinity zinc finger proteins for diverse DNA Holkers M, Maggio I, Henriques SF, Janssen JM, Cathomen
target sites. Science 275: 657– 661. T, Goncalves MA. 2014. Adenoviral vector DNA for ac-
Guilinger JP, Pattanayak V, Reyon D, Tsai SQ, Sander JD, curate genome editing with engineered nucleases. Nat
Joung JK, Liu DR. 2014a. Broad specificity profiling of Methods 11: 1051–1057.
TALENs results in engineered nucleases with improved Holt N, Wang J, Kim K, Friedman G, Wang X, Taupin V,
DNA-cleavage specificity. Nat Methods 11: 429–435. Crooks GM, Kohn DB, Gregory PD, Holmes MC, et al.
Guilinger JP, Thompson DB, Liu DR. 2014b. Fusion of cat- 2010. Human hematopoietic stem/progenitor cells mod-
alytically inactive Cas9 to FokI nuclease improves the ified by zinc-finger nucleases targeted to CCR5 control
specificity of genome modification. Nat Biotechnol 32: HIV-1 in vivo. Nat Biotechnol 28: 839 –847.
577–582. Horvath P, Barrangou R. 2010. CRISPR/Cas, the immune
Guo J, Gaj T, Barbas CF III. 2010. Directed evolution of an system of bacteria and archaea. Science 327: 167–170.
enhanced and highly efficient FokI cleavage domain for Hou Z, Zhang Y, Propson NE, Howden SE, Chu LF, Son-
zinc finger nucleases. J Mol Biol 400: 96–107. theimer EJ, Thomson JA. 2013. Efficient genome engi-
Gupta A, Christensen RG, Rayla AL, Lakshmanan A, neering in human pluripotent stem cells using Cas9 from
Stormo GD, Wolfe SA. 2012. An optimized two-finger Neisseria meningitidis. Proc Natl Acad Sci 110: 15644–
archive for ZFN-mediated gene targeting. Nat Methods 9: 15649.
588–590. Hsu PD, Scott DA, Weinstein JA, Ran FA, Konermann S,
Hammond A, Galizi R, Kyrou K, Simoni A, Siniscalchi C, Agarwala V, Li Y, Fine EJ, Wu X, Shalem O, et al. 2013.
Katsanos D, Gribble M, Baker D, Marois E, Russell S, et al. DNA targeting specificity of RNA-guided Cas9 nucleases.
2016. A CRISPR-Cas9 gene drive system targeting female Nat Biotechnol 31: 827 –832.
reproduction in the malaria mosquito vector Anopheles Hu J, Lei Y, Wong WK, Liu S, Lee KC, He X, You W, Zhou R,
gambiae. Nat Biotechnol 34: 78–83. Guo JT, Chen X, et al. 2014. Direct activation of human
Hart T, Chandrashekhar M, Aregger M, Steinhart Z, Brown and mouse Oct4 genes using engineered TALE and Cas9
KR, MacLeod G, Mis M, Zimmermann M, Fradet-Tur- transcription factors. Nucleic Acids Res 42: 4375–4390.
cotte A, Sun S, et al. 2015. High-resolution CRISPR Hubbard BP, Badran AH, Zuris JA, Guilinger JP, Davis KM,
screens reveal fitness genes and genotype-specific cancer Chen L, Tsai SQ, Sander JD, Joung JK, Liu DR. 2015.
liabilities. Cell 163: 1515– 1526. Continuous directed evolution of DNA-binding proteins
Haun W, Coffman A, Clasen BM, Demorest ZL, Lowy A, Ray to improve TALEN specificity. Nat Methods 12: 939– 942.
E, Retterath A, Stoddard T, Juillerat A, Cedrone F, et al. Hurt JA, Thibodeau SA, Hirsh AS, Pabo CO, Joung JK. 2003.
2014. Improved soybean oil quality by targeted mutagen- Highly specific zinc finger proteins obtained by directed
esis of the fatty acid desaturase 2 gene family. Plant Bio- domain shuffling and cell-based selection. Proc Natl Acad
technol J 12: 934– 940. Sci 100: 12271–12276.
Genome-Editing Technologies
Hwang WY, Fu Y, Reyon D, Maeder ML, Tsai SQ, Sander JD, Kim YK, Wee G, Park J, Kim J, Baek D, Kim JS, Kim VN.
Peterson RT, Yeh JR, Joung JK. 2013. Efficient genome 2013b. TALEN-based knockout library for human micro-
editing in zebrafish using a CRISPR-Cas system. Nat Bio- RNAs. Nat Struct Mol Biol 20: 1458–1464.
technol 31: 227–229. Kim S, Kim D, Cho SW, Kim J, Kim JS. 2014. Highly efficient
Isalan M, Klug A, Choo Y. 2001. A rapid, generally applicable RNA-guided genome editing in human cells via delivery
method to engineer zinc fingers illustrated by targeting of purified Cas9 ribonucleoproteins. Genome Res 24:
the HIV-1 promoter. Nat Biotechnol 19: 656–660. 1012–1019.
Jakociunas T, Bonde I, Herrgard M, Harrison SJ, Kristensen Kleinstiver BP, Prew MS, Tsai SQ, Topkar VV, Nguyen NT,
M, Pedersen LE, Jensen MK, Keasling JD. 2015a. Multi- Zheng Z, Gonzales AP, Li Z, Peterson RT, Yeh JR, et al.
plex metabolic pathway engineering using CRISPR/Cas9 2015. Engineered CRISPR-Cas9 nucleases with altered
in Saccharomyces cerevisiae. Metab Eng 28: 213–222. PAM specificities. Nature 523: 481– 485.
Jakociunas T, Rajkumar AS, Zhang J, Arsovska D, Rodriguez Kleinstiver BP, Pattanayak V, Prew MS, Tsai SQ, Nguyen NT,
A, Jendresen CB, Skjodt ML, Nielsen AT, Borodina I, Zheng Z, Joung JK. 2016. High-fidelity CRISPR-Cas9
Jensen MK, et al. 2015b. CasEMBLR: Cas9-facilitated nucleases with no detectable genome-wide off-target ef-
multiloci genomic integration of in vivo assembled fects. Nature 529: 490– 495.
DNA parts in Saccharomyces cerevisiae. ACS Synth Biol Koike-Yusa H, Li Y, Tan EP, Velasco-Herrera Mdel C, Yusa K.
4: 1226– 1234. 2014. Genome-wide recessive genetic screening in mam-
Jamieson AC, Kim SH, Wells JA. 1994. In vitro selection of malian cells with a lentiviral CRISPR-guide RNA library.
zinc fingers with altered DNA-binding specificity. Bio- Nat Biotechnol 32: 267 –273.
chemistry 33: 5689–5695. Komor AC, Kim YB, Packer MS, Zuris JA, Liu DR. 2016.
Ji H, Jiang Z, Lu P, Ma L, Li C, Pan H, Fu Z, Qu X, Wang P, Programmable editing of a target base in genomic DNA
Deng J, et al. 2016. Specific reactivation of latent HIV-1 without double-stranded DNA cleavage. Nature doi:
by dCas9-SunTag-VP64-mediated guide RNA targeting 10.1038/nature17946.
the HIV-1 promoter. Mol Ther 24: 508–521. Konermann S, Brigham MD, Trevino AE, Hsu PD, Heiden-
Jiang W, Bikard D, Cox D, Zhang F, Marraffini LA. 2013. reich M, Cong L, Platt RJ, Scott DA, Church GM, Zhang F.
RNA-guided editing of bacterial genomes using CRISPR- 2013. Optical control of mammalian endogenous tran-
scription and epigenetic states. Nature 500: 472– 476.
Cas systems. Nat Biotechnol 31: 233 –239.
Konermann S, Brigham MD, Trevino AE, Joung J, Abu-
Jiang F, Taylor DW, Chen JS, Kornfeld JE, Zhou K, Thomp-
dayyeh OO, Barcena C, Hsu PD, Habib N, Gootenberg
son AJ, Nogales E, Doudna JA. 2016. Structures of a
JS, Nishimasu H, et al. 2015. Genome-scale transcrip-
CRISPR-Cas9 R-loop complex primed for DNA cleavage.
tional activation by an engineered CRISPR-Cas9 com-
Science 351: 867– 871.
plex. Nature 517: 583 –588.
Jinek M, Chylinski K, Fonfara I, Hauer M, Doudna JA,
Korkmaz G, Lopes R, Ugalde AP, Nevedomskaya E, Han R,
Charpentier E. 2012. A programmable dual-RNA-guided
Myacheva K, Zwart W, Elkon R, Agami R. 2016. Func-
DNA endonuclease in adaptive bacterial immunity. Sci-
tional genetic screens for enhancer elements in the hu-
ence 337: 816– 821. man genome using CRISPR-Cas9. Nat Biotechnol 34:
Jinek M, East A, Cheng A, Lin S, Ma E, Doudna J. 2013. 192 –198.
RNA-programmed genome editing in human cells. eLife Lebar T, Bezeljak U, Golob A, Jerala M, Kadunc L, Pirs B,
2: e00471. Strazar M, Vucko D, Zupancic U, Bencina M, et al. 2014.
Juillerat A, Pessereau C, Dubois G, Guyot V, Marechal A, A bistable genetic switch based on designable DNA-bind-
Valton J, Daboussi F, Poirot L, Duclert A, Duchateau P. ing domains. Nat Commun 5: 5007.
2015. Optimized tuning of TALEN specificity using non- Lee HJ, Kim E, Kim JS. 2010. Targeted chromosomal dele-
conventional RVDs. Sci Rep 5: 8150. tions in human cells using zinc finger nucleases. Genome
Kiani S, Chavez A, Tuttle M, Hall RN, Chari R, Ter-Ova- Res 20: 81–89.
nesyan D, Qian J, Pruitt BW, Beal J, Vora S, et al. 2015. Li H, Haurigot V, Doyon Y, Li T, Wong SY, Bhagwat AS,
Cas9 gRNA engineering for genome editing, activation Malani N, Anguela XM, Sharma R, Ivanciu L, et al.
and repression. Nat Methods 12: 1051–1054. 2011. In vivo genome editing restores haemostasis in a
Kim YG, Cha J, Chandrasegaran S. 1996. Hybrid restriction mouse model of haemophilia. Nature 475: 217–221.
enzymes: Zinc finger fusions to Fok I cleavage domain. Li Y, Moore R, Guinn M, Bleris L. 2012. Transcription acti-
Proc Natl Acad Sci 93: 1156–1160. vator-like effector hybrids for conditional control and
Kim HJ, Lee HJ, Kim H, Cho SW, Kim JS. 2009. Targeted rewiring of chromosomal transgene expression. Sci Rep
genome editing in human cells with zinc finger nucleases 2: 897.
constructed via modular assembly. Genome Res 19: Li W, Teng F, Li T, Zhou Q. 2013. Simultaneous generation
1279– 1288. and germline transmission of multiple gene mutations
Kim E, Kim S, Kim DH, Choi BS, Choi IY, Kim JS. 2012. in rat using CRISPR-Cas systems. Nat Biotechnol 31:
Precision genome engineering with programmable 684 –686.
DNA-nicking enzymes. Genome Res 22: 1327– 1333. Limsirichai P, Gaj T, Schaffer DV. 2016. CRISPR-mediated
Kim Y, Kweon J, Kim A, Chon JK, Yoo JY, Kim HJ, Kim S, Lee activation of latent HIV-1 expression. Mol Ther 24: 499–
C, Jeong E, Chung E, et al. 2013a. A library of TAL effector 507.
nucleases spanning the human genome. Nat Biotechnol Lin SR, Yang HC, Kuo YT, Liu CJ, Yang TY, Sung KC, Lin YY,
31: 251 –258. Wang HY, Wang CC, Shen YC, et al. 2014. The CRISPR/
T. Gaj et al.
Cas9 system facilitates clearance of the intrahepatic HBV human gene expression using TALE activators. Nat Meth-
templates in vivo. Mol Ther Nucleic Acids 3: e186. ods 10: 243 –245.
Lin J, Chen H, Luo L, Lai Y, Xie W, Kee K. 2015a. Creating a Maggio I, Stefanucci L, Janssen JM, Liu J, Chen X, Mouly V,
monomeric endonuclease TALE-I-SceI with high specif- Goncalves MA. 2016. Selection-free gene repair after ad-
icity and low genotoxicity in human cells. Nucleic Acids enoviral vector transduction of designer nucleases: rescue
Res 43: 1112–1122. of dystrophin synthesis in DMD muscle cell populations.
Lin S, Ewen-Campen B, Ni X, Housden BE, Perrimon N. Nucleic Acids Res 44: 1449–1470.
2015b. In vivo transcriptional activation using CRISPR/ Magnenat L, Blancafort P, Barbas CF III. 2004. In vivo se-
Cas9 in Drosophila. Genetics 201: 433 –442. lection of combinatorial libraries and designed affinity
Liu Q, Segal DJ, Ghiara JB, Barbas CF III. 1997. Design of maturation of polydactyl zinc finger transcription factors
polydactyl zinc-finger proteins for unique addressing for ICAM-1 provides new insights into gene regulation. J
within complex genomes. Proc Natl Acad Sci 94: 5525– Mol Biol 341: 635–649.
5530. Magnenat L, Schwimmer LJ, Barbas CF III. 2008. Drug-
Liu J, Gaj T, Patterson JT, Sirk SJ, Barbas CF III. 2014a. Cell- inducible and simultaneous regulation of endogenous
penetrating peptide-mediated delivery of TALEN pro- genes by single-chain nuclear receptor-based zinc-finger
teins via bioconjugation for genome engineering. PLoS transcription factor gene switches. Gene Ther 15: 1223–
ONE 9: e85755. 1232.
Liu Y, Zeng Y, Liu L, Zhuang C, Fu X, Huang W, Cai Z. Mahiny AJ, Dewerth A, Mays LE, Alkhaled M, Mothes B,
2014b. Synthesizing AND gate genetic circuits based on Malaeksefat E, Loretz B, Rottenberger J, Brosch DM, Re-
CRISPR-Cas9 for identification of bladder cancer cells. autschnig P, et al. 2015. In vivo genome editing using
Nat Commun 5: 5393. nuclease-encoding mRNA corrects SP-B deficiency. Nat
Liu Z, Zhou X, Zhu Y, Chen ZF, Yu B, Wang Y, Zhang CC, Nie Biotechnol 33: 584– 586.
YH, Sang X, Cai YJ, et al. 2014c. Generation of a monkey Mak AN, Bradley P, Cernadas RA, Bogdanove AJ, Stoddard
with MECP2 mutations by TALEN-based gene targeting. BL. 2012. The crystal structure of TAL effector PthXo1
Neurosci Bull 30: 381–386. bound to its DNA target. Science 335: 716 –719.
Liu J, Gaj T, Wallen MC, Barbas CF III. 2015a. Improved cell- Mali P, Aach J, Stranges PB, Esvelt KM, Moosburner M,
penetrating zinc-finger nuclease proteins for precision Kosuri S, Yang L, Church GM. 2013a. CAS9 transcrip-
genome engineering. Mol Ther Nucleic Acids 4: e232. tional activators for target specificity screening and
Liu J, Gaj T, Yang Y, Wang N, Shui S, Kim S, Kanchiswamy paired nickases for cooperative genome engineering.
CN, Kim JS, Barbas CF III. 2015b. Efficient delivery of Nat Biotechnol 31: 833 –838.
nuclease proteins for genome editing in human stem cells Mali P, Yang L, Esvelt KM, Aach J, Guell M, DiCarlo JE,
and primary cells. Nat Protoc 10: 1842–1859. Norville JE, Church GM. 2013b. RNA-guided human
Long C, Amoasii L, Mireault AA, McAnally JR, Li H, San- genome engineering via Cas9. Science 339: 823 –826.
chez-Ortiz E, Bhattacharyya S, Shelton JM, Bassel-Duby Margolin JF, Friedman JR, Meyer WK, Vissing H, Thiesen
R, Olson EN. 2015. Postnatal genome editing partially HJ, Rauscher FJ III. 1994. Kruppel-associated boxes are
restores dystrophin expression in a mouse model of mus- potent transcriptional repression domains. Proc Natl
cular dystrophy. Science 351: 400 –403. Acad Sci 91: 4509–4513.
Lonzaric J, Lebar T, Majerle A, Mancek-Keber M, Jerala R. Marraffini LA, Sontheimer EJ. 2010. CRISPR interference:
2016. Locked and proteolysis-based transcription activa- RNA-directed adaptive immunity in bacteria and ar-
tor-like effector (TALE) regulation. Nucleic Acids Res 44: chaea. Nat Rev Genet 11: 181–190.
1471– 1481. McConnell Smith A, Takeuchi R, Pellenz S, Davis L, Maizels
Luo S, Li J, Stoddard TJ, Baltes NJ, Demorest ZL, Clasen BM, N, Monnat RJ Jr, Stoddard BL. 2009. Generation of a
Coffman A, Retterath A, Mathis L, Voytas DF, et al. 2015. nicking enzyme that stimulates site-specific gene conver-
Non-transgenic plant genome editing using purified se- sion from the I-AniI LAGLIDADG homing endonucle-
quence-specific nucleases. Mol Plant 8: 1425–1427. ase. Proc Natl Acad Sci 106: 5099–5104.
Ma H, Dang Y, Wu Y, Jia G, Anaya E, Zhang J, Abraham S, Meckler JF, Bhakta MS, Kim MS, Ovadia R, Habrian CH,
Choi JG, Shi G, Qi L, et al. 2015. A CRISPR-based screen Zykovich A, Yu A, Lockwood SH, Morbitzer R, Elsaesser
identifies genes essential for West-Nile-virus-induced cell J, et al. 2013. Quantitative analysis of TALE-DNA inter-
death. Cell Rep 12: 673– 683. actions suggests polarity effects. Nucleic Acids Res 41:
Maeder ML, Gersbach CA. 2016. Genome-editing technol- 4118–4128.
ogies for gene and cell therapy. Mol Ther 24: 430 –446. Mendenhall EM, Williamson KE, Reyon D, Zou JY, Ram O,
Maeder ML, Thibodeau-Beganny S, Osiak A, Wright DA, Joung JK, Bernstein BE. 2013. Locus-specific editing
Anthony RM, Eichtinger M, Jiang T, Foley JE, Winfrey RJ, of histone modifications at endogenous enhancers. Nat
Townsend JA, et al. 2008. Rapid “open-source” engineer- Biotechnol 31: 1133–1136.
ing of customized zinc-finger nucleases for highly effi- Menoret S, De Cian A, Tesson L, Remy S, Usal C, Boule JB,
cient gene modification. Mol Cell 31: 294–301. Boix C, Fontaniere S, Creneguy A, Nguyen TH, et al.
Maeder ML, Linder SJ, Cascio VM, Fu Y, Ho QH, Joung JK. 2015. Homology-directed repair in rodent zygotes using
2013a. CRISPR RNA-guided activation of endogenous Cas9 and TALEN engineered proteins. Sci Rep 5: 14410.
human genes. Nat Methods 10: 977– 979. Mercer AC, Gaj T, Fuller RP, Barbas CF 3rd. 2012. Chimeric
Maeder ML, Linder SJ, Reyon D, Angstman JF, Fu Y, Sander TALE recombinases with programmable DNA sequence
JD, Joung JK. 2013b. Robust, synergistic regulation of specificity. Nucleic Acids Res 40: 11163–11172.
Genome-Editing Technologies
Mercer AC, Gaj T, Sirk SJ, Lamb BM, Barbas CF III. 2014. rhabditis elegans using CRISPR-Cas9 ribonucleoprotein
Regulation of endogenous human gene expression by complexes. Genetics 201: 47– 54.
ligand-inducible TALE transcription factors. ACS Synth Papworth M, Moore M, Isalan M, Minczuk M, Choo Y, Klug
Biol 3: 723– 730. A. 2003. Inhibition of herpes simplex virus 1 gene expres-
Miller JC, Holmes MC, Wang J, Guschin DY, Lee YL, Rup- sion by designer zinc-finger transcription factors. Proc
niewski I, Beausejour CM, Waite AJ, Wang NS, Kim KA, Natl Acad Sci 100: 1621–1626.
et al. 2007. An improved zinc-finger nuclease architecture Park KS, Lee DK, Lee H, Lee Y, Jang YS, Kim YH, Yang HY,
for highly specific genome editing. Nat Biotechnol 25: Lee SI, Seol W, Kim JS. 2003. Phenotypic alteration of
778–785. eukaryotic cells using randomized libraries of artificial
Miller JC, Tan S, Qiao G, Barlow KA, Wang J, Xia DF, Meng transcription factors. Nat Biotechnol 21: 1208–1214.
X, Paschon DE, Leung E, Hinkley SJ, et al. 2011. A TALE Parnas O, Jovanovic M, Eisenhaure TM, Herbst RH, Dixit A,
nuclease architecture for efficient genome editing. Nat Ye CJ, Przybylski D, Platt RJ, Tirosh I, Sanjana NE, et al.
Biotechnol 29: 143 –148. 2015. A genome-wide CRISPR screen in primary im-
Miller JC, Zhang L, Xia DF, Campo JJ, Ankoudinova IV, mune cells to dissect regulatory networks. Cell 162:
Guschin DY, Babiarz JE, Meng X, Hinkley SJ, Lam SC, 675 –686.
et al. 2015. Improved specificity of TALE-based genome Pattanayak V, Ramirez CL, Joung JK, Liu DR. 2011. Reveal-
editing using an expanded RVD repertoire. Nat Methods ing off-target cleavage specificities of zinc-finger nucle-
12: 465 –471. ases by in vitro selection. Nat Methods 8: 765– 770.
Mock U, Machowicz R, Hauber I, Horn S, Abramowski P, Pavletich NP, Pabo CO. 1991. Zinc finger-DNA recognition:
Berdien B, Hauber J, Fehse B. 2015. mRNA transfection Crystal structure of a Zif268-DNA complex at 2.1 A. Sci-
of a novel TAL effector nuclease (TALEN) facilitates effi- ence 252: 809 –817.
cient knockout of HIV co-receptor CCR5. Nucleic Acids Perdigao P, Gaj T, Santa-Marta M, Barbas CF III, Goncalves
Res 43: 5560–5571. J. 2016. Reactivation of latent HIV-1 expression by engi-
Moehle EA, Rock JM, Lee YL, Jouvenot Y, DeKelver RC, neered TALE transcription factors. PLoS ONE 11:
Gregory PD, Urnov FD, Holmes MC. 2007. Targeted e0150037.
gene addition into a specified location in the human Perez EE, Wang J, Miller JC, Jouvenot Y, Kim KA, Liu O,
genome using designed zinc finger nucleases. Proc Natl Wang N, Lee G, Bartsevich VV, Lee YL, et al. 2008. Es-
Acad Sci 104: 3055–3060. tablishment of HIV-1 resistance in CD4þ T cells by ge-
Moscou MJ, Bogdanove AJ. 2009. A simple cipher governs nome editing using zinc-finger nucleases. Nat Biotechnol
DNA recognition by TAL effectors. Science 326: 1501. 26: 808– 816.
Mussolino C, Alzubi J, Fine EJ, Morbitzer R, Cradick TJ, Perez-Pinera P, Ousterout DG, Brunger JM, Farin AM, Glass
Lahaye T, Bao G, Cathomen T. 2014. TALENs facilitate KA, Guilak F, Crawford GE, Hartemink AJ, Gersbach CA.
targeted genome editing in human cells with high spe- 2013. Synergistic and tunable human gene activation by
cificity and low cytotoxicity. Nucleic Acids Res 42: 6762– combinations of synthetic transcription factors. Nat
6773. Methods 10: 239– 242.
Nelson CE, Hakim CH, Ousterout DG, Thakore PI, Moreb Piatek A, Ali Z, Baazim H, Li L, Abulfaraj A, Al-Shareef S,
EA, Rivera RMC, Madhavan S, Pan X, Ran FA, Yan WX, et Aouida M, Mahfouz MM. 2015. RNA-guided transcrip-
al. 2015. In vivo genome editing improves muscle func- tional regulation in planta via synthetic dCas9-based
tion in a mouse model of Duchenne muscular dystrophy. transcription factors. Plant Biotechnol J 13: 578 –589.
Science 351: 403– 407. Pollock R, Giel M, Linher K, Clackson T. 2002. Regulation of
Nielsen AA, Voigt CA. 2014. Multi-input CRISPR/Cas ge- endogenous gene expression with a small-molecule di-
netic circuits that interface host regulatory networks. Mol merizer. Nat Biotechnol 20: 729– 733.
Syst Biol 10: 763. Polstein LR, Gersbach CA. 2012. Light-inducible spatiotem-
Nihongaki Y, Yamamoto S, Kawano F, Suzuki H, Sato M. poral control of gene activation by customizable zinc
2015. CRISPR-Cas9-based photoactivatable transcrip- finger transcription factors. J Am Chem Soc 134:
tion system. Chem Biol 22: 169 –174. 16480– 16483.
Nishimasu H, Ran FA, Hsu PD, Konermann S, Shehata SI, Polstein LR, Gersbach CA. 2015. A light-inducible CRISPR-
Dohmae N, Ishitani R, Zhang F, Nureki O. 2014. Crystal Cas9 system for control of endogenous gene activation.
structure of Cas9 in complex with guide RNA and target Nat Chem Biol 11: 198–200.
DNA. Cell 156: 935–949. Porteus MH. 2015. Towards a new era in medicine: Thera-
Nissim L, Perli SD, Fridkin A, Perez-Pinera P, Lu TK. 2014. peutic genome editing. Genome Biol 16: 286.
Multiplexed and programmable regulation of gene net- Porteus MH, Baltimore D. 2003. Chimeric nucleases stim-
works with an integrated RNA and CRISPR/Cas toolkit ulate gene targeting in human cells. Science 300: 763.
in human cells. Mol Cell 54: 698–710. Porteus MH, Carroll D. 2005. Gene targeting using zinc
Osborn MJ, Webber BR, Knipping F, Lonetree CL, Tennis N, finger nucleases. Nat Biotechnol 23: 967– 973.
DeFeo AP, McElroy AN, Starker CG, Lee C, Merkel S, et Pruett-Miller SM, Reading DW, Porter SN, Porteus MH.
al. 2016. Evaluation of TCR gene editing achieved by 2009. Attenuation of zinc finger nuclease toxicity by
TALENs, CRISPR/Cas9, and megaTAL nucleases. Mol small-molecule regulation of protein levels. PLoS Genet
Ther 24: 570 –581. 5: e1000376.
Paix A, Folkmann A, Rasoloson D, Seydoux G. 2015. High Qi LS, Larson MH, Gilbert LA, Doudna JA, Weissman JS,
efficiency, homology-directed genome editing in Caeno- Arkin AP, Lim WA. 2013. Repurposing CRISPR as an
T. Gaj et al.
RNA-guided platform for sequence-specific control of Santiago Y, Chan E, Liu PQ, Orlando S, Zhang L, Urnov FD,
gene expression. Cell 152: 1173– 1183. Holmes MC, Guschin D, Waite A, Miller JC, et al. 2008.
Rahdar M, McMahon MA, Prakash TP, Swayze EE, Bennett Targeted gene knockout in mammalian cells by using
CF, Cleveland DW. 2015. Synthetic CRISPR RNA-Cas9- engineered zinc-finger nucleases. Proc Natl Acad Sci
guided genome editing in human cells. Proc Natl Acad Sci 105: 5809– 5814.
112: E7110– E7117. Sather BD, Romano Ibarra GS, Sommer K, Curinga G, Hale
Ramirez CL, Certo MT, Mussolino C, Goodwin MJ, Cradick M, Khan IF, Singh S, Song Y, Gwiazda K, Sahni J, et al.
TJ, McCaffrey AP, Cathomen T, Scharenberg AM, Joung 2015. Efficient modification of CCR5 in primary human
JK. 2012. Engineered zinc finger nickases induce homol- hematopoietic cells using a megaTAL nuclease and AAV
ogy-directed repair with reduced mutagenic effects. Nu- donor template. Sci Transl Med 7: 307ra156.
cleic Acids Res 40: 5560– 5568. Schmid-Burgk JL, Schmidt T, Kaiser V, Honing K, Hornung
Ran FA, Hsu PD, Lin CY, Gootenberg JS, Konermann S, V. 2013. A ligation-independent cloning technique for
Trevino AE, Scott DA, Inoue A, Matoba S, Zhang Y, et high-throughput assembly of transcription activator-
al. 2013. Double nicking by RNA-guided CRISPR Cas9 like effector genes. Nat Biotechnol 31: 76– 81.
for enhanced genome editing specificity. Cell 154: 1380– Schumann K, Lin S, Boyer E, Simeonov DR, Subramaniam
1389. M, Gate RE, Haliburton GE, Ye CJ, Bluestone JA, Doudna
JA, et al. 2015. Generation of knock-in primary human T
Ran FA, Cong L, Yan WX, Scott DA, Gootenberg JS, Kriz AJ,
cells using Cas9 ribonucleoproteins. Proc Natl Acad Sci
Zetsche B, Shalem O, Wu X, Makarova KS, et al. 2015. In
112: 10437–10442.
vivo genome editing using Staphylococcus aureus Cas9.
Nature 520: 186–191. Segal DJ, Dreier B, Beerli RR, Barbas CF III. 1999. Toward
controlling gene expression at will: Selection and design
Rebar EJ, Huang Y, Hickey R, Nath AK, Meoli D, Nath S,
of zinc finger domains recognizing each of the 50 -GNN-30
Chen B, Xu L, Liang Y, Jamieson AC, et al. 2002. Induc- DNA target sequences. Proc Natl Acad Sci 96: 2758–2763.
tion of angiogenesis in a mouse model using engineered
transcription factors. Nat Med 8: 1427–1432. Segal DJ, Goncalves J, Eberhardy S, Swan CH, Torbett BE, Li
X, Barbas CF III. 2004. Attenuation of HIV-1 replication
Reynolds L, Ullman C, Moore M, Isalan M, West MJ, Clap- in primary human cells with a designed zinc finger tran-
ham P, Klug A, Choo Y. 2003. Repression of the HIV-1 50 scription factor. J Biol Chem 279: 14509–14519.
LTR promoter and inhibition of HIV-1 replication by
Shah AN, Davey CF, Whitebirch AC, Miller AC, Moens CB.
using engineered zinc-finger transcription factors. Proc
2015. Rapid reverse genetic screening using CRISPR in
Natl Acad Sci 100: 1615– 1620.
zebrafish. Nat Methods 12: 535 –540.
Reyon D, Tsai SQ, Khayter C, Foden JA, Sander JD, Joung JK.
Shalem O, Sanjana NE, Hartenian E, Shi X, Scott DA, Mik-
2012. FLASH assembly of TALENs for high-throughput
kelsen TS, Heckl D, Ebert BL, Root DE, Doench JG, et al.
genome editing. Nat Biotechnol 30: 460 –465.
2014. Genome-scale CRISPR-Cas9 knockout screening
Rivenbark AG, Stolzenburg S, Beltran AS, Yuan X, Rots MG, in human cells. Science 343: 84–87.
Strahl BD, Blancafort P. 2012. Epigenetic reprogramming
Sharma R, Anguela XM, Doyon Y, Wechsler T, DeKelver RC,
of cancer cells via targeted DNA methylation. Epigenetics Sproul S, Paschon DE, Miller JC, Davidson RJ, Shivak D,
7: 350 –360. et al. 2015. In vivo genome editing of the albumin locus as
Rouet P, Smih F, Jasin M. 1994. Introduction of double- a platform for protein replacement therapy. Blood 126:
strand breaks into the genome of mouse cells by expres- 1777–1784.
sion of a rare-cutting endonuclease. Mol Cell Biol 14: Siddique AN, Nunna S, Rajavelu A, Zhang Y, Jurkowska RZ,
8096– 8106. Reinhardt R, Rots MG, Ragozin S, Jurkowski TP, Jeltsch
Ryan OW, Skerker JM, Maurer MJ, Li X, Tsai JC, Poddar S, A. 2013. Targeted methylation and gene silencing of
Lee ME, DeLoache W, Dueber JE, Arkin AP, et al. 2014. VEGF-A in human cells by using a designed Dnmt3a-
Selection of chromosomal DNA libraries using a multi- Dnmt3L single-chain fusion protein with increased
plex CRISPR system. eLife doi: 10.7554/eLife.03703. DNA methylation activity. J Mol Biol 425: 479– 491.
Saayman SM, Lazar DC, Scott TA, Hart JR, Takahashi M, Silva G, Poirot L, Galetto R, Smith J, Montoya G, Duchateau
Burnett JC, Planelles V, Morris KV, Weinberg MS. 2016. P, Paques F. 2011. Meganucleases and other tools for tar-
Potent and targeted activation of latent HIV-1 using the geted genome engineering: perspectives and challenges
CRISPR/dCas9 activator complex. Mol Ther 24: 488 – for gene therapy. Curr Gene Ther 11: 11– 27.
498. Sinkins SP, Gould F. 2006. Gene drive systems for insect
Sadowski I, Ma J, Triezenberg S, Ptashne M. 1988. GAL4- disease vectors. Nat Rev Genet 7: 427 –435.
VP16 is an unusually potent transcriptional activator. Sirk SJ, Gaj T, Jonsson A, Mercer AC, Barbas CF III. 2014.
Nature 335: 563–564. Expanding the zinc-finger recombinase repertoire: Di-
Sander JD, Cade L, Khayter C, Reyon D, Peterson RT, Joung rected evolution and mutational analysis of serine recom-
JK, Yeh JR. 2011a. Targeted gene disruption in somatic binase specificity determinants. Nucleic Acids Res 42:
zebrafish cells using engineered TALENs. Nat Biotechnol 4755–4766.
29: 697 –698. Slaymaker IM, Gao L, Zetsche B, Scott DA, Yan WX, Zhang
Sander JD, Dahlborg EJ, Goodwin MJ, Cade L, Zhang F, F. 2016. Rationally engineered Cas9 nucleases with im-
Cifuentes D, Curtin SJ, Blackburn JS, Thibodeau-Be- proved specificity. Science 351: 84–88.
ganny S, Qi Y, et al. 2011b. Selection-free zinc-finger- Smith J, Bibikova M, Whitby FG, Reddy AR, Chandrase-
nuclease engineering by context-dependent assembly garan S, Carroll D. 2000. Requirements for double-strand
(CoDA). Nat Methods 8: 67–69. cleavage by chimeric restriction enzymes with zinc finger
Genome-Editing Technologies
DNA-recognition domains. Nucleic Acids Res 28: 3361– human genome using a ZFN-based nicking enzyme. Ge-
3369. nome Res 22: 1316–1326.
Sorek R, Lawrence CM, Wiedenheft B. 2013. CRISPR-me- Wang H, Hu YC, Markoulaki S, Welstead GG, Cheng
diated adaptive immune systems in bacteria and archaea. AW, Shivalila CS, Pyntikova T, Dadon DB, Voytas DF,
Annu Rev Biochem 82: 237– 266. Bogdanove AJ, et al. 2013. TALEN-mediated editing
Stoddard BL. 2011. Homing endonucleases: From microbial of the mouse Y chromosome. Nat Biotechnol 31:
genetic invaders to reagents for targeted DNA modifica- 530 –532.
tion. Structure 19: 7 –15. Wang T, Wei JJ, Sabatini DM, Lander ES. 2014a. Genetic
Stoddard BL. 2014. Homing endonucleases from mobile screens in human cells using the CRISPR-Cas9 system.
group I introns: Discovery to genome engineering. Mob Science 343: 80–84.
DNA 5: 7. Wang Y, Cheng X, Shan Q, Zhang Y, Liu J, Gao C, Qiu JL.
Streubel J, Blucher C, Landgraf A, Boch J. 2012. TAL effector 2014b. Simultaneous editing of three homoeoalleles in
RVD specificities and efficiencies. Nat Biotechnol 30: hexaploid bread wheat confers heritable resistance to
593–595. powdery mildew. Nat Biotechnol 32: 947– 951.
Sung YH, Kim JM, Kim HT, Lee J, Jeon J, Jin Y, Choi JH, Ban Wang L, Shao Y, Guan Y, Li L, Wu L, Chen F, Liu M, Chen H,
YH, Ha SJ, Kim CH, et al. 2014. Highly efficient gene Ma Y, Ma X, et al. 2015a. Large genomic fragment dele-
knockout in mice and zebrafish with RNA-guided endo- tion and functional gene cassette knock-in via Cas9 pro-
tein mediated genome editing in one-cell rodent embry-
nucleases. Genome Res 24: 125–131.
os. Sci Rep 5: 17517.
Tabebordbar M, Zhu K, Cheng JKW, Chew WL, Widrick JJ,
Wang T, Birsoy K, Hughes NW, Krupczak KM, Post Y, Wei JJ,
Yan WX, Maesner C, Wu EY, Xiao R, Ran FA, et al. 2015.
Lander ES, Sabatini DM. 2015b. Identification and char-
In vivo gene editing in dystrophic mouse muscle and
acterization of essential genes in the human genome.
muscle stem cells. Science 351: 407– 411.
Science 350: 1096– 1101.
Tebas P, Stein D, Tang WW, Frank I, Wang SQ, Lee G, Spratt
Wiedenheft B, Sternberg SH, Doudna JA. 2012. RNA-guid-
SK, Surosky RT, Giedlin MA, Nichol G, et al. 2014. Gene
ed genetic silencing systems in bacteria and archaea. Na-
editing of CCR5 in autologous CD4 T cells of persons
ture 482: 331 –338.
infected with HIV. N Engl J Med 370: 901– 910.
Wilber A, Tschulena U, Hargrove PW, Kim YS, Persons DA,
Tesson L, Usal C, Menoret S, Leung E, Niles BJ, Remy S, Barbas CF III, Nienhuis AW. 2010. A zinc-finger tran-
Santiago Y, Vincent AI, Meng X, Zhang L, et al. 2011. scriptional activator designed to interact with the g-glo-
Knockout rats generated by embryo microinjection of bin gene promoters enhances fetal hemoglobin produc-
TALENs. Nat Biotechnol 29: 695–696. tion in primary human adult erythroblasts. Blood 115:
Thakore PI, D’Ippolito AM, Song L, Safi A, Shivakumar NK, 3033–3041.
Kabadi AM, Reddy TE, Crawford GE, Gersbach CA. Windbichler N, Menichelli M, Papathanos PA, Thyme SB, Li
2015. Highly specific epigenome editing by CRISPR- H, Ulge UY, Hovde BT, Baker D, Monnat RJ Jr, Burt A, et
Cas9 repressors for silencing of distal regulatory ele- al. 2011. A synthetic homing endonuclease-based gene
ments. Nat Methods 12: 1143–1149. drive system in the human malaria mosquito. Nature 473:
Tong Y, Charusanti P, Zhang L, Weber T, Lee SY. 2015. 212 –215.
CRISPR-Cas9 based engineering of actinomycetal ge- Wolfe SA, Nekludova L, Pabo CO. 2000. DNA recognition by
nomes. ACS Synth Biol 4: 1020–1029. Cys2His2 zinc finger proteins. Annu Rev Biophys Biomol
Torres R, Martin MC, Garcia A, Cigudosa JC, Ramirez JC, Struct 29: 183–212.
Rodriguez-Perales S. 2014. Engineering human tumour- Woo JW, Kim J, Kwon SI, Corvalan C, Cho SW, Kim H, Kim
associated chromosomal translocations with the RNA- SG, Kim ST, Choe S, Kim JS. 2015. DNA-free genome
guided CRISPR-Cas9 system. Nat Commun 5: 3964. editing in plants with preassembled CRISPR-Cas9 ribo-
Tsai SQ, Wyvekens N, Khayter C, Foden JA, Thapar V, Reyon nucleoproteins. Nat Biotechnol 33: 1162–1164.
D, Goodwin MJ, Aryee MJ, Joung JK. 2014. Dimeric Wu H, Yang WP, Barbas CF III. 1995. Building zinc fingers
CRISPR RNA-guided FokI nucleases for highly specific by selection: Toward a therapeutic application. Proc Natl
genome editing. Nat Biotechnol 32: 569 –576. Acad Sci 92: 344– 348.
Urnov FD, Miller JC, Lee YL, Beausejour CM, Rock JM, Wu Y, Liang D, Wang Y, Bai M, Tang W, Bao S, Yan Z, Li D, Li
Augustus S, Jamieson AC, Porteus MH, Gregory PD, J. 2013. Correction of a genetic disease in mouse via use of
Holmes MC. 2005. Highly efficient endogenous human CRISPR-Cas9. Cell Stem Cell 13: 659 –662.
gene correction using designed zinc-finger nucleases. Na- Wu H, Wang Y, Zhang Y, Yang M, Lv J, Liu J, Zhang Y. 2015.
ture 435: 646–651. TALE nickase-mediated SP110 knockin endows cattle
Urnov FD, Rebar EJ, Holmes MC, Zhang HS, Gregory PD. with increased resistance to tuberculosis. Proc Natl Acad
2010. Genome editing with engineered zinc finger nucle- Sci 112: E1530–E1539.
ases. Nat Rev Genet 11: 636– 646. Xiao A, Wang Z, Hu Y, Wu Y, Luo Z, Yang Z, Zu Y, Li W,
Wallen MC, Gaj T, Barbas CF III. 2015. Redesigning recom- Huang P, Tong X, et al. 2013. Chromosomal deletions and
binase specificity for safe harbor sites in the human ge- inversions mediated by TALENs and CRISPR/Cas in ze-
nome. PLoS ONE 10: e0139123. brafish. Nucleic Acids Res 41: e141.
Wang J, Friedman G, Doyon Y, Wang NS, Li CJ, Miller JC, Yang J, Zhang Y, Yuan P, Zhou Y, Cai C, Ren Q, Wen D, Chu
Hua KL, Yan JJ, Babiarz JE, Gregory PD, et al. 2012. C, Qi H, Wei W. 2014. Complete decoding of TAL effec-
Targeted gene addition to a predetermined site in the tors for DNA recognition. Cell Res 24: 628 –631.
T. Gaj et al.
Yang Y, Wang L, Bell P, McMenamin D, He Z, White J, Yu H, endonuclease of a class 2 CRISPR-Cas system. Cell 163:
Xu C, Morizono H, Musunuru K, et al. 2016. A dual AAV 759 –771.
system enables the Cas9-mediated correction of a meta- Zetsche B, Volz SE, Zhang F. 2015b. A split-Cas9 architecture
bolic liver disease in newborn mice. Nat Biotechnol 34: for inducible genome editing and transcription modula-
334–338. tion. Nat Biotechnol 33: 139– 142.
Yin H, Xue W, Chen S, Bogorad RL, Benedetti E, Grompe M, Zhang F, Cong L, Lodato S, Kosuri S, Church GM, Arlotta P.
Koteliansky V, Sharp PA, Jacks T, Anderson DG. 2014. 2011. Efficient construction of sequence-specific TAL
Genome editing with Cas9 in adult mice corrects a dis- effectors for modulating mammalian transcription. Nat
ease mutation and phenotype. Nat Biotechnol 32: 551 – Biotechnol 29: 149– 153.
553. Zhang Y, Yin C, Zhang T, Li F, Yang W, Kaminski R, Fagan
Yin H, Song CQ, Dorkin JR, Zhu LJ, Li Y, Wu Q, Park A, PR, Putatunda R, Young WB, Khalili K, et al. 2015.
Yang J, Suresh S, Bizhanova A, et al. 2016. Therapeutic CRISPR/gRNA-directed synergistic activation mediator
genome editing by combined viral and non-viral delivery (SAM) induces specific, persistent and robust reactiva-
of CRISPR system components in vivo. Nat Biotechnol tion of the HIV-1 latent reservoirs. Sci Rep 5: 16277.
34: 328 –333. Zhou Y, Zhu S, Cai C, Yuan P, Li C, Huang Y, Wei W. 2014.
Zalatan JG, Lee ME, Almeida R, Gilbert LA, Whitehead EH, High-throughput screening of a CRISPR/Cas9 library
La Russa M, Tsai JC, Weissman JS, Dueber JE, Qi LS, et for functional genomics in human cells. Nature 509:
al. 2015. Engineering complex synthetic transcriptional 487 –491.
programs with CRISPR RNA scaffolds. Cell 160: 339– Zuris JA, Thompson DB, Shu Y, Guilinger JP, Bessen JL, Hu
350. JH, Maeder ML, Joung JK, Chen ZY, Liu DR. 2015. Cat-
Zetsche B, Gootenberg JS, Abudayyeh OO, Slaymaker IM, ionic lipid-mediated delivery of proteins enables efficient
Makarova KS, Essletzbichler P, Volz SE, Joung J, van der protein-based genome editing in vitro and in vivo. Nat
Oost J, Regev A, et al. 2015a. Cpf1 is a single RNA-guided Biotechnol 33: 73–80.
Cold Spring Harb Perspect Biol 2016; doi: 10.1101/cshperspect.a023754 originally published online
October 6, 2016
Minimal Cells−−Real and Imagined Synthetic DNA Synthesis and Assembly: Putting
John I. Glass, Chuck Merryman, Kim S. Wise, et al. the Synthetic in Synthetic Biology
Randall A. Hughes and Andrew D. Ellington
Synthetic Botany Design Automation in Synthetic Biology
Christian R. Boehm, Bernardo Pollak, Nuri Evan Appleton, Curtis Madsen, Nicholas Roehner,
Purswani, et al. et al.
Synthetic Biology in Cell and Organ Cell-Free Synthetic Biology: Engineering Beyond
Transplantation the Cell
Sean Stevens Jessica G. Perez, Jessica C. Stark and Michael C.
Jewett
Genome-Editing Technologies: Principles and The Need for Integrated Approaches in Metabolic
Applications Engineering
Thomas Gaj, Shannon J. Sirk, Sai-lan Shui, et al. Anna Lechner, Elizabeth Brunk and Jay D. Keasling
Alternative Watson−Crick Synthetic Genetic Synthetic Biology of Natural Products
Systems Rainer Breitling and Eriko Takano
Steven A. Benner, Nilesh B. Karalkar, Shuichi
Hoshika, et al.
Phage Therapy in the Era of Synthetic Biology At the Interface of Chemical and Biological
E. Magda Barbu, Kyle C. Cady and Bolyn Hubby Synthesis: An Expanded Genetic Code
Han Xiao and Peter G. Schultz
Synthetic Morphogenesis Building Spatial Synthetic Biology with
Brian P. Teague, Patrick Guye and Ron Weiss Compartments, Scaffolds, and Communities
Jessica K. Polka, Stephanie G. Hays and Pamela
A. Silver
Engineering Gene Circuits for Mammalian Cell−
Based Applications
Simon Ausländer and Martin Fussenegger
Copyright © 2016 Cold Spring Harbor Laboratory Press; all rights reserved