ArticlePDF AvailableLiterature Review

Review of CRISPR/Cas9 sgRNA Design Tools

Authors:

Abstract

The adaptive immunity system in bacteria and archaea, Clustered Regularly Interspaced Short Palindromic Repeats, CRISPR-associate (CRISPR/Cas), has been adapted as a powerful gene editing tool and got a broad application in genome research field due to its ease of use and cost-effectiveness. The performance of CRISPR/Cas relies on well-designed single-guide RNA (sgRNA), so a lot of bioinformatic tools have been developed to assist the design of highly active and specific sgRNA. These tools vary in design specifications, parameters, genomes and so on. To help researchers to choose their proper tools, we reviewed various sgRNA design tools, mainly focusing on their on-target efficiency prediction model and off-target detection algorithm.
Vol.:(0123456789)
1 3
Interdisciplinary Sciences: Computational Life Sciences
https://doi.org/10.1007/s12539-018-0298-z
REVIEW
Review ofCRISPR/Cas9 sgRNA Design Tools
YingboCui1 · JiamingXu1· MinxiaCheng2· XiangkeLiao1· ShaoliangPeng1,2,3
Received: 6 March 2018 / Revised: 2 April 2018 / Accepted: 4 April 2018
© Springer-Verlag GmbH Germany, part of Springer Nature 2018
Abstract
The adaptive immunity system in bacteria and archaea, Clustered Regularly Interspaced Short Palindromic Repeats, CRISPR-
associate (CRISPR/Cas), has been adapted as a powerful gene editing tool and got a broad application in genome research
field due to its ease of use and cost-effectiveness. The performance of CRISPR/Cas relies on well-designed single-guide
RNA (sgRNA), so a lot of bioinformatic tools have been developed to assist the design of highly active and specific sgRNA.
These tools vary in design specifications, parameters, genomes and so on. To help researchers to choose their proper tools,
we reviewed various sgRNA design tools, mainly focusing on their on-target efficiency prediction model and off-target
detection algorithm.
Keywords CRISPR· CRISPR/Cas9· SgRNA design· On-target efficiency· Off-target detection
1 Introduction
CRISPR/Cas is the adaptive immunity system in many
bacteria and most archaea [13], which provides immunity
against viruses and plasmids. In this system, the DNA of
invaded virus or plasmid will be cleaved into novel spacer
and stored in an array in DNA. When the same virus or plas-
mid invades again, the corresponding invading DNA will be
recognized and interfered [2].
In 2012, researchers ported the CRISPR/Cas system
to genome editing and explain the basic mechanism of
CRISPR/Cas9 [4, 5]. CRISPR/Cas9 consists of two parts:
sgRNA and Cas9 endonuclease. The two components form
a complex to cleave target DNA sites, as shown in Fig.1.
sgRNA is derived from a fusion of the tracrRNA (trans-
activating crRNA) and crRNA (CRISPR RNA) [2, 4].
From the engineering perspective, sgRNA consists of two
parts: constant part (purple in Fig.1), which forms a scaf-
fold by several stem-loop for Cas9 binding, and a 5-end
20-nt altered part (pink in Fig.1) complementary to target
DNA sequence, which is programmable to target different
DNA sites [6]. Cas9 is nuclease derived from Streptococcus
pyogenes (S. pyogenes) [7]. The target site in DNA mainly
contains two parts: protospacer (black in Fig.1) comple-
mentary to the 5-end 20-nt sequence in sgRNA and proto-
spacer adjacent motif (PAM, yellow in Fig.1) bound by
Cas9. PAM is usually short (for SpCas9 is 5-NGG-3) and
directly adjacent to protospacer. The Cas9 protein will not
cleave sequence without the presence of PAM.
If the protospacer pairs with the 5-end 20-nt sequence,
and the Cas9 binds with PAM, a double-strand break (DSB)
will be made. After DSB, DNA repair machinery will start
and catalyze non-homologous end joining (NHEJ) or homol-
ogy-directed repair (HDR), as shown in Fig.1. For NHEJ,
the lost sequence may cannot be recovered and results in
sequence insertions or deletions (indels), which produce
gene loss of function, while for HDR, an introduced exog-
enous DNA template will fill the gap from DSB [8].
Except for gene loss of function (CRISPRko) by making
a DSB, CRISPR/Cas9 system can also repress or boost the
expression of specific gene [911]. Gene repression (CRIS-
PRi) can be achieved by binding a catalytically dead Cas9
lacking cleaving activity to the transcription factor binding
sites [9], and gene activation (CRISPRa) can be realized by
Yingbo Cui and Jiaming Xu are equal contributors.
* Shaoliang Peng
pengshaoliang@nudt.edu.cn
1 College ofComputer, National University ofDefense
Technology, Changsha410073, China
2 College ofComputer Science andElectronic Engineering,
Hunan University, Changsha410082, China
3 National Supercomputing Center inChangsha,
Changsha410082, China
Interdisciplinary Sciences: Computational Life Sciences
1 3
the fusion of an inactive Cas9 to the transcriptional activa-
tion domain [10, 11].
Compared with previous gene editing technologies, zinc-
finger nucleases (ZFNs) [1218] and transcription activa-
tor-like effector nucleases (TALENs) [1922], which bind
to specific DNA sequence by protein-DNA recognition,
CRISPR/Cas9 system identifies specific site by the com-
plementarity between the 20-nt sequence at the 5-end of
sgRNA and the protospacer in DNA, which is easier and
cheaper to implemented in engineering. Besides, some
studies show that CRISPR/Cas9 is more efficient than
ZFNs and TALENs in gene editing [2325]. Due to these
advantages, it has been used in many species and mamma-
lian cells [2634]. In addition, some researchers have used
the CRISPR/Cas9 system for genome-wide genetic screens
invitro and invivo [3543].
Currently, the large amount of sgRNA design tools vary
in design specifications, parameters, genomes and so on [44,
45]. To help researchers choose the most suitable sgRNA
design tools for their experiments, we review current sgRNA
design tools, mainly focusing on the on-target efficiency pre-
diction models and off-target prediction algorithms.
2 Overview ofsgRNA Design Tools
The major work of sgRNA design is to find target sites in
genome, which is pretty easy to implement by scanning
PAM sequence (like 5-NGG-3 for SpCas9). However, sev-
eral challenges exist for designing a good sgRNA: efficacy
and specificity.
In theory, if the 5-end 20-nt sequence in sgRNA is com-
plementary to the target DNA sequence, the sgRNA-Cas9
complex should bind to the site and make a cleavage, but
in practice, some studies suggest that the cutting efficiency
varies significantly among sgRNAs [5, 26, 27, 33, 3537,
4660]. Predictive models to identify target sites with high
efficiency are necessary.
Another challenge is off-target activity of sgRNA (speci-
ficity). The off-target is caused by both sgRNA and Cas9.
A few mismatches between the 5-end 20-nt sequence in
sgRNA and target DNA sequence are tolerated [61]. Some
studies have shown that CRISPR/Cas9 non-specially cleave
DNA sites with several mismatches generating off-target
mutations with considerable frequencies [10, 47, 52, 6273].
The optimal PAM recognized by SpCas9 is 5-NGG-3.
However, SpCas9 also binds to 5-NAG-3 or 5-NGA-3
with low frequencies [10, 57, 62, 74]. It is essential to find
out potential off-target sites and improve sgRNA specificity.
To design sgRNA with high efficacy and specificity, many
models and computational tools have been developed. Some
representative ones are summarized in Table1.
2.1 On‑target Efficiency Prediction
The Root laboratory proposed a rule to predict on-target
efficacy of sgRNAs [75]. They analyzed all possible target
sites for six mouse genes and three human genes, includ-
ing 1841 sgRNAs. In their model, they employed a sup-
port vector machine (SVM) model [90] to choose subsets
with the best generalization error from 586 features [75].
The selected features by SVM were trained with a logis-
tic regression classifier [91] to generate a sgRNA on-target
Fig. 1 CRISPR/Cas9 genome
editing system
Interdisciplinary Sciences: Computational Life Sciences
1 3
Table 1 Representative tools for sgRNA design
Tool Genome specific Nuclease Input On-target predic-
tion
Off-target scoring Websites References
CRISPR.mit Yes (17) SpCas9 DNA sequence No Hsu etal., 2013
[62]
http://crisp r.mit.
edu/
[62]
sgRNA designer No SpCas9
SaCas9
DNA sequence
Transcript ID
Gene ID
Gene Symbol
Rule Set 2 [50] CFD [50]https ://porta
ls.broad insti
tute.org/gpp/
publi c/analy
sis-tools /sgrna
-desig n
[50, 75]
E-CRISP Yes (55) Cas9
Custom PAM
Gene ID
Gene Symbol
DNA sequence
Xu etal., 2015
[51]
Doench etal.,
2014 [75]
E-score [76]
S-score [76]http://www.e-
crisp .org/E-
CRISP /
[76]
CHOPCHOP Yes (111) 7 PAMs
Custom PAM
RefSeq
Gene ID
Genomic region
Rule Set 2 [50]
Xu etal., 2015
[51]
Chari etal., 2015
[56]
Moreno-Mateos
etal., 2015 [58]
Doench etal.,
2014 [75]
G20
Cong etal., 2013
[26]
Hsu etal., 2013
[62]
https ://chopc hop.
rc.fas.harva
rd.edu/
[77, 78]
CRISPRseek No No Software package Doench etal.,
2014 [75]
CFD [50]
Hsu etal., 2013
[62]
http://www.bioco
nduct or.org/
packa ges/relea
se/bioc/html/
CRISP Rseek
.html
(Local R Pack-
age)
[79]
Cas-OFFinder Yes (36) 16 PAMs crRNA sequences No Cas-OFFinder
[80]
http://www.rgeno
me.net/cas-offin
der/
[80]
CRISPRdirect Yes (16) Custom PAM DNA sequence
Genomic region
Accession num-
bers
No kmer + PAM [81]http://crisp r.dbcls
.jp/
[81]
CCTop Yes (65) 11 PAMs DNA sequence CRISPRater [55] Stemmer etal.,
2017 [80]
https ://crisp r.cos.
uni-heide lberg
.de/index .html
[68]
CRISPRscan Yes (13) SpCas9
LbCpf1
AsCpf1
DNA sequence
Gene ID
Gene symbol
Moreno-Mateos
etal., 2015 [58]
Cong etal., 2013
[26]
CFD [50]
Hsu etal., 2013
[62]
http://www.crisp
rscan .org/
[58]
PROTOSPACER No SpCas9 Gene ID
DNA Sequence
Genomic Region
Doench etal.,
2014 [75]
Hsu etal., 2013
[62]
http://www.proto
space r.com/
[82]
sgRNA Scorer
1.0
No SpCas9
StCas9
DNA Sequence Chari etal., 2015
[56]
No https ://crisp r.med.
harva rd.edu/
sgRNA Score
rV1/
[56]
WU-CRISPR Yes (2) SpCas9 DNA Sequence
Gene ID
Gene Symbol
Wong etal., 2015
[49]
Wong etal., 2015
[49]
http://crisp r.wustl
.edu
[49]
SSC No SpCas9 DNA Sequence Xu etal., 2015
[51]
No http://cistr ome.
org/SSC/
[51]
Interdisciplinary Sciences: Computational Life Sciences
1 3
efficacy prediction model. The efficiency score from this
model is in the range of 0–1, where the higher values mean
more effective. They implemented the prediction model in
their online tool, sgRNA Designer [75]. Several tools use
this model to predict sgRNA on-target efficiency, such as
E-CRISP [76], CHOPCHOP [77, 78], PROTOSPACER
[82], CLD [85], CRISPOR [89], and CRISPETa [92]. This
tool supports medium-level throughput in query and one
remarkable feature is that the model in this tool is based on
the experimental data, so it is suitable for simultaneously
generate several reliable guides. CRISPRpred, also resorts
to learning algorithm to improve efficiency in predicting
on-target activity and performs better than SVM model in
Doench etal. [75, 93].
In 2016, the Root laboratory improved their model and
proposed the Rule Set 2 [50]. Comparing with their previous
model, the Rule Set 2 incorporated several new features,
such as counts of position-independent nucleotide, location
of target site in the corresponding gene, and melting tem-
perature. Besides, the authors compared the performance
of several classification-based models in machine learning
and referred to the gradient-bossed regression trees in their
reinforced model. GuideScan [67], CHOPCHOP [77, 78],
CRISPOR [89], and Guide Picker [94] use the Rule Set 2 to
evaluate activity of sgRNA.
The Liu laboratory assessed the features affecting
sgRNA efficiency in CRISPR screen data [51]. They used
Elastic-Net [95] to construct the predictive model and
found some new features, mainly the preference for cyto-
sine for sgRNA. They applied their model in SSC [51],
CRISPR-FOCUS [53] and CRISPR-DO [60]. E-CRISP
[76], CHOPCHOP [77, 78], CLD [85], CRISPOR [89],
Table 1 (continued)
Tool Genome specific Nuclease Input On-target predic-
tion
Off-target scoring Websites References
CROP-IT Yes (2) SpCas9 Genomic Region No Singh etal., 2015
[69]
http://cheet
ah.bioch .virgi
nia.edu/AdliL
ab/CROP-IT/
homep age.html
[69]
CRISPR Multi-
Targeter
No SpCas9
Custom PAM
DNA Sequence
Gene Symbol
Gene ID/
Transcripts ID
RefSeq Sequence
Doench etal.
2014 [75]
GT-Scan [83]
Cas-OFFinder
[80]
http://www.multi
crisp r.net/
[84]
CLD No SpCas9 Gene ID
Genomic Region
Xu etal., 2015
[51]
Doench etal.
2014 [75]
Custom
Heigwer etal.,
2016 [68]
https ://githu
b.com/boutr
oslab /cld (Local
Perl Package)
[85]
CRISPOR Yes (258) 9 PAMs DNA Sequence
Genomic Region
Rule Set 2 [50]
Xu etal., 2015
[51]
Chari etal., 2015
[56]
Doench etal.,
2014 [75]
Housden etal.,
2015 [86]
Prox GC [87, 88]
CFD [50]
Hsu etal., 2013
[62]
http://crisp or.tefor
.net/
[89]
sgRNA Scorer
2.0
No 6 PAMs
Custom PAM
DNA Sequence Chari etal., 2015
[57]
No https ://crisp r.med.
harva rd.edu/
sgRNA Score
rV2/
[57]
GuideScan Yes (6) SpCas9
AsCf1
LbCpf1
Genomic Region
Gene Symbol
Rule Set 2 [50] CFD [50]http://www.guide
scan.com/
[67]
CASPER No Custom PAM DNA Sequence CASPER [52] CASPER [52]https ://githu
b.com/Trinh
Lab/CASPE R
(Local Python
Package)
[52]
Interdisciplinary Sciences: Computational Life Sciences
1 3
and pgRNAFinder [96] also adopt this model. Besides,
authors of SSC found the effect of sequence context on
sgRNA efficiency differs significantly between CRIS-
PRko and CRISPRa/i and further proposed a model to
predict efficiency of sgRNA in CRISPRa/i. Due to the use
of machine learning method, high efficiency of designed
candidate guides is remarkable [97].
As the same demand for targeting invivo, Moreno-
Mateos etal. discovered that guanine enrichment and ade-
nine depletion contribute to the stability, loading and activity
of sgRNA by an analysis of target sites of 128 genes, where
ten sgRNAs for each gene [58]. They used logistic regres-
sion model to select features associated with sgRNA activ-
ity, including features on mononucleotide and dinucleotide,
and they used a linear regression model for sgRNA activ-
ity prediction. The model is integrated in their online tool
CRISPRscan [58]. CHOPCHOP [77, 78] and CRISPOR [89]
also use this model to predict sgRNA activity. Nevertheless,
one factor should be noticed that this efficiency prediction
model is based on the data of zebrafish and applying this
model to other species will receive worse results compared
with other tools.
The Church laboratory developed an invivo methodol-
ogy to evaluate sgRNA activity across thousands of genes
[56]. The two libraries refer to target-site library and sgRNA
library, respectively. They built a SVM classifier model to
score the on-target activity of sgRNA sequences. They also
observed the preference for guanine in position 20, next to
the PAM in SpCas9 and St1Cas9 (Streptococcus thermophi-
lus). The model was implemented in their online tool sgRNA
Scorer. CHOPCHOP [77, 78] and CRISPOR [89] applied
this model for on-target efficiency prediction. Its improved
version sgRNA scorer 2.0 [57] established a more general-
ized model with a new combined SVM, which was proved
to be useful for predicting sgRNA activity for multiple Cas9
orthologs and Cpf1 protein. The SVM model implemented
in this tool highly improved on-target prediction power,
whereas the analysis procedure is slow and the results have
to be returned by email.
WU-CRISPR [49] identified some novel structural and
sequence features from Doenchs dataset [75], and built a
sgRNA potency prediction model with SVM. They used the
Chari dataset [56] to evaluate the model and got the best per-
formance compared with three other tools [51, 56, 75]. This
tool is recommended for its highly efficient sgRNA design
using machine learning method and ease of use.
The Huang laboratory used Elastic-Net algorithm [95]
in their model to predict sgRNA efficiency [59]. They bor-
rowed knowledge from oligonucleotide design in microarray
and found that including position-dependent features can
help the design of effective sgRNA. The authors further
found that the frequency of T and TT affects the efficiency
of sgRNA significantly.
Although some studies found guanine enrichment can
improve the stability and potency of sgRNA [35, 51, 58,
75], Malina etal. discovered that too many PAMs (5-NGG-
3) within the target DNA sites inhibit the CRISPR/Cas9
activity in vivo [54]. Some other studies suggested the pref-
erence for guanines rather than adenines for the stability of
sgRNA [35, 75]. Taking these into consideration, CASPER
[52] integrate the CRISPRscan [58] score, PAM penalty
score [54] and a score indicating guanines preference [35,
75] into its predictive model. Their tool also expanded the
applications in multi-targeting analysis and multi-population
analysis.
Labuhn etal. utilized the surrogate fluorescent reporter
knock-out assays to assess and analyze a dataset of 430
sgRNAs [55]. They found nine additional features affecting
sgRNA activity and generated a linear model-based discrete
system using R package lars [98]. Their model enables the
exclusion of impotent sgRNA, implemented in their online
tool CRISPRater.
Some other tools include more than one on-target effi-
ciency prediction models and provide more choices for
users, such as E-CRISP [76], CHOPCHOP [77, 78], CLD
[85], and CRISPOR [89]. The details of the corresponding
tools are shown in Table1.
2.2 Off‑target Detection
The Zhang laboratory investigated sgRNA target specificity
by evaluating more than 700 sgRNA variants and predicated
off-target loci which is induced by SpCas9 with indel muta-
tion levels > 100 [62]. They found that mismatch tolerance
between SpCas9 complex and DNA is influenced by the
number, position and distribution of mismatches. Based on
this observation, they proposed a penalty matrix (Table2)
to describe the effect of mismatch position. The penalty is
between 0 and 1 where higher value means bigger effect
on cleavage. Referring to this penalty matrix correspond-
ing to each position, each sgRNA can be assigned a score
according to its potential off-target sites, as an evidence to
choose appropriate sgRNAs. The authors also implemented
a web-based software tool to facilitate sgRNA design and
validation (http://crisp r.mit.edu). This method is widely used
for sgRNA specificity score calculation, such as CRISPR-
scan [58], CRISPR-DO [60], CHOPCHOP [77, 78], and
CRISPOR [89]. However, this tool is only suitable for short
sequences (23–500nt) and the analysis is very slow.
Stemmer etal. noticed the limitation of previous sgRNA
off-target detecting tools and developed an online sgRNA
finding tool, CCTop [68]. This tool provides more cus-
tom options for users to choose. A graphical visualization
interface makes it easier to identify the appropriate sgRNA
among the candidates. Many researchers use third party
sequence alignment tools such as BWA [99] and Bowtie
Interdisciplinary Sciences: Computational Life Sciences
1 3
[100] for target site searching [66, 76, 77]. In CCTop, the
candidate sgRNAs are all selected by Bowtie [100] and
ranked by their off-target potential. The off-target score of
each sgRNA is defined as follows:
In this equation,
dist
means the distance between this
off-target site and the closest exon,
scoreoff _target
is calcu-
lated by the relative position of mismatches in protospacer.
total_off _targets
means the total number of candidate off-
target sites. The authors validated the predicted results with
experiments and proved its effectiveness. This tool provides
many custom options which are both favorable for primary
users and advanced users. Additionally, the analysis speed in
simple mode is fast, whereas advanced mode is slow.
The Root laboratory analyzed off-target effects on pooled-
screening data [50], with all possible sgRNAs targeting the
coding regions of human CD33 cell line, regardless of PAM.
The author profiled many mismatch conditions between
sgRNA and DNA, including alternative PAMs, mismatches,
deletions and insertions. Using the experimental data, they
proposed an algorithm to rank potential off-targets, which
is called the Cutting Frequency Determination (CFD) score.
They compared CFD score to off-target effect measurements
of Hsu–Zhang [62]. and CCTop [68]. Throughout various
(1)
Score
=
off _target
log10(dist)+scoreoff _ta rget
total_off _targets total_off _target b±
b24ac
2a
.
different examinations, they concluded that CFD score per-
forms better and could be able to avoid high-frequency off-
target effects. Some other tools apply this scoring algorithm
in their workflows, such as CRISPRScan [58], GuideScan
[67], and CRISPOR [89]. Here a recent study indicates that
CFD score performs better than CCTop [68], MIT-Zhang
[62] and CROP-IT [69] score when analyzing less than four
mismatches [89].
Mendoza etal. found that the previous off-target scor-
ing method cannot quickly accommodate across organisms,
so they proposed a novel algorithm to assess the off-target
activity, named CASPER [52]. The output of this algorithm
consists of three factors: SH, ST, and SS. SH is a score which
is calculated by the experimental data of mismatch posi-
tion and types derived from the Hsu–Zhang matrix [62]
(Table2). ST is calculated by the distance of mismatch
from PAM. SS is a stepped score, associated with the posi-
tion of mismatch in spacer. Overall, SH is derived from the
experimental data and ST, SS are derived from the properties
associated with CRISPR ribonucleoprotein complexes. Fur-
thermore, they adapted their scoring algorithm to as Cpf1
endonucleases and exhibited that CASPER can still effec-
tively assess its off-target activity even without sufficient
experimental data.
There are also many other tools devised their own algo-
rithms to evaluate off-target potential of sgRNA, such
as WU-CRISPR [49], CROP-IT [69], CT-Finder [73],
E-CRISPR [76], CGAT [101], sgRNAcas9 [102], and
CRISTA [103]. Overall, the off-target detection and evalu-
ation algorithms in these tools also have instructive value
in sgRNA design. Actually, after a lot of experiments,
researchers found out that using single off-target model can-
not always be effective and reliable in sgRNA design [89].
Based on this consideration, many tools provide advanced
options for researchers to choose the off-target detection
model or show several scores calculated by widely used
models in their output. CHOPCHOP [77, 78], an online
tool for sgRNA design, provides off-target scoring method
including two models devised by Hsu etal. [62] and Cong
etal. [26], separately. CRISPOR [89] offers scoring models
by Hsu etal. [62] and Doench etal., 2016 [50] (i.e., CFD).
CRISPR-RT [104] aims at crRNA design in CRISPR-C2c2/
Cas13a system and provides a variety of custom options for
users. It returns the candidate crRNA target sites by search-
ing the reference transcriptome with user-input RNA/cDNA
sequence. Users can rank all results by transcript-target num-
bers, gene-target numbers, GC content or more properties
of target sites.
Table 2 Experimentally
determined penalty weight
matrix of mismatch position
[62]
Mismatch rela-
tive position
Penalty weight
1 0
2 0
3 0.014
4 0
5 0
6 0.395
7 0.317
8 0
9 0.389
10 0.079
11 0.445
12 0.508
13 0.613
14 0.851
15 0.732
16 0.828
17 0.615
18 0.804
19 0.685
20 0.583
Interdisciplinary Sciences: Computational Life Sciences
1 3
3 Other Considerations ofsgRNA Design
Throughout various studies in CRISPR system, research-
ers have noticed that many other factors also influence the
gene editing performance of CRISPR. Here we summarize
some of these factors as follows:
Truncated sgRNAs with 17- to 19-nt spacer are more
sensitive to mismatches than others with 20-nt length,
which can effectively reduce off-target mutations [46]. In
addition, the recent study has showed that shorter spacers
such as 17 to 18 nt have higher specificity but perform less
on-target effective than 19- or 20-nt spacers [51]. On the
contrary, longer sgRNAs are less effective than sgRNAs
with 20-nt spacers [58].
Zetsche etal. found that Cpf1 (CRISPR from Prevo-
tella and Francisella 1) protein might be more convenient
in gene editing than Cas9 system, because it only need
mature crRNAs for targeting, while CRISPR/Cas9 system
requires both tracrRNA and crRNA. T-enriched PAM and
staggered DSB at distal end of spacer are notable features
in Cpf1, bringing new options in experimentation [105].
Fonfara etal. have studied the Cas9 homologs and
identified specific PAM sequences and dual-RNA (i.e.,
tracrRNA-crRNA) in some Cas9 orthologous proteins (for
example, S. thermophilus, S. mutans, P. multocida and N.
meningitidis). They also verified the coevolution between
dual-RNA and Cas9 protein through experimentation.
Their study indicates that the Cas9 orthologs and associ-
ated PAM and dual-RNA can become candidate compo-
nents in CRISPR/Cas9 system [106].
Ran etal. devised a double nicking approach by a pair
of sgRNA-guided Cas9n (Cas9 D10A) nickases, which can
effectively reduce the off-target effects. In this gene editing
approach, two modified effectors will bind to two opposite
strands and surround 10 to 31 nt target sequence and then
make a DSB [107]. Some recent studies have applied this
technique in their research [108, 109].
Tsai etal. described a dimeric architecture in CRISPR
system called dimeric RNA-guided FokI nucleases (RFNs).
This kind of dimeric nickases implement cleavage by two
guide RNAs (gRNAs) binding to the opposite strands. This
dimeric architecture can promise any nucleotide at 5 end of
gRNA and relative to paired nickases, which makes steps
forward in improving gRNA specificity [110].
CRISPR gene editing system can be divided into two
categories, CRISPRko and CRISPRa/i. The DNA target
sites vary for these two categories. In CRISPRko, target
sites are usually protein-coding region and Cas9 nucle-
ases produce loss-of-function gene knockouts by inducing
DSB. In CRISPRa/i, sgRNA guides catalytically inactivate
Cas9 (dCas9) to effector domains to activate or repress
gene transcription without DNA cleavage [50, 51].
Canonical sgRNA including 20-nt sequence and PAM
has limited target sites in vivo, because T7 or SP6 promot-
ers in CRISPR/Cas9 system restrict GG or GA in the 5-end
of sgRNA sequence [58]. Experimental results showed
that there are two kinds of alternative truncated sequences
which can become efficient substitutes to replace the 20-nt
sequence, one is truncated sgRNA at 5 end by less than
2-nt and the other is the 20-nt sgRNA with one mismatch
in 5 end [58].
Some researchers investigated the epigenetics features,
and found that chromatin accessibility affects the binding of
dCas9 to DNA sites [111, 112]. Chari etal. also observed
this phenomenon [56]. Whereas Moreno-Mateos etal. did
not notice a strong effect of chromatin accessibility on
CRISPR/Cas9 activity [58].
Four consecutive T (i.e., TTTT) is a signal of termina-
tion for pol III promoter thus the presence of TTTT in target
DNA sequence sites cannot be adoptable. It is necessary to
avoid TTTT in target sites when using pol III promoter [81].
4 Conclusion
CRISPR/Cas9 technology has been used more and more
widely for genome editing. Its efficacy, specificity, easy to
use, cost-effectiveness and versatility, will boost this tech-
nique on more fronts. In this review, we examined various
aspects of sgRNA design tools, including activity prediction
models, and off-target detection algorithms. Almost all of
these models or algorithms depend on large-scale experi-
mental datasets and systematic analysis. Therefore, with
more CRISPR/Cas9 datasets available, more novel sgRNA
design tools will be developed to facilitate the research
community.
Acknowledgements We would like thank to Jingyu Amy Peng and
Chen-Hao Chen from Harvard T.H. Chan School of Public Health,
Shenglin Mei and Jian Ma from Tongji University for their discus-
sion about this work. This work was supported by National Key
R&D Program of China 2017YFB0202602, 2017YFC1311003,
2016YFC1302500, 2016YFB0200400, 2017YFB0202104; NSFC
Grants 61772543, U1435222, 61625202, 61272056; the Funds of State
Key Laboratory of Chemo/Biosensing and Chemometrics; the Funda-
mental Research Funds for the Central Universities; and Guangdong
Provincial Department of Science and Technology under grant No.
2016B090918122.
References
1. Koonin EV, Makarova KS (2009) CRISPR-Cas: an adaptive
immunity system in prokaryotes. F1000 Biology Reports 1:95.
https ://doi.org/10.3410/B1-95
2. Horvath P, Barrangou R (2010) CRISPR/Cas, the immune system
of bacteria and archaea. Science 327(5962):167–170. https ://doi.
org/10.1126/scien ce.11795 55
Interdisciplinary Sciences: Computational Life Sciences
1 3
3. Koonin EV, Makarova KS (2013) CRISPR-Cas: evolution of an
RNA-based adaptive immunity system in prokaryotes. RNA Biol
10(5):679–686. https ://doi.org/10.4161/rna.24022
4. Deltcheva E, Chylinski K, Sharma CM, Gonzales K, Chao Y,
Pirzada ZA, Eckert MR, Vogel J, Charpentier E (2011) CRISPR
RNA maturation by trans-encoded small RNA and host factor
RNase III. Nature 471(7340):602–607. https ://doi.org/10.1038/
natur e0988 6
5. Jinek M, Chylinski K, Fonfara I, Hauer M, Doudna JA, Char-
pentier E (2012) A programmable dual-RNA-guided DNA endo-
nuclease in adaptive bacterial immunity. Science 337:816–821.
https ://doi.org/10.1126/scien ce.12258 29
6. Cong L, Zhang F (2015) Genome engineering using CRISPR-
Cas9 system. Methods Mol Biol 1239:197–217. https ://doi.
org/10.1007/978-1-4939-1862-1_10
7. Heler R, Samai P, Modell JW, Weiner C, Goldberg GW, Bikard
D, Marraffini LA (2015) Cas9 specifies functional viral targets
during CRISPR-Cas adaptation. Nature 519(7542):199–202.
https ://doi.org/10.1038/natur e1424 5
8. Wyman C, Kanaar R (2006) DNA double-strand break repair:
all’s well that ends well. Annu Rev Genet 40(1):363–383. https
://doi.org/10.1146/annur ev.genet .40.11040 5.09045 1
9. Qi LS, Larson MH, Gilbert LA, Doudna JA, Weissman JS, Arkin
AP, Lim WA (2013) Repurposing CRISPR as an RNA-guided
platform for sequence-specific control of gene expression. Cell
152(5):1173–1183. https ://doi.org/10.1016/j.cell.2013.02.022
10. Mali P, Aach J, Stranges PB, Esvelt KM, Moosburner M, Kosuri
S, Yang L, Church GM (2013) CAS9 transcriptional activators
for target specificity screening and paired nickases for coopera-
tive genome engineering. Nat Biotechnol 31(9):833–838. https
://doi.org/10.1038/nbt.2675
11. Gilbert LA, Horlbeck MA, Adamson B, Villalta JE, Chen Y,
Whitehead EH, Guimaraes C, Panning B, Ploegh HL, Bassik
MC, Qi LS, Kampmann M, Weissman JS (2014) Genome-scale
CRISPR-mediated control of gene repression and activation. Cell
159(3):647–661. https ://doi.org/10.1016/j.cell.2014.09.029
12. Urnov FD, Miller JC, Lee YL, Beausejour CM, Rock JM, Augus-
tus S, Jamieson AC, Porteus MH, Gregory PD, Holmes MC
(2005) Highly efficient endogenous human gene correction using
designed zinc-finger nucleases. Nature 435(7042):646–651. https
://doi.org/10.1038/natur e0355 6
13. Urnov FD, Rebar EJ, Holmes MC, Zhang HS, Gregory PD
(2010) Genome editing with engineered zinc finger nucleases.
Nat Rev Genet 11:636–646. https ://doi.org/10.1038/nrg28 42
14. Meng X, Noyes MB, Zhu LJ, Lawson ND, Wolfe SA (2008)
Targeted gene inactivation in zebrafish using engineered zinc-
finger nucleases. Nat Biotechnol 26(6):695–701. https ://doi.
org/10.1038/nbt13 98
15. Gupta A, Meng X, Zhu LJ, Lawson ND, Wolfe SA (2011) Zinc
finger protein-dependent and-independent contributions to the
invivo off- target activity of zinc finger nucleases. Nucleic Acids
Res 39(1):381–392. https ://doi.org/10.1093/nar/gkq78 7
16. Chu SW, Noyes MB, Christensen RG, Pierce BG, Zhu LJ, Weng
Z, Stormo GD, Wolfe SA (2012) Exploring the DNA-recognition
potential of homeodomains. Genome Res 22(10):1889–1898.
https ://doi.org/10.1101/gr.13901 4.112
17. Enuameh MS, Asriyan Y, Richards A, Christensen RG, Hall VL,
Kazemian M, Zhu C, Pham H, Cheng Q, Blatti C, Brasefield JA,
Basciotta MD, Ou J, McNulty JC, Zhu LJ, Celniker SE, Sinha S,
Stormo GD, Brodsky MH, Wolfe SA (2013) Global analysis of
Drosophila Cys2-His2 zinc finger proteins reveals a multitude of
novel recognition motifs and binding determinants. Genome Res
23(6):928–940. https ://doi.org/10.1101/gr.15147 2.112
18. Shekar M, Venugopal MN (2017) Insight into a transcriptional
adaptor zinc finger encoded by a putative protein in the white
spot syndrome virus genome. Interdiscip Sci Comput Life Sci.
https ://doi.org/10.1007/s1253 9-017-0268-x
19. Boch J, Scholze H, Schornack S, Landgraf A, Hahn S, Kay S,
Lahaye T, Nickstadt A, Bonas U (2009) Breaking the code of
DNA binding specificity of TAL-type III effectors. Science
326:1509–1512. https ://doi.org/10.1126/scien ce.11788 11
20. Moscou MJ, Bogdanove AJ (2009) A simple cipher governs
DNA recognition by TAL effectors. Science 326:1501. https ://
doi.org/10.1126/scien ce.11788 17
21. Bogdanove AJ, Voytas DF (2011) TAL effectors: customizable
proteins for DNA targeting. Science 333:1843–1846. https ://doi.
org/10.1126/scien ce.12040 94
22. Miller JC, Tan S, Qiao G, Barlow KA, Wang J, Xia DF, Meng
X, Paschon DE, Leung E, Hinkley SJ, Dulay GP, Hua KL, Ank-
oudinova I, Cost GJ, Urnov FD, Zhang HS, Holmes MC, Zhang
L, Gregory PD, Rebar EJ (2011) A TALE nuclease architecture
for efficient genome editing. Nat Biotechnol 29:143–148. https
://doi.org/10.1038/nbt.1755
23. Ding Q, Regan SN, Xia Y, Oostrom LA, Cowan CA, Musun-
uru K (2013) Enhanced efficiency of human pluripotent stem
cell genome editing through replacing TALENs with CRIS-
PRs. Cell Stem Cell 12(4):393–394. https ://doi.org/10.1016/j.
stem.2013.03.006
24. Ikmi A, McKinney SA, Delventhal KM, Gibson MC (2014)
TALEN and CRISPR/Cas9-mediated genome editing in the
early-branching metazoan Nematostella vectensis. Nat Commun
5:5486. https ://doi.org/10.1038/ncomm s6486
25. Smith C, Gore A, Yan W, Abalde-Atristain L, Li Z, He C, Wang
Y, Brodsky RA, Zhang K, Cheng L, Ye Z (2014) Whole-genome
sequencing analysis reveals high specificity of CRISPR/Cas9 and
TALEN-based genome editing in human iPSCs. Cell Stem Cell
15(1):12–13. https ://doi.org/10.1016/j.stem.2014.06.011
26. Cong L, Ran FA, Cox D, Lin S, Barretto R, Habib N, Hsu
PD, Wu X, Jiang W, Marraffini LA, Zhang F (2013) Multi-
plex genome engineering using CRISPR/Cas systems. Science
339(6121):819–823. https ://doi.org/10.1126/scien ce.12311 43
27. Mali P, Yang L, Esvelt KM, Aach J, Guell M, DiCarlo JE,
Norville JE, Church GM (2013) RNA-guided human genome
engineering via Cas9. Science 339(6121):823–826. https ://doi.
org/10.1126/scien ce.12320 33
28. Friedland AE, Tzur YB, Esvelt KM, Colaiácovo MP, Church
GM, Calarco JA (2013) Heritable genome editing in C. elegans
via a CRISPR-Cas9 system. Nat Methods 10(8):741–743. https
://doi.org/10.1038/nmeth .2532
29. Gratz SJ, Cummings AM, Nguyen JN, Hamm DC, Donohue
LK, Harrison MM, Wildonger J, O’Connor-Giles KM (2013)
Genome engineering of Drosophila with the CRISPR RNA-
guided Cas9 nuclease. Genetics 194(4):1029–1035. https ://doi.
org/10.1534/genet ics.113.15271 0
30. Hou Z, Zhang Y, Propson NE, Howden SE, Chu LF, Sontheimer
EJ, Thomson JA (2013) Efficient genome engineering in human
pluripotent stem cells using Cas9 from Neisseria meningitidis.
Proc Natl Acad Sci USA 110(39):15644–15649. https ://doi.
org/10.1073/pnas.13135 87110
31. Hwang WY, Fu Y, Reyon D, Maeder ML, Tsai SQ, Sander JD,
Peterson RT, Yeh JR, Joung JK (2013) Efficient genome edit-
ing in zebrafish using a CRISPR-Cas system. Nat Biotechnol
31(3):227–229. https ://doi.org/10.1038/nbt.2501
32. Li D, Qiu Z, Shao Y, Chen Y, Guan Y, Liu M, Li Y, Gao N,
Wang L, Lu X, Zhao Y, Liu M (2013) Heritable gene targeting
in the mouse and rat using a CRISPR-Cas system. Nat Biotechnol
31(8):681–683. https ://doi.org/10.1038/nbt.2661
33. Yang H, Wang H, Shivalila CS, Cheng AW, Shi L, Jaenisch
R (2013) One-step generation of mice carrying reporter
and conditional alleles by CRISPR/Cas-mediated genome
Interdisciplinary Sciences: Computational Life Sciences
1 3
engineering. Cell 154(6):1370–1379. https ://doi.org/10.1016/j.
cell.2013.08.022
34. Chakrapani V, Rasal KD, Kumar S, Mohapatra SD, Sundaray
JK, Jayasankar P etal (2017) In silico analysis of nsSNPs of
carp TLR22 gene affecting its binding ability with Poly I:C.
Interdiscip Sci Comput Life Sci. https ://doi.org/10.1007/s1253
9-017-0247-2
35. Wang T, Wei JJ, Sabatini DM, Lander ES (2014) Genetic
screens in human cells using the CRISPR-Cas9 system. Sci-
ence 343(6166):80–84. https ://doi.org/10.1126/scien ce.12469
81
36. Shalem O, Sanjana NE, Hartenian E, Shi X, Scott DA, Mik-
kelson T, Heckl D, Ebert BL, Root DE, Doench JG, Zhang
F (2014) Genome-scale CRISPR-Cas9 knockout screen-
ing in human cells. Science 343(6166):84–87. https ://doi.
org/10.1126/scien ce.12470 05
37. Koike-Yusa H, Li Y, Tan EP, Velasco-Herrera MC, Yusa K
(2014) Genome-wide recessive genetic screening in mamma-
lian cells with a lentiviral CRISPR-guide RNA library. Nat
Biotechnol 32(3):267–273. https ://doi.org/10.1038/nbt.2800
38. Wang T, Birsoy K, Hughes NW, Krupczak KM, Post Y, Wei
JJ, Lander ES, Sabatini DM (2015) Identification and charac-
terization of essential genes in the human genome. Science
350(6264):1096–1101. https ://doi.org/10.1126/scien ce.aac70
41
39. Tzelepis K, Koike-Yusa H, De Braekeleer E, Li Y, Metzakopian
E, Dovey OM, Mupo A, Grinkevich V, Li M, Mazan M, Goz-
decka M, Ohnishi S, Cooper J, Patel M, McKerrell T, Chen B,
Domingues AF, Gallipoli P, Teichmann S, Ponstingl H, McDer-
mott U, Saez-Rodriguez J, Huntly BJP, Iorio F, Pina C, Vassiliou
GS, Yusa K (2016) A CRISPR dropout screen identifies genetic
vulnerabilities and therapeutic targets in acute myeloid leukemia.
Cell reports 17(4):1193–1205. https ://doi.org/10.1016/j.celre
p.2016.09.079
40. Horlbeck MA, Gilbert LA, Villalta JE, Adamson B, Pak RA,
Chen Y, Fields AP, Park CY, Corn JE, Kampmann M, Weiss-
man JS (2016) Compact and highly active next-generation librar-
ies for CRISPR-mediated gene repression and activation. ELife
5:e19760. https ://doi.org/10.7554/eLife .19760
41. Aguirre AJ, Meyers RM, Weir BA, Vazquez F, Zhang CZ, Ben-
David U, Cook A, Ha G, Harrington WF, Doshi MB, Kost-Ali-
mova M, Gill S, Xu H, Ali LD, Jiang G, Pantel S, Lee Y, Goodale
A, Cherniack AD, Oh C, Kryukov G, Cowley GS, Garraway LA,
Stegmaier K, Roberts CW, Golub TR, Meyerson M, Root DE,
Tsherniak A, Hahn WC (2016) Genomic copy number dictates
a gene-independent cell response to CRISPR/Cas9 targeting.
Cancer Discov 6(8):914–929. https ://doi.org/10.1158/2159-8290.
CD-16-0154
42. Chen S, Sanjana NE, Zheng K, Shalem O, Lee K, Shi X, Scott
DA, Song J, Pan JQ, Weissleder R, Lee H, Zhang F, Sharp PA
(2015) Genome-wide CRISPR screen in a mouse model of
tumor growth and metastasis. Cell 160(6):1246–1260. https ://
doi.org/10.1016/j.cell.2015.02.038
43. Meyers RM, Bryan JG, McFarland JM, Weir BA, Sizemore
AE, Xu H, Dharia NV, Montgomery PG, Cowley GS, Pantel S,
Goodale A, Lee Y, Ali LD, Jiang G, Lubonja R, Harrington WF,
Strickland M, Wu T, Hawes DC, Zhivich VA, Wyatt MR, Kalani
Z, Chang JJ, Okamoto M, Stegmaier K, Golub TR, Boehm JS,
Vazquez F, Root DE, Hahn WC, Tsherniak A (2017) Computa-
tional correction of copy number effect improves specificity of
CRISPR–Cas9 essentiality screens in cancer cells. Nat Genet
49(12):1779. https ://doi.org/10.1038/ng.3984
44. Yennmalli RM, Kalra S, Srivastava PA, Garlapati VK (2017)
Computational tools and resources for crispr/cas 9 genome edit-
ing method. MOJ Proteom Bioinform 5(4):00164. https ://doi.
org/10.15406 /mojpb .2017.05.00164
45. Zhu LJ (2015) Overview of guide RNA design tools for
CRISPR-Cas9 genome editing technology. Front Biol
10(4):289–296. https ://doi.org/10.1007/s1151 5-015-1366-y
46. Fu Y, Sander JD, Reyon D, Cascio VM, Joung JK (2014)
Improving CRISPR-Cas nuclease specificity using truncated
guide RNAs. Nat Biotechnol 32(3):279–284. https ://doi.
org/10.1038/nbt.2808
47. Fu Y, Foden JA, Khayter C, Maeder ML, Reyon D, Joung
JK, Sander JD (2013) High-frequency off-target mutagenesis
induced by CRISPR-Cas nucleases in human cells. Nat Bio-
technol 31(9):822–826. https ://doi.org/10.1038/nbt.2623
48. Jinek M, East A, Cheng A, Lin S, Ma E, Doudna J (2013)
RNA-programmed genome editing in human cells. ELife
2:e00471. https ://doi.org/10.7554/eLife .00471
49. Wong N, Liu W, Wang X (2015) WU-CRISPR: character-
istics of functional guide RNAs for the CRISPR/Cas9 sys-
tem. Genome Biol 16:218. https ://doi.org/10.1186/s1305
9-015-0784-0
50. Doench JG, Fusi N, Sullender M, Hegde M, Vaimberg EW,
Donovan KF, Smith I, Tothova Z, Wilen C, Orchard R, Virgin
HW, Listgarten J, Root DE (2016) Optimized sgRNA design to
maximize activity and minimize off-target effects of CRISPR-
Cas9. Nat Biotechnol 34(2):184. https ://doi.org/10.1038/
nbt.3437
51. Xu H, Xiao T, Chen CH, Li W, Meyer CA, Wu Q, Wu D, Cong
L, Zhang F, Liu JS, Brown M, Liu XS (2015) Sequence deter-
minants of improved CRISPR sgRNA design. Genome Res
25(8):1147–1157. https ://doi.org/10.1101/gr.19145 2.115
52. Mendoza BJ, Trinh CT (2018) Enhanced guide-RNA design and
targeting analysis for precise CRISPR genome editing of single
and consortia of industrially relevant and non-model organisms.
Bioinformatics 34(1):16–23. https ://doi.org/10.1093/bioin forma
tics/btx56 4
53. Cao Q, Ma J, Chen CH, Xu H, Chen Z, Li W, Liu XS (2017)
CRISPR-FOCUS: a web server for designing focused CRISPR
screening experiments. PLoS One 12(9):e0184281. https ://doi.
org/10.1371/journ al.pone.01842 81
54. Malina A, Cameron CJ, Robert F, Blanchette M, Dostie J, Pel-
letier J (2015) PAM multiplicity marks genomic target sites as
inhibitory to CRISPR-Cas9 editing. Nat Commun 6:10124. https
://doi.org/10.1038/ncomm s1012 4
55. Labuhn M, Adams FF, Ng M, Knoess S, Schambach A, Char-
pentier EM, Schwarzer A, Mateo JL, Klusmann JH, Heckl D
(2018) Refined sgRNA efficacy prediction improves large- and
small-scale CRISPR-Cas9 applications. Nucleic Acids Res
46(3):1375–1385. https ://doi.org/10.1093/nar/gkx12 68
56. Chari R, Mali P, Moosburner M, Church GM (2015) Unraveling
CRISPR-Cas9 genome engineering parameters via a library-
on-library approach. Nat Methods 12(9):823. https ://doi.
org/10.1038/nmeth .3473
57. Chari R, Yeo NC, Chavez A, Church GM (2017) sgRNA Scorer
2.0: a species-independent model to predict CRISPR/Cas9 activ-
ity. ACS Synth Biol 6(5):902–904. https ://doi.org/10.1021/acssy
nbio.6b003 43
58. Moreno-Mateos MA, Vejnar CE, Beaudoin JD, Fernandez JP,
Mis EK, Khokha MK, Giraldez AJ (2015) CRISPRscan: design-
ing highly efficient sgRNAs for CRISPR-Cas9 targeting invivo.
Nat Methods 12(10):982. https ://doi.org/10.1038/nmeth .3543
59. Kuan PF, Powers S, He S, Li K, Zhao X, Huang B (2017) A sys-
tematic evaluation of nucleotide properties for CRISPR sgRNA
design. BMC Bioinform 18(1):297. https ://doi.org/10.1186/
s1285 9-017-1697-6
60. Ma J, Köster J, Qin Q, Hu S, Li W, Chen C, Cao Q, Wang J, Mei
S, Liu Q, Xu H, Liu XS (2016) CRISPR-DO for genome-wide
CRISPR design and optimization. Bioinformatics 32(21):3336–
3338. https ://doi.org/10.1093/bioin forma tics/btw47 6
Interdisciplinary Sciences: Computational Life Sciences
1 3
61. Lin Y, Cradick TJ, Brown MT, Deshmukh H, Ranjan P, Sarode
N, Wile BM, Vertino PM, Stewart FJ, Bao G (2014) CRISPR/
Cas9 systems have off-target activity with insertions or deletions
between target DNA and guide RNA sequences. Nucleic Acids
Res 42(11):7473–7485. https ://doi.org/10.1093/nar/gku40 2
62. Hsu PD, Scott DA, Weinstein JA, Ran FA, Konermann S, Agar-
wala V, Li Y, Fine EJ, Wu X, Shalem O, Cradick TJ, Marraffini
LA, Bao G, Zhang F (2013) DNA targeting specificity of RNA-
guided Cas9 nucleases. Nat Biotechnol 31(9):827–832. https ://
doi.org/10.1038/nbt.2647
63. Pattanayak V, Lin S, Guilinger JP, Ma E, Doudna JA, Liu DR
(2013) High-throughput profiling of off-target DNA cleavage
reveals RNA-programmed Cas9 nuclease specificity. Nat Bio-
technol 31:839–843. https ://doi.org/10.1038/nbt.2673
64. Cradick TJ, Fine EJ, Antico CJ, Bao G (2013) CRISPR/Cas9
systems targeting β-globin and CCR5 genes have substantial off-
target activity. Nucleic Acids Res 41(20):9584–9592. https ://doi.
org/10.1093/nar/gkt71 4
65. Cho SW, Kim S, Kim Y, Kweon J, Kim HS, Bae S, Kim JS
(2014) Analysis of off-target effects of CRISPR/Cas-derived
RNA-guided endonucleases and nickases. Genome Res
24(1):132–141. https ://doi.org/10.1101/gr.16233 9.113
66. Tsai SQ, Zheng Z, Nguyen NT, Liebers M, Topkar VV, Thapar V,
Wyvekens N, Khayter C, Iafrate AJ, Le LP, Aryee MJ, Joung JK
(2015) GUIDE-seq enables genome-wide profiling of off-target
cleavage by CRISPR-Cas nucleases. Nat Biotechnol 33(2):187.
https ://doi.org/10.1038/nbt.3117
67. Perez AR, Pritykin Y, Vidigal JA, Chhangawala S, Zamparo L,
Leslie CS, Ventura A (2017) GuideScan software for improved
single and paired CRISPR guide RNA design. Nat Biotechnol
35(4):347–349. https ://doi.org/10.1038/nbt.3804
68. Stemmer M, Thumberger T, Del Sol Keyer M, Wittbrodt J, Mateo
JL (2017) CCTop: an intuitive, flexible and reliable CRISPR/
Cas9 target prediction tool. Plos One 12(4):e0176619. https ://
doi.org/10.1371/journ al.pone.01766 19
69. Singh R, Kuscu C, Quinlan A, Qi Y, Adli M (2015) Cas9-chro-
matin binding information enables more accurate CRISPR off-
target prediction. Nucleic Acids Res 43(18):e118. https ://doi.
org/10.1093/nar/gkv57 5
70. Graham DB, Root DE (2015) Resources for the design of
CRISPR gene editing experiments. Genome Biol 16:260. https
://doi.org/10.1186/s1305 9-015-0823-x
71. Jakočiūnas T, Jensen MK, Keasling JD (2015) CRISPR/Cas9
advances engineering of microbial cell factories. Metab Eng
34:44–59. https ://doi.org/10.1016/j.ymben .2015.12.003
72. Oliveros JC, Mònica F, Daniel TM, David SL, Lluis M, Pilar C
etal (2016) Breaking-Cas—interactive design of guide RNAs
for CRISPR-Cas experiments for ENSEMBL genomes. Nucleic
Acids Res 44(W1):W267. https ://doi.org/10.1093/nar/gkw40 7
73. Zhu H, Misel L, Graham M, Robinson ML, Liang C (2016) CT-
Finder: a web service for CRISPR optimal target prediction and
visualization. Sci Rep 6:25516. https ://doi.org/10.1038/srep2
5516
74. Jiang W, Bikard D, Cox D, Zhang F, Marraffini LA (2013) RNA-
guided editing of bacterial genomes using CRISPR-Cas systems.
Nat Biotechnol 31(3):233–239. https ://doi.org/10.1038/nbt.2508
75. Doench JG, Hartenian E, Graham DB, Tothova Z, Hegde M,
Smith I, Sullender M, Ebert BL, Xavier RJ, Root DE (2014)
Rational design of highly active sgRNAs for CRISPR-Cas9-
mediated gene inactivation. Nat Biotechnol 32(12):1262. https
://doi.org/10.1038/nbt.3026
76. Heigwer F, Kerr G, Boutros M (2014) E-CRISP: fast CRISPR
target site identification. Nat Methods 11(2):122–123. https ://doi.
org/10.1038/nmeth .2812
77. Montague TG, Cruz JM, Gagnon JA, Church GM, Valen E
(2014) CHOPCHOP: a CRISPR/Cas9 and TALEN web tool for
genome editing. Nucleic Acids Res 42(Web Server issue):W401–
W407. https ://doi.org/10.1093/nar/gku41 0
78. Labun K, Montague TG, Gagnon JA, Thyme SB, Valen E (2016)
CHOPCHOP v2: a web tool for the next generation of CRISPR
genome engineering. Nucleic Acids Res 44(W1):W272–W276.
https ://doi.org/10.1093/nar/gkw39 8
79. Zhu LJ, Holmes BR, Aronin N, Brodsky MH (2014) CRISPR-
seek: a bioconductor package to identify target-specific guide
RNAs for CRISPR-Cas9 genome-editing systems. Plos One
9(9):e108424. https ://doi.org/10.1371/journ al.pone.01084 24
80. Bae S, Park J, Kim JS (2014) Cas-OFFinder: a fast and versa-
tile algorithm that searches for potential off-target sites of Cas9
RNA-guided endonucleases. Bioinformatics 30(10):1473. https
://doi.org/10.1093/bioin forma tics/btu04 8
81. Naito Y, Hino K, Bono H, Ui-Tei K (2015) CRISPRdirect:
software for designing CRISPR/Cas guide RNA with reduced
off-target sites. Bioinformatics 31(7):1120–1123. https ://doi.
org/10.1093/bioin forma tics/btu74 3
82. MacPherson CR, Scherf A (2015) Flexible guide-RNA design
for CRISPR applications using Protospacer Workbench. Nat Bio-
technol 33(8):805. https ://doi.org/10.1038/nbt.3291
83. O’Brien A, Bailey TL (2014) GT-Scan: identifying unique
genomic targets. Bioinformatics 30(18):2673–2675. https ://doi.
org/10.1093/bioin forma tics/btu35 4
84. Prykhozhij SV, Rajan V, Gaston D, Berman JN (2015) CRISPR
multiTargeter: a web tool to find common and unique CRISPR
single guide RNA targets in a set of similar sequences. PLoS One
10(3):e0119372. https ://doi.org/10.1371/journ al.pone.01193 72
85. Heigwer F, Zhan T, Breinig M, Winter J, Brügemann D, Leible
S, Boutros M (2016) CRISPR library designer (CLD): software
for multispecies design of single guide RNA libraries. Genome
Biol 17(1):55. https ://doi.org/10.1186/s1305 9-016-0915-2
86. Housden BE, Valvezan AJ, Kelley C, Sopko R, Hu Y, Roesel
C, Lin S, Buckner M, Tao R, Yilmazel B, Mohr SE, Manning
BD, Perrimon N (2015) Identification of potential drug targets
for tuberous sclerosis complex by synthetic screens combining
CRISPR-based knockouts with RNAi. Sci Signal 8(393):rs9.
https ://doi.org/10.1126/scisi gnal.aab37 29
87. Ren X, Yang Z, Xu J, Sun J, Mao D, Hu Y, Yang SJ, Qiao HH,
Wang X, Hu Q, Deng P, Liu LP, Ji JY, Li JB, Ni JQ (2014)
Enhanced specificity and efficiency of the CRISPR/Cas9 sys-
tem with optimized sgRNA parameters in Drosophila. Cell Rep
9(3):1151–1162. https ://doi.org/10.1016/j.celre p.2014.09.044
88. Farboud B, Meyer BJ (2015) Dramatic enhancement of genome
editing by CRISPR/Cas9 through improved guide RNA design.
Genetics 199(4):959–971. https ://doi.org/10.1534/genet
ics.115.17516 6
89. Haeussler M, Schönig K, Eckert H, Eschstruth A, Mianné J,
Renaud JB, Schneider-Maunoury S, Shkumatava A, Teboul L,
Kent J, Joly JS, Concordet JP (2016) Evaluation of off-target and
on-target scoring algorithms and integration into the guide RNA
selection tool CRISPOR. Genome Biol 17(1):148. https ://doi.
org/10.1186/s1305 9-016-1012-2
90. Labaj W, Papiez A, Polanski A, Polanska J (2017) Comprehen-
sive analysis of MILE gene expression data set advances dis-
covery of leukaemia type and subtype biomarkers. Interdiscip
Sci Comput Life Sci 9(1):24–35. https ://doi.org/10.1007/s1253
9-017-0216-9
91. Pei Z, Liu J, Liu M, Zhou W, Yan P, Wen S etal (2018) Risk-
predicting model for incident of essential hypertension based on
environmental and genetic factors with support vector machine.
Interdiscip Sci Comput Life Sci. https ://doi.org/10.1007/s1253
9-017-0271-22016
92. Pulido-Quetglas C, Aparicio-Prat E, Arnan C, Polidori T, Her-
moso T, Palumbo E, Ponomarenko J, Guigo R, Johnson AK
(2017) Scalable design of paired CRISPR guide RNAs for
Interdisciplinary Sciences: Computational Life Sciences
1 3
genomic deletion. PLOS Comput Biol 13(3):e1005341. https ://
doi.org/10.1371/journ al.pcbi.10053 41
93. Rahman MK, Rahman MS (2017) CRISPRpred: a flexible
and efficient tool for sgRNAs on-target activity prediction in
CRISPR/cas9 systems. Plos One 12(8):e0181943. https ://doi.
org/10.1371/journ al.pone.01819 43
94. Hough SH, Kancleris K, Brody L, Humphryes-Kirilov N, Wolan-
ski J, Dunaway K, Ajetunmobi A, Dillard V (2017) Guide Picker
is a comprehensive design tool for visualizing and selecting
guides for CRISPR experiments. BMC Bioinform 18:167. https
://doi.org/10.1186/s1285 9-017-1581-4
95. Zou H, Hastie T (2005) Regularization and variable selection via
the elastic net. J Roy Stat Soc 67(2):301–320
96. Xiong Y, Xie X, Wang Y, Ma W, Liang P, Songyang Z, Dai Z
(2017) pgRNAFinder: a web-based tool to design distance inde-
pendent paired-gRNA. Bioinformatics 33(22):3642–3644. https
://doi.org/10.1093/bioin forma tics/btx47 2
97. Chuai GH, Wang QL, Qi L (2016) In silico meets invivo: towards
computational CRISPR-based sgRNA design. Trends Biotechnol
35(1):12. https ://doi.org/10.1016/j.tibte ch.2016.06.008
98. Efron B, Hastie T, Johnstone I, Tibshirani R (2004) Least angle
regression. Ann Stat 32:407–451
99. Li H, Durbin R (2009) Fast and accurate short read alignment
with Burrows-Wheeler transform. Bioinformatics 25(14):1754–
1760. https ://doi.org/10.1093/bioin forma tics/btp32 4
100. Langmead B, Trapnell C, Pop M, Salzberg SL (2009) Ultra-
fast and memory-efficient alignment of short DNA sequences
to the human genome. Genome Biol 10(3):R25. https ://doi.
org/10.1186/gb-2009-10-3-r25
101. Brazelton VA Jr, Zarecor S, Wright DA, Wang Y, Liu J, Chen
K, Yang B, Lawrence-Dill CJ (2015) A quick guide to CRISPR
sgRNA design tools. Gm Crops Food 6(4):266–276. https ://doi.
org/10.1080/21645 698.2015.11376 90
102. Xie S, Shen B, Zhang C, Huang X, Zhang Y (2014) sgRNAcas9:
a software package for designing CRISPR sgRNA and evaluating
potential off-target cleavage sites. Plos One 9(6):e100448. https
://doi.org/10.1371/journ al.pone.01004 48
103. Abadi S, Yan WX, Amar D, Mayrose I (2017) A machine learn-
ing approach for predicting CRISPR-Cas9 cleavage efficien-
cies and patterns underlying its mechanism of action. PLoS
Comput Biol 13(10):e1005807. https ://doi.org/10.1371/journ
al.pcbi.10058 07
104. Zhu H, Richmond E, Liang C (2018) CRISPR-RT: a web appli-
cation for designing CRISPR-C2c2 crRNA with improved
target specificity. Bioinformatics 34(1):117–119. https ://doi.
org/10.1093/bioin forma tics/btx58 0
105. Zetsche B, Gootenberg JS, Abudayyeh OO, Slaymaker IM,
Makarova KS, Essletzbichler P, Volz SE, Joung J, van der Oost
J, Regev A, Koonin EV, Zhang F (2015) Cpf1 is a single RNA-
guided endonuclease of a class 2 CRISPR-Cas system. Cell
163(3):759. https ://doi.org/10.1016/j.cell.2015.09.038
106. Fonfara I, Le Rhun A, Chylinski K, Makarova KS, Lécrivain AL,
Bzdrenga J, Koonin EV, Charpentier E (2014) Phylogeny of Cas9
determines functional exchangeability of dual-RNA and Cas9
among orthologous type II CRISPR-Cas systems. Nucleic Acids
Res 42(4):2577–2590. https ://doi.org/10.1093/nar/gkt10 74
107. Ran FA, Hsu PD, Lin CY, Gootenberg JS, Konermann S, Trevino
AE, Scott DA, Inoue A, Matoba S, Zhang Y, Zhang F (2013)
Double nicking by RNA-guided CRISPR Cas9 for enhanced
genome editing specificity. Cell 154(6):1380–1389. https ://doi.
org/10.1016/j.cell.2013.08.021
108. Mccaffrey J, Sibert J, Zhang B, Zhang Y, Hu W, Riethman H etal
(2016) CRISPR-Cas9 D10A nickase target-specific fluorescent
labeling of double strand DNA for whole genome mapping and
structural variation analysis. Nucleic Acids Res 44(2):e11-e11.
https ://doi.org/10.1093/nar/gkv87 8
109. Chang L, Sun C, Chen X, Yang W, Zhang J, Zhang Y etal.
(2017). Knocking out of human DNAH2 gene in U2OS cells by
CRISPR/Cas9n double nick system. Sheng Wu Gong Cheng Xue
Bao. https ://doi.org/10.13345 /j.cjb.16027 3
110. Tsai SQ, Wyvekens N, Khayter C, Foden JA, Thapar V, Reyon
D, Goodwin MJ, Aryee MJ, Joung JK (2014) Dimeric CRISPR
RNA-guided FokI-dCas9 nucleases directed by truncated gRNAs
for highly specific genome editing. Nat Biotechnol 32(6):569–
576. https ://doi.org/10.1038/nbt.2908
111. Wu X, Scott DA, Kriz AJ, Chiu AC, Hsu PD, Dadon DB, Cheng
AW, Trevino AE, Konermann S, Chen S, Jaenisch R, Zhang F,
Sharp PA (2014) Genome-wide binding of the CRISPR endonu-
clease Cas9 in mammalian cells. Nat Biotechnol 32(7):670–676.
https ://doi.org/10.1038/nbt.2889
112. Kuscu C, Arslan S, Singh R, Thorpe J, Adli M (2014) Genome-
wide analysis reveals characteristics of off-target sites bound by
the Cas9 endonuclease. Nat Biotechnol 32(7):677–683. https ://
doi.org/10.1038/nbt.2916
... The first and third steps in particular require direct computational intervention (Fig. 2). The most important consideration when designing a specific gRNA is to use computational tools or algorithms to search the entire genome sequence for a unique target region with the lowest off-target specificity [39,40]. While, the post-editing events can be predicted using sequencing and comparing the regions of edited sites using microhomology analysis. ...
... To ensure successful gene editing research, sgRNA design should be extremely specific. With advancement of the genomic resources, CRISPR-Cas techniques, and computational algorithms, several software tools, including web tools have become available for designing and selecting sgRNA sequences for a variety of CRISPR-Cas applications [14,35,[37][38][39][40][41]47]. Most of these tools require input of the DNA sequence, species name, and in some cases the genomic location/gene ID/gene name. ...
Article
Full-text available
Recent advances in genome editing tools and CRISPR-Cas technologies have enabled plant genome engineering reach new heights. The current regulatory exemptions for certain categories of genome edited products, such as those derived from SDN-1 and SDN-2, which are free of any transgene, have significantly accelerated genome editing research in a number of agricultural crop plants in different countries. Although CRISPR-Cas technology is becoming increasingly popular, it is still important to carefully consider a number of factors before planning and carrying conducting CRISPR-Cas studies. To attempt genome editing in a plant, a high-quality genome sequence and a repeatable tissue culture protocol for in vitro regeneration are essential. One of the most important steps in plant genome editing is the designing of a CRISPR construct, which involves selecting the appropriate Cas protein, sgRNA sequence, and appropriate regulatory sequence to trigger expression. Computational tools and algorithms play a crucial role in construct design and gRNA selection to minimize off-target effects and also to optimize their delivery techniques. Researchers may need to select appropriate software tools capable of analyzing post-editing detection of mutation events and other DNA sequence abnormalities to identify off-target effects. To fully fulfill the potential of plant genome editing, continued advances in computational biology are essential to meet the challenges it faces today.
... Despite its broad potential, CRISPR-Cas technology present limitations such as requiring the presence of a 2-6 bp protospacer adjacent motif (PAM), hindering its use in PAM-independent applications 5 . Additionally, limitations such as large size, unstable and charged single guide RNA (sgRNA), and cell membrane barriers constrain its delivery 6,7 . ...
... The purified (wt)RusA, (d)RusA, and FokI-(d)RusA proteins were run on an SDS-PAGE gel with increasing concentrations, (wt)RusA(15,30, and 50 µM); (d)RusA(6,12, and 24 µM); andFokI-(d)RusA (0.4, 0.8, and 1.2 µM) to check for any minor impurities. Qualitative assessment of the gel confirmed that all proteins were pure enough to proceed for further characterizations (Figure S2).We initially tested the activity of (wt)RusA and (d)RusA on a designed HJ named Double Cleavage Strand 1 and 3 (DC-1&3) which allows for proper binding of dimerized RusA then correctly positioning of the dinucleotide within the core homology sequence at the cleavage site of RusA for cleavage (Figure S3a). ...
Preprint
Full-text available
Programmable site-specific nucleases have revolutionized the genome editing. However, these systems still face challenges such as guide dependency, delivery issues, and off-target effects. Harnessing the natural functions of structure-guided nucleases offer promising alternatives for generating site-specific double-strand DNA breaks. Yet, structure-guided nucleases require precise reaction conditions and validation for in-vivo applicability. To address these limitations, we developed the PNA-Coupled FokI-(d)RusA (PC-FIRA) system. PC-FIRA combines the sequence-specific binding ability of peptide nucleic acids (PNAs) with the catalytic efficiency of FokI nuclease fused to a structurally-guided inactive RusA resolvase (FokI-(d)RusA). This system allows for precise double-strand DNA breaks without the constraints of existing site-specific nuclease and structure-guided nucleases. Through in vitro optimizations, we achieved high target specificity and cleavage efficiency. This included adjusting incubation temperature, buffer composition, ion concentration, and cleavage timing. Diverse DNA structures, such as Holliday Junctions, linear, and circular DNA, were tested demonstrating the potential activity on different target forms. Further investigation has revealed the PC-FIRA system capacity for facilitating the precise deletion of large DNA fragments. This can be useful in cloning, large-fragment DNA assembly, and genome engineering, with promising applications in biotechnology, medicine, agriculture, and synthetic biology.
... E-CRISP (http://www.e-crisp.org/E-CRISP /), dan sebagainya (Cui et al., 2018). ...
Book
Full-text available
... However, currently available tools for gRNA design do not allow for determining their secondary structure (Hassan et al., 2021). Guides are predicted by assessing their activity through various tools that have been developed (Bae et al., 2014b;Montague et al., 2014;Moreno-Mateos et al., 2015;Chuai et al., 2018;Concordet and Haeussler, 2018;Cui et al., 2018). Algorithms such as Rule Set one and Rule Set 2 have been developed for on-target activity prediction Heigwer et al., 2014;Wang et al., 2014;Xu et al., 2015). ...
Chapter
Development of the programmable nuclease Ca9 has increased our understanding of the molecular basis of the physiological processes and diseases demonstrating a remarkable potential across various fields, including basic research, applied biotechnology, and biomedical research. CRISPR-Cas9 is a gene-editing technique that uses a guide RNA to direct the Cas9 enzyme to specific DNA sequences, where it creates double-strand breaks. These breaks can be repaired through either nonhomologous end joining (NHEJ), leading to gene mutations, or homology-directed repair (HDR), enabling precise edits. These characteristics render Cas9 an ideal candidate for various genome editing applications, particularly for the generation of knockout cells to study gene function either in vitro or in vivo. Delivery of the CRISPR molecular machinery is a crucial step for successful editing. Lentiviral vectors have been shown as an extremely effective method to transduce a wide range of cell types including neurons, astrocytes, adult neuronal stem cells, and oligodendrocytes. Here, we provide a useful protocol based on the lentiviral approach to develop CRISPR edition, facilitating the study of gene function in mammalian cells.
Article
Full-text available
The CRISPR-Cas9 technology, one of the groundbreaking genome editing methods for addressing genetic disorders, has emerged as a powerful, precise, and efficient tool. However, its clinical translation remains hindered by challenges in delivery efficiency and targeting specificity. This review provides a comprehensive analysis of the structural features, advantages, and potential applications of various non-viral and stimuli-responsive systems, examining recent progress to emphasize the potential to address these limitations and advance CRISPR-Cas9 therapeutics. We describe how recent reports emphasize that nonviral vectors, including lipid-based nanoparticles, extracellular vesicles, polymeric nanoparticles, gold nanoparticles, and mesoporous silica nanoparticles, can offer diverse advantages to enhance stability, cellular uptake, and biocompatibility, based on their structures and physio-chemical stability. We also summarize recent progress on stimuli-responsive nanoformulations, a type of non-viral vector, to introduce precision and control in CRISPR-Cas9 delivery. Stimuli-responsive nanoformulations are designed to respond to pH, redox states, and external triggers, facilitate controlled and targeted delivery, and minimize off-target effects. The insights in our review suggest future challenges for clinical applications of gene therapy technologies and highlight the potential of delivery systems to enhance CRISPR-Cas9’s clinical efficacy, positioning them as pivotal tools for future gene-editing therapies.
Chapter
Micronutrient starvation, commonly known as hidden hunger, is very common in more than two billion people worldwide and the improvement of nutritional values via biofortification employing selective breeding and genetic enhancement methods represents a potential solution to mitigate this issue. Recognizing their historical significance in providing a nutritious diet and sustaining agricultural production, researchers have diligently pursued the acquisition of new genetic traits, including enhanced yield, biotic and abiotic stress tolerance, and nutrient quality, in legumes, which have long served as rich sources of protein and phytochemicals for human consumption over millennia. In last few years, the notable expansion of legume germplasms has laid the foundation for the utilization of state-of-the-art breeding methodologies, including genetic engineering, genomic editing, and selection, to enhance efforts for betterment of the crops. The emergence of advanced genetic engineering techniques like legume transformation and genome editing techniques, such as CRISPR/Cas9, Zinc Finger Nucleases (ZFNs), Transcription Activator-like Effector Nucleases (TALENs) empower molecular scientists to precisely and efficiently manipulate genes. Various methods for delivering DNA exist, with Agrobacterium-mediated and particle bombardment being commonly used for optimizing traits. Nonetheless, challenges such as recalcitrance and genotype-dependence hinder successful transformations. In this book chapter, we have discussed the recent progress, evolution and future prospects of these genetic engineering and genome editing tools with special emphasis to nutritional quality improvement of different legumes.
Article
Clustered regularly interspaced short palindromic repeats (CRISPR) technology has transformed molecular biology and the future of gene-targeted therapeutics. CRISPR systems comprise a CRISPR-associated (Cas) endonuclease and a guide RNA (gRNA) that can be programmed to guide sequence-specific binding, cleavage, or modification of complementary DNA or RNA. However, the application of CRISPR-based therapeutics is challenged by factors such as molecular size, prokaryotic or phage origins, and an essential gRNA cofactor requirement, which impact efficacy, delivery and safety. This Review focuses on chemical modification and engineering approaches for gRNAs to enhance or enable CRISPR-based therapeutics, emphasizing Cas9 and Cas12a as therapeutic paradigms. Issues that chemically modified gRNAs seek to address, including drug delivery, physiological stability, editing efficiency and off-target effects, as well as challenges that remain, are discussed.
Article
Full-text available
Genome editing with the CRISPR-Cas9 system has enabled unprecedented efficacy for reverse genetics and gene correction approaches. While off-target effects have been successfully tackled, the effort to eliminate variability in sgRNA efficacies-which affect experimental sensitivity-is in its infancy. To address this issue, studies have analyzed the molecular features of highly active sgRNAs, but independent cross-validation is lacking. Utilizing fluorescent reporter knock-out assays with verification at selected endogenous loci, we experimentally quantified the target efficacies of 430 sgRNAs. Based on this dataset we tested the predictive value of five recently-established prediction algorithms. Our analysis revealed a moderate correlation (r = 0.04 to r = 0.20) between the predicted and measured activity of the sgRNAs, and modest concordance between the different algorithms. We uncovered a strong PAM-distal GC-content-dependent activity, which enabled the exclusion of inactive sgRNAs. By deriving nine additional predictive features we generated a linear model-based discrete system for the efficient selection (r = 0.4) of effective sgRNAs (CRISPRater). We proved our algorithms' efficacy on small and large external datasets, and provide a versatile combined on- and off-target sgRNA scanning platform. Altogether, our study highlights current issues and efforts in sgRNA efficacy prediction, and provides an easily-applicable discrete system for selecting efficient sgRNAs.
Article
Full-text available
The transcriptional adaptor zinc (TAZ) fingers are a specialized class of zinc finger domains reported to exist only in eukaryotic transcriptional coactivator proteins. A putative protein within the shrimp white spot syndrome virus (WSSV) encodes for a TAZ domain, which is unique as no virus so far has been reported for the presence of this domain. Our study shows the viral TAZ domain to be similar to TAZ2 rather than TAZ1 domain of eukaryotic CREB-binding proteins and its paralog p300 proteins. Furthermore, as with eukaryotic TAZ2 domain which interacts and binds to several transcriptional factors including the p53 tumor suppressor protein, an in silico docking study of the WSSV-TAZ and the shrimp p53 transcriptional factor showed the two protein domains to be involved in a protein–protein interaction.
Article
Full-text available
The CRISPR–Cas9 system has revolutionized gene editing both at single genes and in multiplexed loss-of-function screens, thus enabling precise genome-scale identification of genes essential for proliferation and survival of cancer cells1,2. However, previous studies have reported that a gene-independent antiproliferative effect of Cas9-mediated DNA cleavage confounds such measurement of genetic dependency, thereby leading to false-positive results in copy number–amplified regions3,4. We developed CERES, a computational method to estimate gene-dependency levels from CRISPR–Cas9 essentiality screens while accounting for the copy number–specific effect. In our efforts to define a cancer dependency map, we performed genome-scale CRISPR–Cas9 essentiality screens across 342 cancer cell lines and applied CERES to this data set. We found that CERES decreased false-positive results and estimated sgRNA activity for both this data set and previously published screens performed with different sgRNA libraries. We further demonstrate the utility of this collection of screens, after CERES correction, for identifying cancer-type-specific vulnerabilities.
Article
Full-text available
The adaptation of the CRISPR-Cas9 system as a genome editing technique has generated much excitement in recent years owing to its ability to manipulate targeted genes and genomic regions that are complementary to a programmed single guide RNA (sgRNA). However, the efficacy of a specific sgRNA is not uniquely defined by exact sequence homology to the target site, thus unintended off-targets might additionally be cleaved. Current methods for sgRNA design are mainly concerned with predicting off-targets for a given sgRNA using basic sequence features and employ elementary rules for ranking possible sgRNAs. Here, we introduce CRISTA (CRISPR Target Assessment), a novel algorithm within the machine learning framework that determines the propensity of a genomic site to be cleaved by a given sgRNA. We show that the predictions made with CRISTA are more accurate than other available methodologies. We further demonstrate that the occurrence of bulges is not a rare phenomenon and should be accounted for in the prediction process. Beyond predicting cleavage efficiencies, the learning process provides inferences regarding patterns that underlie the mechanism of action of the CRISPR-Cas9 system. We discover that attributes that describe the spatial structure and rigidity of the entire genomic site as well as those surrounding the PAM region are a major component of the prediction capabilities.
Article
Full-text available
CRISPR-Cas systems have been successfully applied in genome editing. Recently, the CRISPR-C2c2 system has been reported as a tool for RNA editing. Here we describe CRISPR-RT (CRISPR RNA-Targeting), the first web application to help biologists design the crRNA with improved target specificity for the CRISPR-C2c2 system. CRISPR-RT allows users to set up a wide range of parameters, making it highly flexible for current and future research in CRISPR-based RNA editing. CRISPR-RT covers major model organisms and can be easily extended to cover other species. CRISPR-RT will empower researchers in RNA editing. Availability: Freely available at http://bioinfolab.miamioh.edu/CRISPR-RT . Contact: liangc@miamioh.edu. Supplementary information: Supplementary data are available in Supplemental.Info.pdf.
Article
Full-text available
The CRISPR/Cas System has been shown to be an efficient and accurate genome-editing technique. There exist a number of tools to design the guide RNA sequences and predict potential off-target sites. However, most of the existing computational tools on gRNA design are restricted to small deletions. To address this issue, we present pgRNAFinder, with an easy-to-use web interface, which enables researchers to design single or distance-free paired-gRNA sequences. The web interface of pgRNAFinder contains both gRNA search and scoring system. After users input query sequences, it searches gRNA by 3' protospacer-adjacent motif (PAM), and possible off-targets, and scores the conservation of the deleted sequences rapidly. Filters can be applied to identify high-quality CRISPR sites. PgRNAFinder offers gRNA design functionality for 8 vertebrate genomes. Furthermore, to keep pgRNAFinder open, extensible to any organism, we provide the source package for local use. Availability and implementation: The pgRNAFinder is freely available at http://songyanglab.sysu.edu.cn/wangwebs/pgRNAFinder/ , and the source code and user manual can be obtained from https://github.com/xiexiaowei/pgRNAFinder .
Article
Full-text available
The recently developed CRISPR screen technology, based on the CRISPR/Cas9 genome editing system, enables genome-wide interrogation of gene functions in an efficient and cost-effective manner. Although many computational algorithms and web servers have been developed to design single-guide RNAs (sgRNAs) with high specificity and efficiency, algorithms specifically designed for conducting CRISPR screens are still lacking. Here we present CRISPR-FOCUS, a web-based platform to search and prioritize sgRNAs for CRISPR screen experiments. With official gene symbols or RefSeq IDs as the only mandatory input, CRISPR-FOCUS filters and prioritizes sgRNAs based on multiple criteria, including efficiency, specificity, sequence conservation, isoform structure, as well as genomic variations including Single Nucleotide Polymorphisms and cancer somatic mutations. CRISPR-FOCUS also provides pre-defined positive and negative control sgRNAs, as well as other necessary sequences in the construct (e.g., U6 promoters to drive sgRNA transcription and RNA scaffolds of the CRISPR/Cas9). These features allow users to synthesize oligonucleotides directly based on the output of CRISPR-FOCUS. Overall, CRISPR-FOCUS provides a rational and high-throughput approach for sgRNA library design that enables users to efficiently conduct a focused screen experiment targeting up to thousands of genes. (CRISPR-FOCUS is freely available at http://cistrome.org/crispr-focus/)
Article
Full-text available
The CRISPR/Cas9-sgRNA system has recently become a popular tool for genome editing and a very hot topic in the field of medical research. In this system, Cas9 protein is directed to a desired location for gene engineering and cleaves target DNA sequence which is complementary to a 20-nucleotide guide sequence found within the sgRNA. A lot of experimental efforts, ranging from in vivo selection to in silico modeling, have been made for efficient designing of sgRNAs in CRISPR/Cas9 system. In this article, we present a novel tool, called CRISPRpred, for efficient in silico prediction of sgRNAs on-target activity which is based on the applications of Support Vector Machine (SVM) model. To conduct experiments, we have used a benchmark dataset of 17 genes and 5310 guide sequences where there are only 20% true values. CRISPRpred achieves Area Under Receiver Operating Characteristics Curve (AUROC-Curve), Area Under Precision Recall Curve (AUPR-Curve) and maximum Matthews Correlation Coefficient (MCC) as 0.85, 0.56 and 0.48, respectively. Our tool shows approximately 5% improvement in AUPR-Curve and after analyzing all evaluation metrics, we find that CRISPRpred is better than the current state-of-the-art. CRISPRpred is enough flexible to extract relevant features and use them in a learning algorithm. The source code of our entire software with relevant dataset can be found in the following link: https://github.com/khaled-buet/CRISPRpred.
Article
Motivation: Genetic diversity of non-model organisms offers a repertoire of unique phenotypic features for exploration and cultivation for synthetic biology and metabolic engineering applications. To realize this enormous potential, it is critical to have an efficient genome editing tool for rapid strain engineering of these organisms to perform novel programmed functions. Results: To accommodate the use of CRISPR/Cas systems for genome editing across organisms, we have developed a novel method, named CASPER (CRISPR Associated Software for Pathway Engineering and Research), for identifying on- and off-targets with enhanced predictability coupled with an analysis of non-unique (repeated) targets to assist in editing any organism with various endonucleases. Utilizing CASPER, we demonstrated a modest 2.4% and significant 30.2% improvement (F-test, p<0.05) over the conventional methods for predicting on- and off-target activities, respectively. Further we used CASPER to develop novel applications in genome editing: multitargeting analysis (i.e. simultaneous multiple-site modification on a target genome with a sole guide-RNA (gRNA) requirement) and multispecies population analysis (i.e. gRNA design for genome editing across a consortium of organisms). Our analysis on a selection of industrially relevant organisms revealed a number of non-unique target sites associated with genes and transposable elements that can be used as potential sites for multitargeting. The analysis also identified shared and unshared targets that enable genome editing of single or multiple genomes in a consortium of interest. We envision CASPER as a useful platform to enhance the precise CRISPR genome editing for metabolic engineering and synthetic biology applications. Availability: https://github.com/TrinhLab/CASPER. Contact: ctrinh@utk.edu. Supplementary information: Supplementary data are available at Bioinformatics online.
Article
Immune response mediated by toll-like receptor 22 (TLR22), only found in teleost/amphibians, is triggered by double-stranded RNA binding to its LRR (leucine-rich repeats) ecto-domain. Accumulated evidences suggested that missense mutations in TLR genes affect its function. However, information on mutation linked pathogen recognition for TLR22 was lacking. The present study was commenced for predicting the effect of non-synonymous single-nucleotide polymorphisms (nsSNPs) on the pathogen recognizable LRR domain of TLR22 of farmed carp, Labeo rohita. The sequence-based algorithms (SIFT, PROVEAN and I-Mutant2.0) indicated that three SNPs (out of 27) such as p.L159F (rs76759876) and p.L529P (rs749355507) of LRR, and p.I836M (rs750758397) of intracellular motifs could potentially disrupt protein function. The 3D structure was generated using MODELLER 9.13 and further validated by SAVEs server. The simulated molecular docking of native TLR22 and mutants with poly I:C ligand indicated that mutations positioned at p.L159F and p.L529P of the LRR region affects the binding affinity significantly. This is the first kind of study of predicting nsSNPs of teleost TLR22 with disturbed ligand binding affinity with its extra-cellular LRR domain and thereby likely hindrance in subsequent signal transduction. This study serves as a guide for in vivo evaluation of impact of mutation on immune response mediated by teleost TLR22 gene.