Abstract
Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR)–CRISPR associated protein (CRISPR-Cas) has emerged and evolved as a revolutionary genome editing technology, transforming research across diverse biological disciplines. Over the past decade, this technology has unveiled numerous opportunities for precise genome manipulation. However, the processes of discovering Cas proteins, repurposing them as editing tools, selecting appropriate candidate tool from the CRISPR-toolbox, designing experiments, and analyzing data are often complex and require careful consideration. To support researchers at every stage of CRISPR experimentation, a wide array of web resources has been developed. In this article, we provide a comprehensive overview of standalone and web-based tools that assist in the identification of CRISPR-Cas systems and the design of guide RNAs (gRNAs). We also highlight tools for evaluating gRNA efficiency, predicting CRISPR-Cas9 mutation profiles, as well as tools for base editing and prime editing, and the analysis and visualization of experimental results. Additionally, we introduce CRISPR–Gateway for Accessing Tools and Resources (CRISPR-GATE), an all-inclusive web repository that consolidates publicly available tools for genome editing research. This repository offers a categorized and user-friendly interface, allowing researchers to quickly access relevant tools based on their specific needs. CRISPR-GATE aims to streamline the search for CRISPR resources, facilitating both education and accelerating innovation. The web repository can be accessed from https://crispr-gate.daasbioinfromaticsteam.in/.
Keywords: CRISPR-Cas tools, gRNA design, mutation prediction, base editing, prime editing, TnpB, web resources, CRISPR-GATE
Introduction
On account of its extraordinary efficiency, ease, directness, versatility, and ability to target both DNA and RNA, the Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR)–CRISPR associated protein (CRISPR-Cas) system has emerged as a promising genome and transcriptome manipulation technology and has swiftly superseded its forerunners in the past decade [1]. It finds applications in various fields of biology and beyond, such as gene therapy, crop improvement, and industrial biotechnology [2, 3].
Recent advancements have significantly expanded CRISPR-Cas functionalities. Dead Cas9 (dCas9) fused with effector proteins enables precise regulation of gene expression via CRISPR activation (CRISPRa) or interference (CRISPRi) [4, 5]. Cas-induced DNA breaks enable precise nucleotide replacement, insertion, or deletion via homology-directed repair (HDR) using a donor template. Fusion of nucleotide deaminases with nickase Cas9 (nCas9) or dCas9 facilitates base editing, enabling precise nucleotide changes without introducing double-strand breaks (DSBs) or requiring donor templates [6–8]. Prime editing allows versatile genomic modifications, including insertions/deletions, all types of base substitutions, and a combination of the above in the DNA [9]. Additionally, CRISPR technologies are extensively utilized for epigenetic studies [10], genetic screening [11], artificial chromatin loop formations [12], genome organization control systems [13], real-time chromatin and RNA imaging [14, 15], RNA editing [16], molecular diagnostics [17], gene drives [18], and much more.
The exploration of CRISPR-Cas systems in prokaryotic genomes initially relied on sequence similarity searches against known sequences, but later shifted toward signature-based methods that use algorithms to detect characteristic features. The combination of these approaches has enabled the effective discovery of CRISPR arrays, Cas genes, protospacer adjacent motif (PAM) sequences, and anti-CRISPR proteins (Acrs) [19–22]. The discovery of functional CRISPR-Cas systems in natural organisms has laid the foundation for their repurposing as genome editing tools.
Selecting and manually designing guide RNAs (gRNAs), evaluating different strategies, avoiding off-targets in complex genomes, predicting outcomes, and analyzing experimental data can be challenging for researchers new to the field. Fortunately, a wide range of computational tools has made it easier to perform CRISPR-Cas experiments in a more streamlined and user-friendly manner. Many of these tools incorporate diverse features to evaluate and optimize gRNAs, aiming to maximize efficiency and specificity in experimental setups [23, 24]. Moreover, gRNA design for base editing and prime editing is technically more complex, requiring dedicated tools for accurate modeling and interpretation. In the case of base editors, additional considerations such as bystander effects, influence of neighboring sequences, spacer position, editor-specific activity windows, and potential off-target effects are crucial for effective gRNA design [25, 26]. For prime editors, prime editing gRNA (pegRNA) design involves additional components, making the process more intricate and subject to various influencing factors [9, 27].
Although Cas-induced DNA double-strand breaks (DSBs) were long thought to produce random indels, recent studies have shown that these mutations can be predicted [28, 29]. The availability of prediction tools has made it easier to design more effective experiments [30–34]. The complexity of plant genomes, longer experimental timelines, and emerging applications like multiplexing and gene regulation underscore the critical need for CRISPR-Cas tools specifically optimized for plants.
The final hurdles often lie in data analysis and interpretation. To address this, both standalone and web-based tools have been developed to analyze mutation data using either Sanger sequencing and/or high-throughput sequencing datasets [35, 36]. More advanced tools have since emerged to support the analysis and visualization of outcomes from diverse experiments, including base editing, prime editing, and pooled CRISPR screens [37, 38].
Currently, studies with adequate details of the full spectrum of CRISPR-Cas tools, enlisting the majority of available tools, starting from the discovery of CRISPR-Cas systems to downstream analysis of editing experiments, are scarce. Researchers often need to browse through numerous resources to find the appropriate tools that suit their experimental needs. For instance, a conventional gRNA design tool cannot serve the purpose of base editing and prime editing experiments, highlighting the need for a unified resource. While several resources have compiled CRISPR-Cas tools, such as the CRISPR Software Matchmaker (https://tinyurl.com/e7nt8dxd), the Wikipedia page on CRISPR/Cas tools (https://en.wikipedia.org/wiki/CRISPR/Cas_tools), WeReview: CRISPR Tools [39], and the awesome-CRISPR GitHub repository (https://github.com/davidliwei/awesome-CRISPR), they often suffer from outdated content, limited scope, inconsistent maintenance, lack of depth, and advanced interactivity. Similarly, there have been extended reviews on computational tools and resources for CRISPR-Cas genome editing [40, 41]. However, none of them has produced an elaborate application-wise compilation with an integrated web resource.
In this review, we investigate and comprehensively discuss various CRISPR-Cas computational tools, including those used for the detection of CRISPR-Cas systems in prokaryotic genomes, gRNA design, evaluation of gRNA efficiency, prediction of CRISPR-Cas9 mutation profiles, base editing and prime editing techniques, and analysis and visualization of editing results (Fig. 1). We have placed greater emphasis on freely available tools over commercial ones. To address the challenges discussed earlier and to ensure easy access to CRISPR-Cas tools, we have tabulated and classified them based on the nature of experiments, expected outcomes, and popularity. The technical aspects of these tools, as they relate to specific tasks, are also reviewed in detail.
Figure 1.

Schematic pipeline for selecting different categories of bioinformatics tools for CRISPR-based experimentation.
We conducted a comparative analysis to help readers understand and make informed decisions based on the utilities of each tool. Each tool was systematically evaluated against a predefined set of main and sub-features. A three-tier scoring system was employed at the sub-feature level, where a score of 1 indicates full support, 0.5 denotes partial support or support with limitations, and 0 signifies no support. The data were then normalized to a consistent bounded scale (0–1) using the average score within the set for better visualisation and to enable fair comparison across tools. These results are visualized as heatmaps, providing a clear overview of each tool’s capabilities. Additionally, we developed a one-stop web resource, CRISPR-GATE, to help the scientific community identify suitable tools and minimizing time investment.
Discovery of novel CRISPR-Cas systems from prokaryotes
To repurpose CRISPR-Cas systems as genome editing tools, it is essential to thoroughly explore their fundamental components. In this section, we discuss the computational tools available for such initial exploration, including the identification of CRISPR arrays, Cas genes, PAM sequences, and other critical elements that constitute a functional system. Identifying the class, type, and subtype of CRISPR systems present in a genome of interest is crucial, as not all CRISPR systems possess desirable properties for genome editing applications. For example, Class 2 systems—specifically Type II and Type V—are particularly valuable due to the presence of a single-effector Cas nuclease, making them highly suitable for tool development. At a minimum, a computational tool should be capable of identifying CRISPR arrays and their associated Cas genes. More advanced tools offer system-wide classification by type or subtype and can identify critical components such as PAM sequences. Tools that determine the transcriptional orientation of arrays and support metagenomic datasets are also particularly useful for specific research contexts.
There are various tools and approaches for discovering CRISPR-Cas systems, either through sequence similarity searches or signature-based methods that employ machine learning algorithms. Bioinformatics tools can detect CRISPR arrays based on characteristics such as the presence of conserved repeats and spacers. Cas genes can be detected mainly by utilizing tools based on sequence similarity to known Cas genes. Eventually, a combination of these technologies will allow us to effectively uncover CRISPR-Cas systems in a genome or metagenome, as listed in Fig. 2.
Figure 2.
Tools for exploration of CRISPR-Cas systems and identifying anti-CRISPR proteins.
In the early days, sequence similarity-based matching tools such as BLAST [42], HMM [43], and PatScan [44] were used. However, those tools suffer from many drawbacks and demand manual curation of the outputs. Later, advanced tools with higher processing capabilities were developed like PILER-CR [45], CRISPR Recognition Tool (CRT) [46], CRISPRFinder [47], CRISPRDetect [48], CRISPRdigger [49], CRF [50], CRISPRidentify [51], and CRISPRclassify [52] for finding CRISPR repeats from both assembled and unassembled genomic sequences. PILER-CR is the first specialized tool for CRISPR detection, whereas CRF is the first tool to harness machine learning algorithms. CRISPRidentify produce very less to zero false positive rate, while CRISPRclassify employed a novel repeat-based method independent of Cas genes. CRISPRDirection [53], CRISPRstrand [54], and Potential Orientation and Cas Orientation [55] tools enable predicting an array’s transcriptional direction for exploring non-canonical functions and identifying leader regions. CRISPRDirection integrated into CRISPRCasFinder [20, 47] and CRISPRDetect, while CRISPRstrand has been integrated into the CRISPRmap [19]. CRISPRleader is a dedicated tool for the identification of CRISPR leader sequences [56].
Although many Cas proteins share structural similarities, significant variability exists even within the same subtype [57]. In the last decade, numerous computational tools have been developed for CRISPR-Cas system analysis—though most focus on CRISPR arrays rather than Cas genes [45, 48, 52]. Efforts to classify Cas genes based on the nature of effector proteins, sequences, and Cas operon architecture have resulted in a classification system with two classes, six types, and 33 subtypes [58, 59]. This has spurred the development of tools such as MacSyFinder [21], HMMCAS [60], CRISPRcasIdentifier [61], Casboundary [62], CASPredict [63], and CRISPRCasStack [64]. MacSyFinder and HMMCAS follow reference-guided approaches, whereas CRISPRcasIdentifier, Casboundary, CASPredict, and CRISPRCasStack employ machine learning techniques. HMMCAS, CASPredict, and CRISPRCasStack offer web tools, whereas MacSyFinder, CRISPRcasIdentifier, and Casboundary are standalone programs. An online resource of Cas genes was built through similarity search and genomic neighborhood analysis of metagenome datasets [65]. As a database, CasPDB serves as an informative and annotated resource [66]. Using CRISPR-Cas-Docker, optimal crRNA-Cas protein pair compatibility can be studied in silico before conducting expensive and time-consuming experiments [67]. Recently, CasPEDIA has come up with a comprehensive database of the Class 2 CRISPR-Cas proteins incorporating their enzymatic properties [68].
In order to support single-platform analysis, some tools and web servers have integrated CRISPR array and Cas gene identification. Such tools for exploration of CRISPR-Cas systems include CRISPRmap, CRISPRdisco [69], CRISPRCasFinder, CRISPRCasTyper [70], and CRISPRloci [71]. Both CRISPRmap and CRISPRloci tools are available under the Freiburg RNA tools platform [72]. The CRISPRmap tool detects CRISPR arrays using CRISPRFinder and CRT, and detects Cas genes using HMMER. CRISPRdisco makes use of MinCED [46] and BLAST tools for detecting CRISPR arrays and Cas genes, respectively. The CRISPRCasFinder web tool is accessible from the CRISPR-Cas++ platform [20], detects CRISPR arrays using an improved version of CRISPRFinder. The most recent tools, CRISPRCasTyper and CRISPRloci, use machine learning approaches based on signature genes. CRISPRCasTyper employs MinCED for detection of CRISPR arrays and HMMER for identification of Cas proteins. On the other hand, CRISPRloci has integrated several tools to support the discovery of CRISPR-Cas systems with detailed annotations. There are also web-based interactive databases equipped with graphical tools that facilitate the identification of CRISPR arrays and Cas genes, such as CRISPI [73], CRISPRBank [48], CRISPRone [74], CRISPRminer [75], CRISPRCasdb [76, 77], and CRISPRimmunity [78].
Identifying CRISPR-Cas systems from metagenome sequences
Discovery from complex environmental samples requires specialized tools optimized for fragmented data. These tools can identify CRISPR components from either assembled sequences or raw, unassembled sequencing reads. Though many of the available tools to detect CRISPR arrays, like CRT and CRISPRFinder, can be used for metagenomic assembly data, dedicated tools optimized for the complexity of such data were developed only later. The standalone metaCRT tool helps in the detection of CRISPRs from whole-metagenome assemblies [79]. However, Crass supports a raw read-based method for the identification of CRISPR arrays from metagenomes [80]. Likewise, metaCRISPR [81] and MetaCRAST [82] detect CRISPR arrays using unassembled raw metagenomic datasets. The functionalities for identification and annotation of CRISPRs from environmental datasets are also facilitated by tools like MinCED and CRISPRCasMeta [20].
Determining PAM
PAM is conserved and crucial for Cas effector to identify and cleave target sequence [83]. Prior to the availability of in silico analysis, the PAM sequences were determined via time-consuming experimental methods such as in vivo and in vitro selections [84, 85]. CASPERpam is a dedicated tool that makes it possible to identify and access the PAMs by analyzing CRISPR spacer sequences mined from genomic data within NCBI’s public repositories. The resulting PAM sequence database and groups are made available as a new feature in the CASPER software platform [86]. Spacer2PAM [87] and PAMpredict [88] also support the detection of functional PAMs. PAM prediction using PAMpredict is found to exhibit high accuracy [88].
Identifying Acrs
Acrs are inhibitors produced by bacteriophages and mobile genetic elements that counteract CRISPR-Cas immune systems [89]. Potential Acrs can act as natural off-switches for CRISPR-Cas systems, making them valuable tools for regulating genome editing. CRISPRminer and CRISPRimmunity provide database support for identifying self-targeting and anti-CRISPR proteins. Initially, a simple database was created to track Acr names and prevent nomenclature confusion [90]. Later, more advanced databases with detailed annotations were developed, such as AcrDB [91], AcrHub [92], and Anti-CRISPRdb [93, 94]. Among these, only AcrDB supports anti-CRISPR-associated proteins (Acas) exploration.
Several tools have been developed for Acr prediction, including AcRanker [22], AcrFinder [95], PaCRISPR [96], AcaFinder [97], DeepAcr [98], PreAcrs [99], and AcrNET [100]. Predictions have led to the discovery of many potential Acrs [22, 101]. Among these, AcrFinder, PaCRISPR, AcaFinder, and AcrNET are web-based tools. For Aca predictions, AcaFinder is the first exclusive tool, with integrated CRISPRCasTyper for additional assessments.
The development of tools for CRISPR-Cas system discovery reflects the evolution of bioinformatics programming paradigms. Early and foundational tools were implemented in Perl and Java, while modern integrated platforms are predominantly built using Python. Overall, a wide range of tools facilitate the discovery of CRISPR-Cas systems and their essential components, like CRISPR arrays, Cas proteins, Acr and Aca proteins, and PAM sequences. To help an end-user make an informed decision when choosing the best tool from the comprehensive options available, we have provided a feature-wise comparison heatmap in Fig. 3. This figure scores the major features of each tool based on its underlying capabilities, and the full details are available in Supplementary Table 1.
Figure 3.
Comparative account of tools to identify different components of CRISPR-Cas system and anti- CRISPR proteins. Scoring rule: 1, fully compatible; 0.5, partial support or support with limitations; 0, not compatible. Normalized scores, calculated as the average of sub-features, were used to enable a fair comparison across tools.
Guide RNA design for canonical CRISPR-Cas editing (knock-out or random genetic variation)
Once a suitable CRISPR-Cas9 system is identified and characterized, it could be utilized in genome editing applications. The gRNA provides information required for Cas9 to bind to and cleave a specific genetic locus. The success of CRISPR-Cas experiments depends largely on the efficiency (high on-target activity) and specificity (minimal off-target activity) of the gRNAs used. The central challenge in evaluating gRNA design tools is achieving an optimal balance between these two factors. Tools that employ learning-based approaches generally offer more accurate performance predictions. Furthermore, a tool’s utility is greatly enhanced by its versatility—such as support for multiple nuclease variants and advanced applications like transcriptional regulation or allele-specific targeting. Tools that incorporate the most advanced design rules—including considerations of GC content, secondary structure, tracrRNA sequence, and position-dependent nucleotide preferences—are preferable for achieving optimal outcomes. Several computational tools have employed various features for the evaluation and optimization of gRNAs to achieve maximum efficiency and specificity in experiments [23, 24]. Notable features include the position-dependent nucleotide preferences [102, 103], other sequence characteristics of the gRNA and target [104, 105], epigenetic features [106, 107], and structural properties [108, 109].
Previous studies have categorized gRNA design tools into three major types based on gRNA selection criteria: alignment-based, hypothesis-driven, and learning-based methods [23, 110, 111]. Alignment-based methods select gRNAs based solely on alignment criteria, while hypothesis-driven methods rely on empirically derived rule sets for gRNA selection and prediction. In contrast, learning-based tools use machine learning algorithms with experimental data to predict gRNA efficiency and specificity. Most of the tool development was carried out using the Python programming language, followed by Perl, Java, R and others.
Alignment-based and hypothesis-driven tools
Alignment-based tools generally perform a rapid search for potential gRNA target sites by detecting PAM sequences across the genome, without incorporating efficiency or specificity scoring metrics. In contrast, hypothesis-driven tools conduct a more elaborate off-target search by employing diverse sequence alignment algorithms such as BLAST, Bowtie [112], Jellyfish [113], or GGGenome [113], as well as specially developed algorithms tailored for genome-wide guide design [114, 115]. OffScan [116] uses an FM-index [117], while CRISPR-SE [118] employs an optimized brute force algorithm for efficient off-target identification. Different gRNA design platforms adopt a variety of on-target activity scoring schemes to balance predictive power and interpretability. Early tools often relied on empirically derived rule sets—Rule Set 1, based on experimental data from mammalian cells, consider sequence features that most strongly influence CRISPR-Cas9 cutting efficiency [102]. To enhance accuracy, Rule Set 2 integrated additional, larger datasets and introduced new features—position-independent nucleotide counts and the relative location of the gRNA target site within the gene [103], and is used by tools such as CRISPOR [119], and many others. The recently developed Rule Set 3 accounts for variations in the tracrRNA sequence that significantly affect gRNA activity and efficacy [120], which was further validated independently [121], and used to develop CRISPR-BEasy [122] and update CRISPOR. MIT and CFD scores are two widely used specificity scores [103, 123]. CHOPCHOP offers candidate primers, the display of restriction sites, and more, and is enriched with several functionalities with the latest update for conducting CRISPR-Cas experiments [112, 124, 125]. It now supports better visualization of results, targets more genomes, incorporates machine learning-derived evaluation scores, and allows for Cas13-based RNA editing. Some tools provide multiple scores for gRNA activity evaluations, such as CRISPOR and Guide Picker [126]. To help end-users make informed choices among the many available options, Fig. 4 presents a comparative heatmap scoring each tool’s key features based on their underlying functions, with Supplementary Table 2 offering a more detailed analysis.
Figure 4.
Feature-wise comparison of alignment-based and hypothesis-driven gRNA designing tools. Scoring rule: 1, fully compatible; 0.5, partial support or support with limitations; 0, not compatible. Normalized scores, calculated as the average of sub-features, were used to enable a fair comparison across tools.
The CRISPR-RGEN tools platform [127] provides two tools that facilitate gRNA designs, Cas-OFFinder [128] and Cas-Designer [129]. CT-Finder is found to support single and paired-gRNA designs for Cas9, nCas9, and RNA-guided FokI nucleases (RFNs) [130]. PhytoCRISP-Ex specializes in phytoplankton genome targeting [131]. The targetDesign tool from CRISPR-GE software toolkit, supports off-target evaluation and scoring with an associated offTarget program [132]. CRISPR-FOCUS has been optimized for CRISPR screening applications with batch mode functionality [133]. Interestingly, GuideMaker comes with support to facilitate gRNA designs for non-model organisms [134]. GuideScan2 overcomes the limitations of its previous version and provides a memory-efficient approach for scalable design of guide sequences [135, 136]. RNA/DNA bulges contribute to off-target effects [137], and is addressed by tools like Cas-Designer, CT-Finder, and CRISPRitz [138]. Recently, DANGER has employed a unique technique for on-target and off-target assessment without the need for a reference genome [139].
CRISPR-Cas13-based RNA editing experiments are supported by tools like CHOPCHOP, crisprVerse [140], and CASPER [141, 142]. The crisprVerse provides gRNA design for knockout, activation, and interference experiments. CASPER supports multitargeting and multispecies population analysis while facilitating the application of different CRISPR-Cas systems. Multitargeting functionality with support for multiple types of CRISPR-Cas systems is also provided by CRISPR MultiTargeter [143] and CRISPR-broad [144]. CRISPR MultiTargeter allows designing identical target sites in multiple genes, whereas CRISPR-broad facilitates experiments on large genomes.
Allele-specific gRNA design is crucial in precision applications because it enables CRISPR to selectively target mutant alleles, thereby minimizing off-target effects in dominantly inherited traits and similar contexts [145]. Only a few tools support allele-specific gRNA designs, which include CrisPam [146], SNP-CRISPR [147], AlleleAnalyzer [148], AsCRISPR [145], and AlPaCas [149]. AlPaCas is found to offer diverse functionalities and features, such as support for a broader range of Cas enzymes, structural analysis for Cas protein engineering, interactive visualizations, and greater customization.
Learning-based tools
The learning-based tools contribute to gRNA design by providing meaningful evaluations through the prediction of on-target and off-target activities, addressing the limitations of handcrafted rule-based scoring systems [111]. Learning-based tools can effectively comprehend large datasets and numerous features [150]. The common machine learning algorithms for gRNA design include regression [151, 152], classification [102, 153], and ensemble methods [154, 155], whereas deep learning tools mostly use artificial neural networks and convolutional neural networks (CNNs) for predictions [156–158].
The efficiency and specificity of tools employing machine learning algorithms depend on how well the model is trained. Recent tools that facilitate on-target prediction include Uni-deepSG [159], CIAO [160], CRISPR-Aidit [161, 162], and TIGER [163]. Tools specific for off-target activity prediction include CRISOT [164], CRISPR-HW [165], piCRISPR [166], CRISPR-M [167], and CRISPR-DIPOFF [168]. However, some tools have incorporated both on and off-target prediction modules, such as Azimuth 2.0 [103, 169], DeepCRISPR [170], DeepCpf1 [171], Cas12a_predictor [172], CRISep [173], DeepCas13 [174], CRISPR-AIdit, and TIGER. The off-target prediction tool Elevation has been integrated with Azimuth [169]. Likewise, CRISPRater can be accessed through the CCTop online platform [175].
Tools like sgRNA Scorer 2.0 [106, 176], CRISPick [103, 177], AttnToMismatch_CNN and AttnToCrispr_CNN [178], DeepGuide [179], and GuideHOM [180] support predictions for both Cas9 and Cas12a (formerly Cpf1) experiments. Attention-based modules are leveraged by AttnToMismatch_CNN and AttnToCrispr_CNN, CRISPR-ONT and CRISPR-OFFT [181], AttCRISPR [182], CRISPR-IP [183], CRISPR-HW, and CRISPR-M, whereas DeepCRISTL [184] and crisprHAL [185] make use of transfer learning. Similarly, CrisprDNT [186] and CRISPR-DIPOFF makes use of a transformer-based approach. The features utilized by tools for prediction also vary significantly; some tools use only sequence-based features, whereas other tools make use of epigenetic features as well [167, 170, 171]. Thermodynamic features were included in Azimuth, CRISPRpred [187], DeepHF [188], GNL-Scorer [189], CRISPRon and CRISPRoff [190], and BoostMEC [191]. Some tools have incorporated secondary structure features of gRNAs, such as WU-CRISPR [104], CRISTA [192], DeepHF, AttCRISPR, and DeepCRISTL. Interestingly, piCRISPR incorporates underutilized physically informed features.
Data augmentation techniques are employed by tools such as DeepCRISPR, CRISPRLearner [193], and DL-CRISPR [194]. CnnCrispr [195] utilizes the GloVe model [196], which addresses the limitations of the one-hot encoding approach for data embedding. Indels between target DNA and gRNA (RNA/DNA bulges) have been shown to contribute to off-target effects and CRISPR-Net can predict them [197]. Meaningful visualization of the results is supported by CNN-SVR [198], C-RNNCrispr [199], CRISPR-Net, CRISPR-ONT, DeepCRISTL, CRISPRedict [200], and CRISPR-M. DeepCpf1 and Cas12a_predictor support CRISPR-Cas12a systems, whereas DeepCas13 and TIGER support CRISPR-Cas13d systems. Machine learning models have enabled reliable off-target predictions for plant systems as well [201].
TnpB, an emerging next-generation genome-editing nuclease significantly smaller than conventional Cas9 and Cas12a, offers several advantages, including more efficient delivery [202, 203]. TnpB is guided by ωRNA and has distinct motif requirements in the target DNA. Recently, a deep learning model, TEEP (TnpB Editing Efficiency Predictor), has been developed to predict ωRNA activity [204]. Fig. 5 presents a feature-wise heatmap comparing learning-based tools to guide end-users in selecting the optimal option, with full details provided in Supplementary Table 3.
Figure 5.
Heatmap showing a comparative account of available learning-based gRNA designing tools. Scoring rule: 1, fully compatible; 0.5, partial support or support with limitations; 0, not compatible. Normalized scores, calculated as the average of sub-features, were used to enable a fair comparison across tools.
Prediction of Cas9-induced mutation outcome
Designing a gRNA to make a cut is just the first step toward achieving the desired edit. The next set of tools helps predict how the cell will repair the cut and what changes will result. It was a prevalent dogma that error-prone cell repair mechanisms (canonical NHEJ and MMEJ) lead to random mutations at Cas-induced DSBs. However, recent studies have invariably pointed out that such mutations are not random and are predictable [28]. Interestingly, it was shown that local sequence properties could be used to predict repair mechanisms and mutations [30, 205]. The stakes of such predictions and predictive models are very high for generating precise indels [206]. In plant systems, the study of specific DNA sequences at a target region has allowed scientists to predict the outcome of NHEJ-mediated single nucleotide indels and can introduce beneficial polymorphisms and allelic mutations [29, 207].
Many computational models have been developed to predict outcomes of non-templated DSB repair [28]. The development of models for predicting Cas9-induced mutation outcomes is intrinsically linked to the Python data science ecosystem, leveraging libraries for model training and deployment. The inDelphi is one of the earliest tools reported for the prediction of Cas9-induced indels and was experimentally validated [31]. Later, Lindel [32] and FORECasT [30] were developed for mutation prediction. Similarly, SPROUT, developed based on large experimental data, enables prediction of repair outcome [33]. The predictive accuracy of CROTON, a deep learning framework, was reported to be higher than that of SPROUT, FORECasT, and inDelphi [34]. It was also found that for the prediction of frameshift mutations, deep learning-based models such as Apindel and CROTON did not perform well [208]. FORECasT-repair extends FORECasT model to repair-deficient contexts [209]. Recently, the AIdit_DSB model was developed and demonstrated to outperform Lindel and FORECasT models for K652 cells [162]. Although a model trained exclusively on plant repair data is not available yet, Molla et al. [29] showed that Cas9-induced single nucleotide insertion can be efficiently predicted using inDelphi, FORECasT, and SPROUT. To select a tool for predicting Cas9-induced mutations, one must understand that DNA repair is not random but a predictable process influenced by local sequence context. The most valuable tools provide precise and quantitative predictions of both the class of indel and the frequency of each specific repair outcome. Advanced tools further enhance utility by forecasting the functional impact, such as frameshift mutations, and integrating cell-type-specific biological contexts. To assist users in selecting the most suitable tool, Fig. 6 presents a heatmap scoring the main features of all available tools based on their functionalities, with full details provided in Supplementary Table 4.
Figure 6.
Tools for predicting Cas9-induced mutation (indels) outcome. Scoring rule: 1, fully compatible; 0.5, partial support or support with limitations; 0, not compatible. Normalized scores, calculated as the average of sub-features, were used to enable a fair comparison across tools.
Tools for base editing and prime editing
The canonical CRISPR-Cas tools are highly efficient in generating random indels at target sites, which are useful in knocking out genes and creating random genetic variations at regulatory elements. Whereas, base editing and prime editing support precise alteration of genetic letters [210, 211]. This section covers specialized tools that can design complex gRNAs and predict outcomes for experiments aimed at correcting or rewriting specific genetic information. Computational tools for designing base and prime editing experiments are predominantly offered as web services to handle the complex design rules. These tools are built on diverse web frameworks, with back-end logic implemented in languages such as Perl and Python. To choose the optimal tool for base or prime editing experiments, a user must evaluate several critical design features.
Base editing
Base editors (BEs) are engineered by fusing a nucleotide deaminase to a nuclease-defective effector protein, mostly nCas9 or dCas9. The technique introduces precise, single nucleotide substitutions in a DNA or RNA strand without generating DSBs or indels. BE is directed to a genomic locus by a gRNA bound to the Cas enzyme, where the deaminase could edit its target base within a permissible activity window [206]. BEs mainly fall into two broad categories, DNA BEs and RNA BEs, and the DNA BEs could be further subdivided into cytosine BEs (CBEs), adenine BEs (ABEs), C-to-G BEs (CGBEs), dual-base editors, and organellar BEs [211]. The gRNA design for base editing is more complex owing to specific prerequisites for optimal activity window and suitably placed PAM and becomes more challenging when developing genome-wide gRNA libraries.
To support experimentations with BEs, user-friendly computational tools and web interfaces have been built by several groups [211]. BE-Designer is one of the earliest tools for gRNA design [26]. The beditor tool supports multiprocessing for the design of sizable gRNA libraries [25]. CRISPR-BEST is a dedicated base editing system for actinobacteria [212]. CRISPy-web 2.0 was developed to design gRNA for base editing experiments [114, 213]. BEable-GPS [214] and BE-FF [215] tools assist in choosing the most efficient BEs for correcting single nucleotide variations (SNVs). Tools that provide gRNA design assistance for plants include iSTOP [216], BE-Designer, PnB Designer [217], Betarget [218], CRISPR-BETS [219], and CRISPRbase [220]. Both Cas9- and Cas12a-based BEs are supported by BEtarget, which is accessible from the CRISPR-GE web toolkit. Tools specifically designed for BE-mediated gene knockout experiments with the induction of stop codons include iSTOP, gBIG [221], CRISPR-CBEI [222], and CRISPR-BETS. The EditABLE tool streamlines the identification of highly reliable CRISPR editors and gRNAs that are computationally validated, including base editing [223]. BEscreen [224] and CRISPR-BEasy [122] are two recently developed tools for the design of gRNA libraries for base editing screens.
The prediction of editing efficiency and outcome patterns for BEs have been made feasible with tools like DeepBaseEditor [225], BE-Hive [226], CGBE-Hive [227], CGBE-SMART [228], BE-DICT [229], Bedeepon [230], FORECasT-BE [231], Bedeepoff [232], and DeepBE [233]. DeepBaseEditor employs two models, namely DeepABE and DeepCBE. The CGBE-Hive was built by training the BE-Hive model. For ABEs and CBEs, BEdeepon enables the prediction of editing efficiency and outcome frequencies, and BEdeepoff offers off-target site prediction. Recently, DeepBE was coded to predict the editing efficiencies and outcomes of various BEs generated by different Cas9 variants.
For base editing, key criteria for selecting a tool include support for major editor types, the ability to predict both editing efficiency and product purity, definition of activity windows for each editor, options for managing bystander editing, and application-specific features such as designing guides for stop codon introduction or correction. The heatmap in Fig. 7 offers a visual guide for choosing the best tool from the comprehensive tools available, with further information provided in Supplementary Table 5.
Figure 7.
Available tools for base editing experiment designing. Scoring rule: 1, fully compatible; 0.5, partial support or support with limitations; 0, not compatible. Normalized scores, calculated as the average of sub-features, were used to enable a fair comparison across tools.
Prime editing
Even though BEs are capable of effectively introducing point mutations, they cannot generate accurate indels [210, 211, 234]. Prime editors (PEs), in contrast to BEs, allow for all 12 types of substitutions and indels, as well as their combinations, with favorable intended-editing to by-product indel ratios [9, 235–238]. The prime editing technique combines a prime editing guide RNA (pegRNA) with a nCas9 that has been fused to a reverse transcriptase [9]. Also, unlike BEs, PEs exhibit a PAM proximal activity window. A pegRNA contains three components: a spacer with scaffold, a primer binding site (PBS), and a reverse transcriptase template (RTT). An engineered pegRNA (epegRNA) addresses the limitations of pegRNA by incorporating a structured RNA motif at the 3′ terminus and has been proved to significantly improve editing efficiency [239]. The pegRNA designs, which must be unique for each edit, can be challenging, time-consuming, and tricky, and they have a significant impact on the effectiveness of prime editors [9, 27, 240]. The pegRNA’s extra components, the RTT and PBS sequence, significantly increase design complexity. PBS sequence with a melting temperature of 30°C and paired-pegRNA methods have been shown to further enhance efficiency [241].
Currently, many pegRNA design tools have been developed to accommodate various versions of PEs, most of which are web tools such as pegFinder [242], PlantPegDesigner [241], PrimeDesign [243], Primeedit [244], PE-Designer [245], Easy-Prime [246], PnB Designer [217], and EditABLE [223], whereas multicrispr [247] and PINE-CONE [248] need to be locally installed. Only a few tools provide dedicated pegRNA design support for plants. PlantPegDesigner is designed exclusively for plants, while PE-Designer and PnB Designer can also be utilized. PrimeDesign offers the provision for visualization of pegRNA secondary structure. For the design of epegRNAs, pegLIT identifies non-interfering nucleotide linkers between pegRNAs and 3′ motifs [239]. PETAL aids in the identification of all valid target sites around the sequence of interest [249]. The PRIME-Del was developed to facilitate the design of pegRNAs for achieving large deletions using the PRIME-Del method [250]. To design thousands of pegRNAs at once, the Prime Editing Guide Generator (PEGG), a free-to-use Python package, facilitates the screening of sensor libraries of genetic variants [251].
DeepPE is the first tool developed for the efficiency prediction of pegRNAs [252]. The Easy-Prime tool facilitates efficiency prediction for all small-sized edits. Later, DeepPrime emerged as an improvement upon the DeepPE model, offering enhanced accuracy [253]. DeepPrime and its variant, DeepPrime-FT, support predictions for various prime editing systems and cell types, while DeepPrime-Off offers predictions of off-target effects for PE systems [253]. The MinsePIE tool allows for the prediction of prime editing insertion efficiencies [254]. PRIDICT was developed for the prediction of pegRNA efficiency and unintended editing rates for short insertions and deletions and 1 bp replacements [255]. PRIDICT2.0 overcomes the shortcomings of its predecessor, while ePRIDICT enables pegRNA efficiency prediction based on chromatin contexts [256]. In prime editing, the central challenge lies in the complex design of the pegRNA. Therefore, the best tools are evaluated on their ability to optimize all three of its components, incorporate advanced versions of prime editors, and predict both editing efficiency and purity (Fig. 8 and Supplementary Table 6).
Figure 8.
Prime editing experiment designing tools. Scoring rule: 1, fully compatible; 0.5, partial support or support with limitations; 0, not compatible. Normalized scores, calculated as the average of sub-features, were used to enable a fair comparison across tools.
Tools exclusively for plant-based CRISPR-Cas experiments
CRISPR-Cas technology was first demonstrated in model organisms, particularly in bacteria and animals [257, 258], and soon extended to plants [259]. This section covers tools developed exclusively to overcome challenges unique to plant genome editing, such as handling multiple gene copies (polyploidy), ensuring the CRISPR machinery works efficiently in plant cells, and editing several genes at once.
CRISPR-P is noted to be the first plant-specific gRNA design tool [260]. The CRISPR-PLANT, which offers a database, supports model plants and other important agricultural crops [261]. The CRISPR Genome Analysis Tool (CGAT) provides off-target prediction and also ranks the potential target sites [262]. CRISPR-P 2.0 [263] is an updated version of the CRISPR-P, whereas CRISPR-PLANT v2 [264] is an updated version of CRISPR-PLANT, with improved off-target detection strategies. WheatCRISPR aids in specific gRNA design for editing the polyploid genome of wheat [265]. CRISPR-Local is well optimized for non-reference genomes and targeting multiple genes at once [266]. PhytoCRISP-Ex allows designing guide RNAs for algal genomes [131]. The gRNA sequence and the chromatin state of the target site are important factors governing CRISPR-Cas editing efficiency [267]. CRISPR-Cereal accounts for chromatin accessibility and single-nucleotide polymorphisms (SNPs) in the target region for the three major food crops—wheat, maize, and rice [268]. GB4.0 [269, 270], integrated into the GoldenBraid platform [271], provides tools to design and assemble CRISPR-Cas single guide and multiplex guide RNA constructs either for gene editing or gene regulation.
To select the most suitable tool for plant genome editing, researchers must consider features that address the unique challenges of plant biology. Specialized support for polyploid genomes and the ability to design guides that are specific to homoeologous genes are important. Advanced features include support for genomic target context annotation and genome-wide off-target identification. Finally, tool utility is enhanced by its support for multiplex gRNA design, which is critical for studying complex traits governed by multiple genes. A comparative user guide is given in Fig. 9 and Supplementary Table 7.
Figure 9.
Exclusive tools for plants to design canonical CRISPR-Cas experiments. PlantPegDesigner is not included here; it is listed under prime editing tools. Scoring rule: 1, fully compatible; 0.5, partial support or support with limitations; 0, not compatible. Normalized scores, calculated as the average of sub-features, were used to enable a fair comparison across tools.
Tools for downstream analysis and visualization
After an experiment is performed in the lab, it is crucial to verify the outcome, which is the purpose of this final section of tools. They differ from all preceding design tools by analyzing experimental sequencing data to quantify the efficiency and precision of the edits, thus validating the entire workflow. Mutations induced by CRISPR-Cas9 often result in uniform biallelic or heterozygous conditions in the first transgenic population of diploid organisms. However, in many cases, chimeric mutations occur frequently, where three or more allelic edits are present within a single individual [272, 273]. Identifying the mutated sequences of the targeted alleles is crucial. However, when using the Sanger method, the mutation sites produce overlapping sequencing peaks. Accurate identification through cloning the mutation-containing amplicons and sequencing multiple clones for each target site is labor-intensive, costly, and time-consuming. Issues like sequencing inconsistencies, variable sequence quality, ambiguous indel alignments, and deconvolution of mixed HDR-NHEJ outcomes further complicate the analysis [274]. This complexity is further exacerbated by the limitations of Sanger sequencing for evaluating a range of indels in large cell populations, as well as the analytical challenges posed by pooled experiments where different target sites are part of a single sequencing library. Therefore, simple and quick methods to accurately characterize and quantify the induced mutations become essential.
Many computational tools have been developed to address these challenges and simplify the analysis of CRISPR edits, as listed in Fig. 10. These tools are mostly equipped for the analysis of data pertaining to specific editing results, such as the NHEJ and HDR data, base editing and prime editing data, and pooled CRISPR screening data. To select the best tool for analyzing CRISPR editing outcomes, a user must first consider the type of experimental data generated, primarily Sanger sequencing for single clones or Next-Generation Sequencing (NGS) for pooled populations. The core analytical capabilities are then evaluated based on the tool’s ability to accurately quantify the frequency of different editing events, including NHEJ-mediated indels, HDR-templated repairs, and the specific substitutions generated by base or prime editing. Finally, the overall utility of a tool is determined by its support for high-throughput batch processing and its capacity to generate clear, informative visualizations, such as indel distribution plots and allele frequency charts.
Figure 10.
Tools for data analysis of different categories of editing experiments.
Tools for NHEJ and HDR data analysis
These tools are designed to quantify the outcomes of conventional CRISPR editing by analyzing sequencing data for indels and HDR events. They are essential for measuring the efficiency of standard gene knockout or knock-in experiments. CRISPR-GA [35] is the first computational tool with web support, whereas TIDE [36] offers a rapid computational method that identifies the predominant types of indels and quantifies the efficiency of genome editing. However, TIDE is limited in its applicability to templated genome editing via HDR; to address this, it was updated to TIDER [275]. From Sanger sequencing data, TIDER estimates templated point mutations and small indels and can also assist in quantifying non-templated indels.
CrispRVariants allows mutation analysis of CRISPR-Cas9 experimental data and is made available as a web tool that supports small-scale experiments, named CrispRVariantsLite [276]. BATCH-GE aids in the detection of Cas9-induced indel mutations and supports batch analysis [277]. CRISPR-DAV supports the detection and analysis of small and large indels [278]. DSDecode is used to decode Sanger sequencing chromatograms for genotyping targeted mutations, but it has limitations when analyzing chromatograms from edits obtained from PCR amplicons [279]. DSDecodeM, available under the CRISPR-GE software package and built over DSDecode, addresses these drawbacks and offers enhanced proficiency [132]. The Cas-Analyzer allows for genome editing assessment with support for paired nucleases [280]. The Hi-TOM allows for high-throughput identification of all types of mutations induced by CRISPR-Cas systems [281]. Also, CRIS.py [282] and CRISPRpic [283] facilitate downstream data analysis. DECODR is a versatile web tool capable of even analyzing indels of length ≥ 50 bases around every edit site from single guide or multi-guide CRISPR-Cas experiments [284].
CRISPAltRations is a cloud-hosted software tool available as the back-end tool of the rhAmpSeq™ CRISPR Analysis Tool provided by the IDT web platform [285]. It aids in the batch analysis of single and multiplex samples (up to 500 targets) at the same time. ICE tool provides quick and accurate analysis and is a simple-to-use software program available on the Synthego platform [286]. The edge of ICE is that it does not require uploading a full reference genome. However, the ICE tool may sometimes misrepresent templated editing data, as reported earlier [29]. Further, the CRISPRroots aids in off-target and on-target assessment of edits from RNA-seq data [287]. The CRISPR-GRANT offers a graphical user interface (GUI) for high-throughput assessment of indels from single or pooled amplicons and NGS data, and is widely accepted [288]. CrisprStitch is one of the latest tools for analyzing CRISPR-Cas editing that offers fast and efficient evaluation [289]. This serverless application allows researchers to analyze data directly in their web browser and supports the calculation of mutation frequency, editing efficiency, accuracy, and even visualization of the results.
Tools like TIDE, TIDER, DSDecodeM, DECODR, and ICE support Sanger sequencing data, while CRISPR-GA, BATCH-GE, CRISPR-DAV, Cas-Analyzer, Hi-TOM, CRIS.py, CRISPRpic, CRISPAltRations, CRISPRroots, CRISPR-GRANT, and CrisprStitch support high-throughput sequencing data for analysis and visualization. However, AGEseq [290], ampliconDIVider [291], and CrispRVariants support both types of datasets. The tools Cas-Analyzer, CRISPRMatch [292], DECODR, CRISPAltRations, and CrisprStitch offer support for both Cas9 and Cas12a nucleases. There are a number of tools available that support the analysis of HDR events along with batch analysis and visualization, including BATCH-GE, CRISPR-DAV, CRISPRMatch, CRISPRpic, CRISPAltRations, ICE, and CrisprStitch.
Tools for base editing and prime editing data analysis
Some tools are capable of extending their analytical competency beyond conventional CRISPR-Cas experimental data. This specialized group of tools analyzes precise nucleotide substitutions and complex edits generated by base and prime editors. They are necessary because the editing outcomes are different from the indels created by canonical CRISPR, requiring unique algorithms to assess substitution patterns and frequencies. The CRIS.py tool offers an assessment of multiple sequence modifications induced by base editing. Whereas the CRISPResso2 [37] suite, an updated and widely accepted version of CRISPResso [274], supports batch analysis of data from Cas9 knockout and knock-in, base editing, and prime editing experiments under a single platform that no other software packages facilitate. It allows for the estimation, visualization, and comparison of mutations from pooled amplicon experiments and NGS data sets. The ampliCan tool aids the analysis and visualization of indels, template-based repair, and base editing experiments [293]. AlleleProfileR supports NHEJ, HDR, and base editing data analysis [294]. Although CRISPResso2, ampliCan, and AlleleProfileR provide evaluations of substitution frequency, they were not developed exclusively for CRISPR-mediated substitution experiments. CRISPR-Sub is a dedicated tool built solely for the purpose of systematic assessment of substitution patterns and frequencies from the NGS datasets [295]. Hence, CRISPR-Sub could be useful in the study of base editing and prime editing experiments. At the same time, base editing outcome analysis is supported by specialized tools like BEEP [296], EditR [297], BE-Analyzer [26], and BEAT [298]. BEEP, BEAT, and EditR take Sanger sequencing data as input, whereas BE-Analyzer supports NGS data. BE-Analyzer allows for defining more parameters while performing the analysis when compared to other tools. Nevertheless, PE-Analyzer is the dedicated tool available for the assessment and visualization of insertions, deletions, and base conversions from prime editing results [245]. PE-Analyzer aids in the batch analysis of NGS data without the need for uploading data to the server. The tools CRISPR-Sub, BE-Analyzer, and PE-Analyzer are accessible from the CRISPR-RGEN tools platform.
Tools for pooled CRISPR screening data analysis
The emergence of CRISPR-Cas technology has brought a significant rise in pooled screening data [302]. Tools in this category are essential for the analysis of data from large-scale screens where thousands of genes are targeted simultaneously. They identify which gRNAs are enriched or depleted in the cell population, allowing researchers to link specific genes to cellular functions or phenotypes. Among the available tools, only PinAPL-Py [299], CRISPR-SURF [300], CRISPRcloud2 [301, 302], and VISPR-online [38] offer user-friendly web interfaces for analysis and visualization. The VISPR-online aids in the visualization of CRISPR screen outputs generated by MAGeCK [303], BAGEL [304], and JACKS [305], and thereby streamlines the data evaluation process. CRISPRO supports the analysis and visualization of saturating mutagenesis CRISPR screens from NGS data [306]. BAGEL2 is another popular tool with enhanced performance, specificity, sensitivity, and performance [304, 307]. MAGeCKFlute, an improvement over its earlier version, demonstrates better performance in downstream functional data analysis [303, 308, 309].
Most of the tools are optimized for coding regions with clear functional boundaries and thus are not capable of analyzing tiling screens in non-coding regions where the exact locations of functional sequences are unknown. However, this challenge is addressed by CRISPR-SURF and RELICS [310]. MAUDE also takes care of some parts of non-coding screen evaluation [311]. The ecosystem for downstream analysis of editing outcomes is characterized by two major platforms. The comprehensive pipelines such as CRISPResso2 are typically developed in Python, while tools focused on statistical analysis and visualization, like ampliCan, are often implemented as R packages within the Bioconductor framework. To empower end-users to make an informed choice from numerous options, we present a comparative heatmap in Supplementary File 8, and complete details are documented in Supplementary Table 9.
Web resource development
One of the key challenges in the CRISPR-Cas research landscape is the lack of a comprehensive, centralized online platform that systematically consolidates the wide array of computational tools used throughout the entire workflow—from exploring CRISPR-Cas systems and designing experiments to conducting downstream data analysis. This study addresses that gap by developing an online resource, CRISPR-GATE (Fig. 11), which is planned to be continually updated to meet the evolving needs of the research community. CRISPR-GATE is a meticulously curated repository that offers streamlined access to over 250 CRISPR-Cas tools that are designed by various groups to meet the specific needs of researchers in the field.
Figure 11.
Overview of the CRISPR-GATE web repository. Snapshot showing (a) home page of CRISPR-GATE web resource; (b–d) multiple tools available under different categories.
CRISPR-GATE provides a user-friendly interface, making it an invaluable resource for scientists working with genome editing technologies, from editing tool development to applications in basic biology, medicine, and agriculture. The front-end architecture is built using HTML, CSS, and JavaScript, while the back-end leverages PHP and a MySQL database. A standout feature of CRISPR-GATE is its intuitive browsing and filtering functionality, allowing users to quickly locate relevant tools based on specific categories. This efficiency not only saves researchers’ significant time and effort but also enables them to focus on their experimental goals without the burden of extensive searches. CRISPR-GATE categorizes tools into eight primary areas of application, each accompanied by subcategory lists and filter options for enhanced navigation. Rather than merely listing available tools, CRISPR-GATE goes further by providing detailed insights into each tool’s key features and limitations. Additionally, users can sort tools by several criteria, such as popularity, which is based on the total citation counts as indexed by Google Scholar. All this critical information empowers researchers to make informed decisions about the most suitable and relevant tools for their specific projects. The CRISPR-GATE web resource, its contents, and all associated data are made available under the ICAR Data Use License. The full terms of the license can be viewed on the resource’s website.
Conclusion and future direction
The rapid development of reliable and user-friendly tools has fueled the realization of the potential of CRISPR-Cas technology in transforming the domain of genome manipulation in diverse organisms. Currently, the majority of biologists use CRISPR-Cas tools to add desired dimensions and prove their hypothesis in a short span of time. The contributions of computer programming experts and bioinformaticians have made CRISPR-Cas technologies more easily accessible, overcoming earlier limitations associated with genome editing technologies like ZFNs and TALENs. The simplicity of targeting genomic areas has enabled its exponential adoption across labs and institutions. Web resources have further accelerated the adoption by reducing complexity and computational demands.
Most of the tools available for the detection of CRISPR-Cas systems do not output unanimous results and development of more accurate tools is still underway. The detection of novel Cas proteins and the delineation of the functions of auxiliary Cas proteins provide an attractive direction for future research. Although specific tools already available for the identification of CRISPR-Cas systems from metagenomics data, they still struggle to cover the environmental sample heterogeneity, highlighting the need for robust web databases and tools to manage large dataset and associated computational burden. Additionally, developing new tools to mine microbial genomes for emerging genome editor nucleases like TnpB, IscB will facilitate enriching genome editing reagents.
Improvement of gRNA efficiency and specificity remains an active research area, and the use of advanced machine learning algorithms that can handle and analyze complex datasets could help in making better decisions. Deep learning holds promise for high-impact predictions once sufficiently large datasets become available. Incorporation of features such as folding pattern analysis and target association with nucleosome could enhance gRNA design. Plant-specific, learning-based tools for predicting Cas9-induced mutations are lacking, and validated web tools for NHEJ outcome prediction are in high demand. Moreover, underfunded labs would benefit from tools that can accurately quantify edit types and percentages from simple Sanger chromatograms.
Downstream analysis tools could perform better through machine learning integration, improved sequencing methodologies, and enhanced noise filtration techniques. More web tools are needed to support designing for diverse naturally available and engineered nucleases. Tools for designing BE and PE experiments encompassing diverse genomes are the need of the time. Nevertheless, researchers often struggle to find the most suitable tool for their experiments, even when multiple options are available. The CRISPR-GATE web resource streamlines this process and would speed up and democratize various applications of CRISPR-Cas technology across multiple disciplines.
Key Points
CRISPR-GATE is a comprehensive, user-friendly online repository that brings together computational resources for each step of CRISPR-Cas genome editing, from identifying Cas proteins to designing gRNAs and analyzing results.
This article systematically classifies available computational tools (standalone and web-based), helping researchers quickly select the best tools tailored to their experimental needs.
Detailed coverage is given to computational tools for multiple CRISPR applications, including traditional gene knockout, base and prime editing, and precise mutation outcome prediction and downstream analysis.
The article highlights the recent integration of machine learning and deep learning techniques, significantly improving predictions of gRNA efficiency and reducing off-target risks.
CRISPR-GATE aims to streamline research by saving time, reducing complexity, and promoting innovation in genome editing by offering easy access to categorized and regularly updated computational resources.
Supplementary Material
Acknowledgements
Research was supported by the Indian Council of Agricultural Research, Department of Agricultural Research and Education, Government of India. K.M., M.J.B, and MD would like to acknowledge funding from the Indian Council of Agricultural Research (ICAR), New Delhi, in the form of the Plan Scheme—‘Incentivizing Research in Agriculture’ project. The financial grants to DABin, ICAR-IASRI, and DBT-JRF fellowship to AAV are duly acknowledged. SP would like to acknowledge funding from DST-INSPIRE, New Delhi. We also appreciate the support from the Director, ICAR-CRRI and Director, ICAR-IASRI.
Contributor Information
Asif Ali Vadakkethil, The Graduate School, ICAR-Indian Agricultural Research Institute, PUSA Campus, New Delhi 110012, India; Division of Agricultural Bioinformatics, ICAR-Indian Agricultural Statistics Research Institute, Library Avenue, New Delhi 110012, India.
Sonali Panda, ICAR- Central Rice Research Institute, Cuttack 753006, Odisha, India; Department of Botany, Ravenshaw University, Cuttack 753003, Odisha, India.
Aranya Mitra, Institute of Health Sciences, Presidency University, Kolkata, West Bengal 700156, India.
Manaswini Dash, ICAR- Central Rice Research Institute, Cuttack 753006, Odisha, India.
Mirza J Baig, ICAR- Central Rice Research Institute, Cuttack 753006, Odisha, India.
Ulavappa B Angadi, Division of Agricultural Bioinformatics, ICAR-Indian Agricultural Statistics Research Institute, Library Avenue, New Delhi 110012, India.
Dinesh Kumar, Division of Agricultural Bioinformatics, ICAR-Indian Agricultural Statistics Research Institute, Library Avenue, New Delhi 110012, India.
Sarika Jaiswal, Division of Agricultural Bioinformatics, ICAR-Indian Agricultural Statistics Research Institute, Library Avenue, New Delhi 110012, India.
Mir Asif Iquebal, Division of Agricultural Bioinformatics, ICAR-Indian Agricultural Statistics Research Institute, Library Avenue, New Delhi 110012, India.
Kutubuddin A Molla, ICAR- Central Rice Research Institute, Cuttack 753006, Odisha, India.
Author contributions
Kutubuddin A. Molla, Mir Asif Iquebal, and Sarika Jaiswa conceptualized,designed and supervised the study, and provided resources. Asif Ali Vadakkethil, Sonali Panda, Aranya Mitra, and Manaswini Dash did the data curation and analysis. Asif Ali Vadakkethil and Ulavappa. B. Angadi were involved in web resource and software development. Asif Ali Vadakkethil, Sonali Panda, Kutubuddin A. Molla, Aranya Mitra, Mir Asif Iquebal, and Sarika Jaiswal wrote the first draft of the manuscript. Kutubuddin A. Molla, Mir Asif Iquebal, Sarika Jaiswa, Dinesh Kumar, and Mirza J. Baig administered the project, reviewed and edited the manuscript. All authors contributed to the article and approved the submitted version.
Conflict of interest
None declared.
Funding
Indian Council of Agricultural Research (ICAR), New Delhi.
Data availability
Data presented in the study are included as tables in the Supplementary material. All supplementary materials are available via Zenodo at https://doi.org/10.5281/zenodo.15254840.
References
- 1. Ran FA, Hsu PD, Lin C-Y. et al. Double nicking by RNA-guided CRISPR Cas9 for enhanced genome editing specificity. Cell 2013;154:1380–9. 10.1016/j.cell.2013.08.021 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2. Barrangou R, Doudna JA. Applications of CRISPR technologies in research and beyond. Nat Biotechnol 2016;34:933–41. 10.1038/nbt.3659 [DOI] [PubMed] [Google Scholar]
- 3. Wright AV, Nuñez JK, Doudna JA. Biology and applications of CRISPR systems: harnessing Nature’s toolbox for genome engineering. Cell 2016;164:29–44. 10.1016/j.cell.2015.12.035 [DOI] [PubMed] [Google Scholar]
- 4. Maeder ML, Linder SJ, Cascio VM. et al. CRISPR RNA–guided activation of endogenous human genes. Nat Methods 2013;10:977–9. 10.1038/nmeth.2598 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5. Qi LS, Larson MH, Gilbert LA. et al. Repurposing CRISPR as an RNA-guided platform for sequence-specific control of gene expression. Cell 2013;152:1173–83. 10.1016/j.cell.2013.02.022 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6. Gaudelli NM, Komor AC, Rees HA. et al. Programmable base editing of A•T to G•C in genomic DNA without DNA cleavage. Nature 2017;551:464–71. 10.1038/nature24644 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7. Komor AC, Kim YB, Packer MS. et al. Programmable editing of a target base in genomic DNA without double-stranded DNA cleavage. Nature 2016;533:420–4. 10.1038/nature17946 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8. Nishida K, Arazoe T, Yachie N. et al. Targeted nucleotide editing using hybrid prokaryotic and vertebrate adaptive immune systems. Science (1979) 2016;353:aaf8729. 10.1126/science.aaf8729 [DOI] [PubMed] [Google Scholar]
- 9. Anzalone AV, Randolph PB, Davis JR. et al. Search-and-replace genome editing without double-strand breaks or donor DNA. Nature 2019;576:149–57. 10.1038/s41586-019-1711-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10. Hilton IB, D’Ippolito AM, Vockley CM. et al. Epigenome editing by a CRISPR-Cas9-based acetyltransferase activates genes from promoters and enhancers. Nat Biotechnol 2015;33:510–7. 10.1038/nbt.3199 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11. Doench JG. Am I ready for CRISPR? A user’s guide to genetic screens. Nat Rev Genet 2018;19:67–80. 10.1038/nrg.2017.97 [DOI] [PubMed] [Google Scholar]
- 12. Morgan SL, Mariano NC, Bermudez A. et al. Manipulation of nuclear architecture through CRISPR-mediated chromosomal looping. Nat Commun 2017;8:15993. 10.1038/ncomms15993 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13. Wang H, Xu X, Nguyen CM. et al. CRISPR-mediated programmable 3D genome positioning and nuclear organization. Cell 2018;175:1405–17. 10.1016/j.cell.2018.09.013 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14. Ma H, Naseri A, Reyes-Gutierrez P. et al. Multicolor CRISPR labeling of chromosomal loci in human cells. Proc Natl Acad Sci 2015;112:3002–7. 10.1073/pnas.1420024112 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15. Yang L-Z, Wang Y, Li S-Q. et al. Dynamic imaging of RNA in living cells by CRISPR-Cas13 systems. Mol Cell 2019;76:981–997.e7. 10.1016/j.molcel.2019.10.024 [DOI] [PubMed] [Google Scholar]
- 16. Abudayyeh OO, Gootenberg JS, Essletzbichler P. et al. RNA targeting with CRISPR–Cas13. Nature 2017;550:280–4. 10.1038/nature24049 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17. Gootenberg JS, Abudayyeh OO, Lee JW. et al. Nucleic acid detection with CRISPR-Cas13a/C2c2. Science 1979;2017:438–42. 10.1126/science.aam9321 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18. Kyrou K, Hammond AM, Galizi R. et al. A CRISPR–Cas9 gene drive targeting doublesex causes complete population suppression in caged Anopheles gambiae mosquitoes. Nat Biotechnol 2018;36:1062–6. 10.1038/nbt.4245 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19. Lange SJ, Alkhnbashi OS, Rose D. et al. CRISPRmap: an automated classification of repeat conservation in prokaryotic adaptive immune systems. Nucleic Acids Res 2013;41:8034–44. 10.1093/nar/gkt606 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20. Couvin D, Bernheim A, Toffano-Nioche C. et al. CRISPRCasFinder, an update of CRISRFinder, includes a portable version, enhanced performance and integrates search for Cas proteins. Nucleic Acids Res 2018;46:W246–51. 10.1093/nar/gky425 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21. Abby SS, Néron B, Ménager H. et al. MacSyFinder: a program to mine genomes for molecular systems with an application to CRISPR-Cas systems. PloS One 2014;9:e110726. 10.1371/journal.pone.0110726 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22. Eitzinger S, Asif A, Watters KE. et al. Machine learning predicts new anti-CRISPR proteins. Nucleic Acids Res 2020;48:4698–708. 10.1093/nar/gkaa219 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23. Konstantakos V, Nentidis A, Krithara A. et al. CRISPR–Cas9 gRNA efficiency prediction: an overview of predictive tools and the role of deep learning. Nucleic Acids Res 2022;50:3616–37. 10.1093/nar/gkac192 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24. Schindele P, Wolter F, Puchta H. CRISPR Guide RNA Design Guidelines for Efficient Genome Editing. In: Heinlein M. (eds) RNA Tagging 2020. Methods in Molecular Biology, 2166;331–42. Humana, New York, NY. 10.1007/978-1-0716-0712-1_19 [DOI] [PubMed] [Google Scholar]
- 25. Dandage R, Després PC, Yachie N. et al. beditor: a computational workflow for designing libraries of guide RNAs for CRISPR-Mediated Base editing. Genetics 2019;212:377–85. 10.1534/genetics.119.302089 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26. Hwang G-H, Park J, Lim K. et al. Web-based design and analysis tools for CRISPR base editing. BMC Bioinformatics 2018;19:542. 10.1186/s12859-018-2585-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27. Lin Q, Zong Y, Xue C. et al. Prime genome editing in rice and wheat. Nat Biotechnol 2020;38:582–5. 10.1038/s41587-020-0455-x [DOI] [PubMed] [Google Scholar]
- 28. Molla KA, Yang Y. Predicting CRISPR/Cas9-induced mutations for precise genome editing. Trends Biotechnol 2020;38:136–41. 10.1016/j.tibtech.2019.08.002 [DOI] [PubMed] [Google Scholar]
- 29. Molla KA, Shih J, Wheatley MS. et al. Predictable NHEJ insertion and assessment of HDR editing strategies in plants. Front Genome Ed 2022;4:4. 10.3389/fgeed.2022.825236 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30. Allen F, Crepaldi L, Alsinet C. et al. Predicting the mutations generated by repair of Cas9-induced double-strand breaks. Nat Biotechnol 2019;37:64–72. 10.1038/nbt.4317 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31. Shen MW, Arbab M, Hsu JY. et al. Predictable and precise template-free CRISPR editing of pathogenic variants. Nature 2018;563:646–51. 10.1038/s41586-018-0686-x [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32. Chen W, McKenna A, Schreiber J. et al. Massively parallel profiling and predictive modeling of the outcomes of CRISPR/Cas9-mediated double-strand break repair. Nucleic Acids Res 2019;47:7989–8003. 10.1093/nar/gkz487 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33. Leenay RT, Aghazadeh A, Hiatt J. et al. Large dataset enables prediction of repair after CRISPR–Cas9 editing in primary T cells. Nat Biotechnol 2019;37:1034–7. 10.1038/s41587-019-0203-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34. Li VR, Zhang Z, Troyanskaya OG. CROTON: an automated and variant-aware deep learning framework for predicting CRISPR/Cas9 editing outcomes. Bioinformatics 2021;37:i342–8. 10.1093/bioinformatics/btab268 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35. Güell M, Yang L, Church GM. Genome editing assessment using CRISPR genome Analyzer (CRISPR-GA). Bioinformatics 2014;30:2968–70. 10.1093/bioinformatics/btu427 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36. Brinkman EK, Chen T, Amendola M. et al. Easy quantitative assessment of genome editing by sequence trace decomposition. Nucleic Acids Res 2014;42:e168–8. 10.1093/nar/gku936 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37. Clement K, Rees H, Canver MC. et al. CRISPResso2 provides accurate and rapid genome editing sequence analysis. Nat Biotechnol 2019;37:224–6. 10.1038/s41587-019-0032-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38. Cui Y, Wang Z, Köster J. et al. VISPR-online: a web-based interactive tool to visualize CRISPR screening experiments. BMC Bioinformatics 2021;22:344. 10.1186/s12859-021-04275-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39. Torres-Perez R, Garcia-Martin JA, Montoliu L. et al. WeReview: CRISPR tools—live repository of computational tools for assisting CRISPR/Cas experiments. Bioengineering 2019;6:63. 10.3390/bioengineering6030063 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40. Alipanahi R, Safari L, Khanteymoori A. CRISPR genome editing using computational approaches: a survey. Frontiers in Bioinformatics 2023;2:1001131. 10.3389/fbinf.2022.1001131 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41. Li C, Chu W, Gill RA. et al. Computational tools and resources for CRISPR/Cas genome editing. Genomics Proteomics Bioinformatics 2023;21:108–26. 10.1016/j.gpb.2022.02.006 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42. Altschul SF, Gish W, Miller W. et al. Basic local alignment search tool. J Mol Biol 1990;215:403–10. 10.1016/S0022-2836(05)80360-2 [DOI] [PubMed] [Google Scholar]
- 43. Eddy SR. Profile hidden Markov models. Bioinformatics 1998;14:755–63. 10.1093/bioinformatics/14.9.755 [DOI] [PubMed] [Google Scholar]
- 44. Dsouza M, Larsen N, Overbeek R. Searching for patterns in genomic data. Trends Genet 1997;13:497–8. 10.1016/S0168-9525(97)01347-4 [DOI] [PubMed] [Google Scholar]
- 45. Edgar RC. PILER-CR: fast and accurate identification of CRISPR repeats. BMC Bioinformatics 2007;8:18. 10.1186/1471-2105-8-18 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46. Bland C, Ramsey TL, Sabree F. et al. CRISPR recognition tool (CRT): a tool for automatic detection of clustered regularly interspaced palindromic repeats. BMC Bioinformatics 2007;8:209. 10.1186/1471-2105-8-209 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47. Grissa I, Vergnaud G, Pourcel C. CRISPRFinder: a web tool to identify clustered regularly interspaced short palindromic repeats. Nucleic Acids Res 2007;35:W52–7. 10.1093/nar/gkm360 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48. Biswas A, Staals RHJ, Morales SE. et al. CRISPRDetect: a flexible algorithm to define CRISPR arrays. BMC Genomics 2016;17:356. 10.1186/s12864-016-2627-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49. Ge R, Mai G, Wang P. et al. CRISPRdigger: detecting CRISPRs with better direct repeat annotations. Sci Rep 2016;6:32942. 10.1038/srep32942 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50. Wang K, Liang C. CRF: detection of CRISPR arrays using random forest. PeerJ 2017;5:e3219. 10.7717/peerj.3219 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51. Mitrofanov A, Alkhnbashi OS, Shmakov SA. et al. CRISPRidentify: identification of CRISPR arrays using machine learning approach. Nucleic Acids Res 2021;49:e20–0. 10.1093/nar/gkaa1158 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52. Nethery MA, Korvink M, Makarova KS. et al. CRISPRclassify: repeat-based classification of CRISPR loci. CRISPR J 2021;4:558–74. 10.1089/crispr.2021.0021 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53. Biswas A, Fineran PC, Brown CM. Accurate computational prediction of the transcribed strand of CRISPR non-coding RNAs. Bioinformatics 2014;30:1805–13. 10.1093/bioinformatics/btu114 [DOI] [PubMed] [Google Scholar]
- 54. Alkhnbashi OS, Costa F, Shah SA. et al. CRISPRstrand: predicting repeat orientations to determine the crRNA-encoding strand at CRISPR loci. Bioinformatics 2014;30:i489–96. 10.1093/bioinformatics/btu459 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55. Milicevic O, Repac J, Bozic B. et al. A simple criterion for inferring CRISPR Array direction. Front Microbiol 2019;10:10. 10.3389/fmicb.2019.02054 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56. Alkhnbashi OS, Shah SA, Garrett RA. et al. Characterizing leader sequences of CRISPR loci. Bioinformatics 2016;32:i576–85. 10.1093/bioinformatics/btw454 [DOI] [PubMed] [Google Scholar]
- 57. Koonin EV, Makarova KS, Zhang F. Diversity, classification and evolution of CRISPR-Cas systems. Curr Opin Microbiol 2017;37:67–78. 10.1016/j.mib.2017.05.008 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58. Makarova KS, Wolf YI, Alkhnbashi OS. et al. An updated evolutionary classification of CRISPR–Cas systems. Nat Rev Microbiol 2015;13:722–36. 10.1038/nrmicro3569 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59. Makarova KS, Wolf YI, Iranzo J. et al. Evolutionary classification of CRISPR–Cas systems: a burst of class 2 and derived variants. Nat Rev Microbiol 2020;18:67–83. 10.1038/s41579-019-0299-x [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60. Chai G, Yu M, Jiang L. et al. HMMCAS: a web tool for the identification and domain annotations of CAS proteins. IEEE/ACM Trans Comput Biol Bioinform 2019;16:1313–5. 10.1109/TCBB.2017.2665542 [DOI] [PubMed] [Google Scholar]
- 61. Padilha VA, Alkhnbashi OS, Shah SA. et al. CRISPRcasIdentifier: machine learning for accurate identification and classification of CRISPR-Cas systems. Gigascience 2020;9:1–12. 10.1093/gigascience/giaa062 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62. Padilha VA, Alkhnbashi OS, Tran VD. et al. Casboundary: automated definition of integral Cas cassettes. Bioinformatics 2021;37:1352–9. 10.1093/bioinformatics/btaa984 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63. Yang S, Huang J, He B. CASPredict: a web service for identifying Cas proteins. PeerJ 2021;9:e11887. 10.7717/peerj.11887 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64. Zhang T, Jia Y, Li H. et al. CRISPRCasStack: a stacking strategy-based ensemble learning framework for accurate identification of Cas proteins. Brief Bioinform 2022;23:bbac335. 10.1093/bib/bbac335 [DOI] [PubMed] [Google Scholar]
- 65. Zhang Q, Doak TG, Ye Y. Expanding the catalog of cas genes with metagenomes. Nucleic Acids Res 2014;42:2448–59. 10.1093/nar/gkt1262 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66. Tang Z, Chen S, Chen A. et al. CasPDB: an integrated and annotated database for Cas proteins from bacteria and archaea. Database 2019;2019. 10.1093/database/baz093 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67. Park H, Won J, Park Y. et al. CRISPR-Cas-Docker: web-based in silico docking and machine learning-based classification of crRNAs with Cas proteins. BMC Bioinformatics 2023;24:167. 10.1186/s12859-023-05296-y [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68. Adler BA, Trinidad MI, Bellieny-Rabelo D. et al. CasPEDIA database: a functional classification system for class 2 CRISPR-Cas enzymes. Nucleic Acids Res 2024;52:D590–6. 10.1093/nar/gkad890 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69. Crawley AB, Henriksen JR, Barrangou R. CRISPRdisco: An automated pipeline for the discovery and analysis of CRISPR-Cas systems. CRISPR J 2018;1:171–81. 10.1089/crispr.2017.0022 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70. Russel J, Pinilla-Redondo R, Mayo-Muñoz D. et al. CRISPRCasTyper: automated identification, annotation, and classification of CRISPR-Cas loci. CRISPR J 2020;3:462–9. 10.1089/crispr.2020.0059 [DOI] [PubMed] [Google Scholar]
- 71. Alkhnbashi OS, Mitrofanov A, Bonidia R. et al. CRISPRloci: comprehensive and accurate annotation of CRISPR–Cas systems. Nucleic Acids Res 2021;49:W125–30. 10.1093/nar/gkab456 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72. Raden M, Ali SM, Alkhnbashi OS. et al. Freiburg RNA tools: a central online resource for RNA-focused research and teaching. Nucleic Acids Res 2018;46:W25–9. 10.1093/nar/gky329 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73. Rousseau C, Gonnet M, Le Romancer M. et al. CRISPI: a CRISPR interactive database. Bioinformatics 2009;25:3317–8. 10.1093/bioinformatics/btp586 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74. Zhang Q, Ye Y. Not all predicted CRISPR–Cas systems are equal: isolated cas genes and classes of CRISPR like elements. BMC Bioinformatics 2017;18:92. 10.1186/s12859-017-1512-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75. Zhang F, Zhao S, Ren C. et al. CRISPRminer is a knowledge base for exploring CRISPR-Cas systems in microbe and phage interactions. Commun Biol 2018;1:180. 10.1038/s42003-018-0184-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76. Pourcel C, Touchon M, Villeriot N. et al. CRISPRCasdb a successor of CRISPRdb containing CRISPR arrays and cas genes from complete genome sequences, and tools to download and query lists of repeats and spacers. Nucleic Acids Res 2019;48:D535–44. 10.1093/nar/gkz915 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77. Grissa I, Vergnaud G, Pourcel C. The CRISPRdb database and tools to display CRISPRs and to generate dictionaries of spacers and repeats. BMC Bioinformatics 2007;8:172. 10.1186/1471-2105-8-172 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78. Zhou F, Yu X, Gan R. et al. CRISPRimmunity: an interactive web server for CRISPR-associated important molecular events and modulators used in geNome edIting tool identifYing. Nucleic Acids Res 2023;51:W93–107. 10.1093/nar/gkad425 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79. Rho M, Wu Y-W, Tang H. et al. Diverse CRISPRs evolving in human microbiomes. PLoS Genet 2012;8:e1002441. 10.1371/journal.pgen.1002441 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80. Skennerton CT, Imelfort M, Tyson GW. Crass: identification and reconstruction of CRISPR from unassembled metagenomic data. Nucleic Acids Res 2013;41:e105–5. 10.1093/nar/gkt183 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81. Lei J, Sun Y. Assemble CRISPRs from metagenomic sequencing data. Bioinformatics 2016;32:i520–8. 10.1093/bioinformatics/btw456 [DOI] [PubMed] [Google Scholar]
- 82. Moller AG, Liang C. MetaCRAST: reference-guided extraction of CRISPR spacers from unassembled metagenomes. PeerJ 2017;5:e3788. 10.7717/peerj.3788 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83. Cebrian-Serrano A, Davies B. CRISPR-Cas orthologues and variants: optimizing the repertoire, specificity and delivery of genome engineering tools. Mamm Genome 2017;28:247–61. 10.1007/s00335-017-9697-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84. Karvelis T, Gasiunas G, Young J. et al. Rapid characterization of CRISPR-Cas9 protospacer adjacent motif sequence elements. Genome Biol 2015;16:253. 10.1186/s13059-015-0818-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85. Leenay RT, Maksimchuk KR, Slotkowski RA. et al. Identifying and visualizing functional PAM diversity across CRISPR-Cas systems. Mol Cell 2016;62:137–47. 10.1016/j.molcel.2016.02.031 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86. Mendoza BJ, Trinh CT. In Silico processing of the complete CRISPR-Cas spacer space for identification of PAM sequences. Biotechnol J 2018;13:e1700595. 10.1002/biot.201700595 [DOI] [PubMed] [Google Scholar]
- 87. Rybnicky GA, Fackler NA, Karim AS. et al. Spacer2PAM: A computational framework to guide experimental determination of functional CRISPR-Cas system PAM sequences. Nucleic Acids Res 2022;50:3523–34. 10.1093/nar/gkac142 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88. Ciciani M, Demozzi M, Pedrazzoli E. et al. Automated identification of sequence-tailored Cas9 proteins using massive metagenomic data. Nat Commun 2022;13:6474. 10.1038/s41467-022-34213-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89. Borges AL, Davidson AR, Bondy-Denomy J. The discovery, mechanisms, and evolutionary impact of anti-CRISPRs. Annu Rev Virol 2017;4:37–59. 10.1146/annurev-virology-101416-041616 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90. Bondy-Denomy J, Davidson AR, Doudna JA. et al. A unified resource for tracking anti-CRISPR names. CRISPR J 2018;1:304–5. 10.1089/crispr.2018.0043 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91. Huang L, Yang B, Yi H. et al. AcrDB: a database of anti-CRISPR operons in prokaryotes and viruses. Nucleic Acids Res 2021;49:D622–9. 10.1093/nar/gkaa857 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92. Wang J, Dai W, Li J. et al. AcrHub: an integrative hub for investigating, predicting and mapping anti-CRISPR proteins. Nucleic Acids Res 2021;49:D630–8. 10.1093/nar/gkaa951 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93. Dong C, Hao G-F, Hua H-L. et al. Anti-CRISPRdb: a comprehensive online resource for anti-CRISPR proteins. Nucleic Acids Res 2018;46:D393–8. 10.1093/nar/gkx835 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94. Dong C, Wang X, Ma C. et al. Anti-CRISPRdb v2.2: an online repository of anti-CRISPR proteins including information on inhibitory mechanisms, activities and neighbors of curated anti-CRISPR proteins. Database 2022;2022:1–9. 10.1093/database/baac010 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95. Yi H, Huang L, Yang B. et al. AcrFinder: genome mining anti-CRISPR operons in prokaryotes and their viruses. Nucleic Acids Res 2020;48:W358–65. 10.1093/nar/gkaa351 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96. Wang J, Dai W, Li J. et al. PaCRISPR: a server for predicting and visualizing anti-CRISPR proteins. Nucleic Acids Res 2020;48:W348–57. 10.1093/nar/gkaa432 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97. Yang B, Zheng J, Yin Y. AcaFinder: genome Mining for Anti-CRISPR-associated genes. mSystems 2022;7:7. 10.1128/msystems.00817-22 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98. Wandera KG, Alkhnbashi OS, Bassett HV. et al. Anti-CRISPR prediction using deep learning reveals an inhibitor of Cas13b nucleases. Mol Cell 2022;82:2714–2726.e4. 10.1016/j.molcel.2022.05.003 [DOI] [PubMed] [Google Scholar]
- 99. Zhu L, Wang X, Li F. et al. PreAcrs: a machine learning framework for identifying anti-CRISPR proteins. BMC Bioinformatics 2022;23:444. 10.1186/s12859-022-04986-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100. Li Y, Wei Y, Xu S. et al. AcrNET: predicting anti-CRISPR with deep learning. Bioinformatics 2023;39. 10.1093/bioinformatics/btad259 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101. Gussow AB, Park AE, Borges AL. et al. Machine-learning approach expands the repertoire of anti-CRISPR protein families. Nat Commun 2020;11:3784. 10.1038/s41467-020-17652-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102. Doench JG, Hartenian E, Graham DB. et al. Rational design of highly active sgRNAs for CRISPR-Cas9–mediated gene inactivation. Nat Biotechnol 2014;32:1262–7. 10.1038/nbt.3026 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103. Doench JG, Fusi N, Sullender M. et al. Optimized sgRNA design to maximize activity and minimize off-target effects of CRISPR-Cas9. Nat Biotechnol 2016;34:184–91. 10.1038/nbt.3437 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104. Wong N, Liu W, Wang X. WU-CRISPR: characteristics of functional guide RNAs for the CRISPR/Cas9 system. Genome Biol 2015;16:218. 10.1186/s13059-015-0784-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105. Zhang J-P, Li X-L, Neises A. et al. Different effects of sgRNA length on CRISPR-mediated gene knockout efficiency. Sci Rep 2016;6:28566. 10.1038/srep28566 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106. Chari R, Mali P, Moosburner M. et al. Unraveling CRISPR-Cas9 genome engineering parameters via a library-on-library approach. Nat Methods 2015;12:823–6. 10.1038/nmeth.3473 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107. Wu X, Scott DA, Kriz AJ. et al. Genome-wide binding of the CRISPR endonuclease Cas9 in mammalian cells. Nat Biotechnol 2014;32:670–6. 10.1038/nbt.2889 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108. Liang G, Zhang H, Lou D. et al. Selection of highly efficient sgRNAs for CRISPR/Cas9-based plant genome editing. Sci Rep 2016;6:21451. 10.1038/srep21451 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109. Nishimasu H, Ran FA, Hsu PD. et al. Crystal structure of Cas9 in complex with guide RNA and target DNA. Cell 2014;156:935–49. 10.1016/j.cell.2014.02.001 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110. Liu G, Zhang Y, Zhang T. Computational approaches for effective CRISPR guide RNA design and evaluation. Comput Struct Biotechnol J 2020;18:35–44. 10.1016/j.csbj.2019.11.006 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111. Wang J, Zhang X, Cheng L. et al. An overview and metanalysis of machine and deep learning-based CRISPR gRNA design tools. RNA Biol 2020;17:13–22. 10.1080/15476286.2019.1669406 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112. Montague TG, Cruz JM, Gagnon JA. et al. CHOPCHOP: a CRISPR/Cas9 and TALEN web tool for genome editing. Nucleic Acids Res 2014;42:W401–7. 10.1093/nar/gku410 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113. Naito Y, Hino K, Bono H. et al. CRISPRdirect: software for designing CRISPR/Cas guide RNA with reduced off-target sites. Bioinformatics 2015;31:1120–3. 10.1093/bioinformatics/btu743 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114. Blin K, Pedersen LE, Weber T. et al. CRISPy-web: An online resource to design sgRNAs for CRISPR applications. Synth Syst Biotechnol 2016;1:118–21. 10.1016/j.synbio.2016.01.003 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115. Pliatsika V, Rigoutsos I. “Off-spotter”: very fast and exhaustive enumeration of genomic lookalikes for designing CRISPR/Cas guide RNAs. Biol Direct 2015;10:4. 10.1186/s13062-015-0035-z [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116. Cui Y, Liao X, Peng S. et al. OffScan: a universal and fast CRISPR off-target sites detection tool. BMC Genomics 2020;21:872. 10.1186/s12864-019-6241-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117. Ferragina P, Manzini G. Opportunistic data structures with applications. In: Proceedings 41st Annual Symposium on Foundations of Computer Science, pp. 390–8, 2000. 10.1109/SFCS.2000.892127 [DOI]
- 118. Li B, Chen PB, Diao Y. CRISPR-SE: a brute force search engine for CRISPR design. NAR Genom Bioinform 2021;3:3. 10.1093/nargab/lqab013 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119. Concordet J-P, Haeussler M. CRISPOR: intuitive guide selection for CRISPR/Cas9 genome editing experiments and screens. Nucleic Acids Res 2018;46:W242–5. 10.1093/nar/gky354 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120. DeWeirdt PC, McGee AV, Zheng F. et al. Accounting for small variations in the tracrRNA sequence improves sgRNA activity predictions for CRISPR screening. Nat Commun 2022;13:5255. 10.1038/s41467-022-33024-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121. Riesenberg S, Kanis P, Karlic R. et al. Robust prediction of synthetic gRNA activity and cryptic DNA repair by disentangling cellular CRISPR cleavage outcomes. Nat Commun 2025;16:4717. 10.1038/s41467-025-59947-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122. Chapdelaine-Trépanier V, Shenoy S, Masud W. et al. CRISPR-BEasy: a free web-based service for designing sgRNA tiling libraries for CRISPR-dependent base editing screens. Nucleic Acids Res 2025;53:W193–202. 10.1093/nar/gkaf382 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123. Hsu PD, Scott DA, Weinstein JA. et al. DNA targeting specificity of RNA-guided Cas9 nucleases. Nat Biotechnol 2013;31:827–32. 10.1038/nbt.2647 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124. Labun K, Montague TG, Krause M. et al. CHOPCHOP v3: expanding the CRISPR web toolbox beyond genome editing. Nucleic Acids Res 2019;47:W171–4. 10.1093/nar/gkz365 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125. Labun K, Montague TG, Gagnon JA. et al. CHOPCHOP v2: a web tool for the next generation of CRISPR genome engineering. Nucleic Acids Res 2016;44:W272–6. 10.1093/nar/gkw398 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126. Hough SH, Kancleris K, Brody L. et al. Guide picker is a comprehensive design tool for visualizing and selecting guides for CRISPR experiments. BMC Bioinformatics 2017;18:167. 10.1186/s12859-017-1581-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127. Hwang GH, Kim JS, Bae S. Web-Based CRISPR Toolkits: Cas-OFFinder, Cas-Designer, and Cas-Analyzer. In: Fulga TA, Knapp DJHF, Ferry QRV. (eds) CRISPR Guide RNA Design. Methods in Molecular Biology, vol 2162. Humana, New York, NY. 2021. 10.1007/978-1-0716-0687-2_2 [DOI] [PubMed] [Google Scholar]
- 128. Bae S, Park J, Kim J-S. Cas-OFFinder: a fast and versatile algorithm that searches for potential off-target sites of Cas9 RNA-guided endonucleases. Bioinformatics 2014;30:1473–5. 10.1093/bioinformatics/btu048 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129. Park J, Bae S, Kim J-S. Cas-designer: a web-based tool for choice of CRISPR-Cas9 target sites. Bioinformatics 2015;31:4014–6. 10.1093/bioinformatics/btv537 [DOI] [PubMed] [Google Scholar]
- 130. Zhu H, Misel L, Graham M. et al. CT-finder: a web service for CRISPR optimal target prediction and visualization. Sci Rep 2016;6:25516. 10.1038/srep25516 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131. Rastogi A, Murik O, Bowler C. et al. PhytoCRISP-ex: a web-based and stand-alone application to find specific target sequences for CRISPR/CAS editing. BMC Bioinformatics 2016;17:261. 10.1186/s12859-016-1143-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132. Xie X, Ma X, Zhu Q. et al. CRISPR-GE: a convenient software toolkit for CRISPR-based genome editing. Mol Plant 2017;10:1246–9. 10.1016/j.molp.2017.06.004 [DOI] [PubMed] [Google Scholar]
- 133. Cao Q, Ma J, Chen C-H. et al. CRISPR-FOCUS: a web server for designing focused CRISPR screening experiments. PloS One 2017;12:e0184281. 10.1371/journal.pone.0184281 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134. Poudel R, Rodriguez LT, Reisch CR. et al. GuideMaker: software to design CRISPR-Cas guide RNA pools in non-model genomes. Gigascience 2022;11:11. 10.1093/gigascience/giac007 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135. Perez AR, Pritykin Y, Vidigal JA. et al. GuideScan software for improved single and paired CRISPR guide RNA design. Nat Biotechnol 2017;35:347–9. 10.1038/nbt.3804 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136. Schmidt H, Zhang M, Mourelatos H. et al. Genome-wide CRISPR guide RNA design and specificity analysis with GuideScan2. Genome Biol 2025;26:41. 10.1186/s13059-025-03488-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137. Lin Y, Cradick TJ, Brown MT. et al. CRISPR/Cas9 systems have off-target activity with insertions or deletions between target DNA and guide RNA sequences. Nucleic Acids Res 2014;42:7473–85. 10.1093/nar/gku402 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138. Cancellieri S, Canver MC, Bombieri N. et al. CRISPRitz: rapid, high-throughput and variant-aware in silico off-target site identification for CRISPR genome editing. Bioinformatics 2020;36:2001–8. 10.1093/bioinformatics/btz867 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139. Nakamae K, Bono H. DANGER analysis: risk-averse on/off-target assessment for CRISPR editing without a reference genome. Bioinformatics Advances 2023;3:vbad114. 10.1093/bioadv/vbad114 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140. Hoberecht L, Perampalam P, Lun A. et al. A comprehensive Bioconductor ecosystem for the design of CRISPR guide RNAs across nucleases and technologies. Nat Commun 2022;13:6568. 10.1038/s41467-022-34320-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141. Mendoza BJ, Trinh CT. Enhanced guide-RNA design and targeting analysis for precise CRISPR genome editing of single and consortia of industrially relevant and non-model organisms. Bioinformatics 2018;34:16–23. 10.1093/bioinformatics/btx564 [DOI] [PubMed] [Google Scholar]
- 142. Mendoza B, Fry T, Dooley D. et al. CASPER: An integrated software platform for rapid development of CRISPR tools. CRISPR J 2022;5:609–17. 10.1089/crispr.2022.0025 [DOI] [PubMed] [Google Scholar]
- 143. Prykhozhij SV, Rajan V, Gaston D. et al. CRISPR MultiTargeter: a web tool to find common and unique CRISPR single guide RNA targets in a set of similar sequences. PloS One 2015;10:e0119372. 10.1371/journal.pone.0119372 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144. Veluchamy A, Teles K, Fischle W. CRISPR-broad: combined design of multi-targeting gRNAs and broad, multiplex target finding. Sci Rep 2023;13:19717. 10.1038/s41598-023-46212-x [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145. Zhao G, Li J, Tang Y. AsCRISPR: a web server for allele-specific single guide RNA Design in Precision Medicine. CRISPR J 2020;3:512–22. 10.1089/crispr.2020.0071 [DOI] [PubMed] [Google Scholar]
- 146. Rabinowitz R, Almog S, Darnell R. et al. CrisPam: SNP-derived PAM analysis tool for allele-specific targeting of genetic variants using CRISPR-Cas systems. Front Genet 2020;11:11. 10.3389/fgene.2020.00851 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147. Chen C-L, Rodiger J, Chung V. et al. SNP-CRISPR: a web tool for SNP-specific genome editing. G3 Genes|Genomes|Genetics 2020;10:489–94. 10.1534/g3.119.400904 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148. Keough KC, Lyalina S, Olvera MP. et al. AlleleAnalyzer: a tool for personalized and allele-specific sgRNA design. Genome Biol 2019;20:167. 10.1186/s13059-019-1783-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149. Rosignoli S, Lustrino E, Conci A. et al. AlPaCas: allele-specific CRISPR gene editing through a protospacer-adjacent-motif (PAM) approach. Nucleic Acids Res 2024;52:W29–38. 10.1093/nar/gkae419 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150. O’Brien AR, Burgio G, Bauer DC. Domain-specific introduction to machine learning terminology, pitfalls and opportunities in CRISPR-based gene editing. Brief Bioinform 2021;22:308–14. 10.1093/bib/bbz145 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151. Moreno-Mateos MA, Vejnar CE, Beaudoin J-D. et al. CRISPRscan: designing highly efficient sgRNAs for CRISPR-Cas9 targeting in vivo. Nat Methods 2015;12:982–8. 10.1038/nmeth.3543 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152. Xu H, Xiao T, Chen C-H. et al. Sequence determinants of improved CRISPR sgRNA design. Genome Res 2015;25:1147–57. 10.1101/gr.191452.115 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 153. Wilson LOW, Reti D, O’Brien AR. et al. High activity target-site identification using phenotypic independent CRISPR-Cas9 Core functionality. CRISPR J 2018;1:182–90. 10.1089/crispr.2017.0021 [DOI] [PubMed] [Google Scholar]
- 154. Peng H, Zheng Y, Blumenstein M. et al. CRISPR/Cas9 cleavage efficiency regression through boosting algorithms and Markov sequence profiling. Bioinformatics 2018;34:3069–77. 10.1093/bioinformatics/bty298 [DOI] [PubMed] [Google Scholar]
- 155. Hiranniramol K, Chen Y, Liu W. et al. Generalizable sgRNA design for improved CRISPR/Cas9 editing efficiency. Bioinformatics 2020;36:2684–9. 10.1093/bioinformatics/btaa041 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156. Sahu M, Dash R. A survey on deep learning: convolution neural network (CNN). Intelligent and Cloud Computing: Proceedings of ICICC 2019;2021:317–25. 10.1007/978-981-15-6202-0_32 [DOI] [Google Scholar]
- 157. Webb S. Deep learning for biology. Nature 2018;554:555–7. 10.1038/d41586-018-02174-z [DOI] [PubMed] [Google Scholar]
- 158. Wu Y, Feng J. Development and application of artificial neural network. Wirel Pers Commun 2018;102:1645–56. 10.1007/s11277-017-5224-x [DOI] [Google Scholar]
- 159. Zhong Z, Li Z, Yang J. et al. Unified model to predict gRNA efficiency across diverse cell lines and CRISPR-Cas9 systems. J Chem Inf Model 2023;63:7320–9. 10.1021/acs.jcim.3c01339 [DOI] [PubMed] [Google Scholar]
- 160. Yu Y, Gawlitt S, LB A e S. et al. Improved prediction of bacterial CRISPRi guide efficiency from depletion screens through mixed-effect machine learning and data integration. Genome Biol 2024;25:25. 10.1186/s13059-023-03153-y [DOI] [PMC free article] [PubMed] [Google Scholar]
- 161. Li J, Wu P, Cao Z. et al. Machine learning-based prediction models to guide the selection of Cas9 variants for efficient gene editing. Cell Rep 2024;43:113765. 10.1016/j.celrep.2024.113765 [DOI] [PubMed] [Google Scholar]
- 162. Zhang H, Yan J, Lu Z. et al. Deep sampling of gRNA in the human genome and deep-learning-informed prediction of gRNA activities. Cell Discov 2023;9:48. 10.1038/s41421-023-00549-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 163. Wessels H-H, Stirn A, Méndez-Mancilla A. et al. Prediction of on-target and off-target activity of CRISPR–Cas13d guide RNAs using deep learning. Nat Biotechnol 2024;42:628–37. 10.1038/s41587-023-01830-8 [DOI] [PubMed] [Google Scholar]
- 164. Chen Q, Chuai G, Zhang H. et al. Genome-wide CRISPR off-target prediction and optimization using RNA-DNA interaction fingerprints. Nat Commun 2023;14:7521. 10.1038/s41467-023-42695-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 165. Yang Y, Li J, Zou Q. et al. Prediction of CRISPR-Cas9 off-target activities with mismatches and indels based on hybrid neural network. Comput Struct Biotechnol J 2023;21:5039–48. 10.1016/j.csbj.2023.10.018 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 166. Störtz F, Mak JK, Minary P. piCRISPR: physically informed deep learning models for CRISPR/Cas9 off-target cleavage prediction. Artificial Intelligence in the Life Sciences 2023;3:100075. 10.1016/j.ailsci.2023.100075 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 167. Sun J, Guo J, Liu J. CRISPR-M: predicting sgRNA off-target effect using a multi-view deep learning network. PLoS Comput Biol 2024;20:20. 10.1371/journal.pcbi.1011972 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 168. Toufikuzzaman M, Hassan Samee MA, Sohel RM. CRISPR-DIPOFF: an interpretable deep learning approach for CRISPR Cas-9 off-target prediction. Brief Bioinform 2024;25:bbad530. 10.1093/bib/bbad530 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 169. Listgarten J, Weinstein M, Kleinstiver BP. et al. Prediction of off-target activities for the end-to-end design of CRISPR guide RNAs. Nat Biomed Eng 2018;2:38–47. 10.1038/s41551-017-0178-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 170. Chuai G, Ma H, Yan J. et al. DeepCRISPR: optimized CRISPR guide RNA design by deep learning. Genome Biol 2018;19:80. 10.1186/s13059-018-1459-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 171. Luo J, Chen W, Xue L. et al. Prediction of activity and specificity of CRISPR-Cpf1 using convolutional deep learning neural networks. BMC Bioinformatics 2019;20:332. 10.1186/s12859-019-2939-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 172. O’Brien A, Bauer DC, Burgio G. Predicting CRISPR-Cas12a guide efficiency for targeting using machine learning. PloS One 2023;18:e0292924. 10.1371/journal.pone.0292924 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 173. Liu Y, Fan R, Yi J. et al. A fusion framework of deep learning and machine learning for predicting sgRNA cleavage efficiency. Comput Biol Med 2023;165:107476. 10.1016/j.compbiomed.2023.107476 [DOI] [PubMed] [Google Scholar]
- 174. Cheng X, Li Z, Shan R. et al. Modeling CRISPR-Cas13d on-target and off-target effects using machine learning approaches. Nat Commun 2023;14:752. 10.1038/s41467-023-36316-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 175. Labuhn M, Adams FF, Ng M. et al. Refined sgRNA efficacy prediction improves large- and small-scale CRISPR–Cas9 applications. Nucleic Acids Res 2018;46:1375–85. 10.1093/nar/gkx1268 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 176. Chari R, Yeo NC, Chavez A. et al. sgRNA scorer 2.0: a species-independent model to predict CRISPR/Cas9 activity. ACS Synth Biol 2017;6:902–4. 10.1021/acssynbio.6b00343 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 177. Kim HK, Min S, Song M. et al. Deep learning improves prediction of CRISPR–Cpf1 guide RNA activity. Nat Biotechnol 2018;36:239–41. 10.1038/nbt.4061 [DOI] [PubMed] [Google Scholar]
- 178. Liu Q, He D, Xie L. Prediction of off-target specificity and cell-specific fitness of CRISPR-Cas system using attention boosted deep learning and network-based gene feature. PLoS Comput Biol 2019;15:e1007480. 10.1371/journal.pcbi.1007480 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 179. Baisya D, Ramesh A, Schwartz C. et al. Genome-wide functional screens enable the prediction of high activity CRISPR-Cas9 and -Cas12a guides in Yarrowia lipolytica. Nat Commun 2022;13:922. 10.1038/s41467-022-28540-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 180. Kirillov B, Savitskaya E, Panov M. et al. Uncertainty-aware and interpretable evaluation of Cas9–gRNA and Cas12a–gRNA specificity for fully matched and partially mismatched targets with deep kernel learning. Nucleic Acids Res 2022;50:e11–1. 10.1093/nar/gkab1065 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 181. Zhang G, Zeng T, Dai Z. et al. Prediction of CRISPR/Cas9 single guide RNA cleavage efficiency and specificity by attention-based convolutional neural networks. Comput Struct Biotechnol J 2021;19:1445–57. 10.1016/j.csbj.2021.03.001 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 182. Xiao L-M, Wan Y-Q, Jiang Z-R. AttCRISPR: a spacetime interpretable model for prediction of sgRNA on-target activity. BMC Bioinformatics 2021;22:589. 10.1186/s12859-021-04509-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 183. Zhang Z-R, Jiang Z-R. Effective use of sequence information to predict CRISPR-Cas9 off-target. Comput Struct Biotechnol J 2022;20:650–61. 10.1016/j.csbj.2022.01.006 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 184. Elkayam S, Orenstein Y. DeepCRISTL: deep transfer learning to predict CRISPR/Cas9 functional and endogenous on-target editing efficiency. Bioinformatics 2022;38:i161–8. 10.1093/bioinformatics/btac218 [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- 185. Ham DT, Browne TS, Banglorewala PN. et al. A generalizable Cas9/sgRNA prediction model using machine transfer learning with small high-quality datasets. Nat Commun 2023;14:5514. 10.1038/s41467-023-41143-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 186. Guan Z, Jiang Z. Transformer-based anti-noise models for CRISPR-Cas9 off-target activities prediction. Brief Bioinform 2023;24:bbad127. 10.1093/bib/bbad127 [DOI] [PubMed] [Google Scholar]
- 187. Rahman MK, Rahman MS. CRISPRpred: a flexible and efficient tool for sgRNAs on-target activity prediction in CRISPR/Cas9 systems. PloS One 2017;12:e0181943. 10.1371/journal.pone.0181943 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 188. Wang D, Zhang C, Wang B. et al. Optimized CRISPR guide RNA design for two high-fidelity Cas9 variants by deep learning. Nat Commun 2019;10:4284. 10.1038/s41467-019-12281-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 189. Wang J, Xiang X, Bolund L. et al. GNL-scorer: a generalized model for predicting CRISPR on-target activity by machine learning and featurization. J Mol Cell Biol 2021;12:909–11. 10.1093/jmcb/mjz116 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 190. Xiang X, Corsi GI, Anthon C. et al. Enhancing CRISPR-Cas9 gRNA efficiency prediction by data integration and deep learning. Nat Commun 2021;12:3238. 10.1038/s41467-021-23576-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 191. Zarate OA, Yang Y, Wang X. et al. BoostMEC: predicting CRISPR-Cas9 cleavage efficiency through boosting models. BMC Bioinformatics 2022;23:446. 10.1186/s12859-022-04998-z [DOI] [PMC free article] [PubMed] [Google Scholar]
- 192. Abadi S, Yan WX, Amar D. et al. A machine learning approach for predicting CRISPR-Cas9 cleavage efficiencies and patterns underlying its mechanism of action. PLoS Comput Biol 2017;13:e1005807. 10.1371/journal.pcbi.1005807 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 193. Dimauro G, Colagrande P, Carlucci R. et al. CRISPRLearner: a deep learning-based system to predict CRISPR/Cas9 sgRNA on-target cleavage efficiency. Electronics (Basel) 2019;8:1478. 10.3390/electronics8121478 [DOI] [Google Scholar]
- 194. Zhang Y, Long Y, Yin R. et al. DL-CRISPR: a deep learning method for off-target activity prediction in CRISPR/Cas9 with data augmentation. IEEE Access 2020;8:76610–7. 10.1109/ACCESS.2020.2989454 [DOI] [Google Scholar]
- 195. Liu Q, Cheng X, Liu G. et al. Deep learning improves the ability of sgRNA off-target propensity prediction. BMC Bioinformatics 2020;21:51. 10.1186/s12859-020-3395-z [DOI] [PMC free article] [PubMed] [Google Scholar]
- 196. Pennington J, Socher R, Manning C. Glove: Global vectors for word representation. In: Proceedings of the 2014 Conference on Empirical Methods in Natural Language Processing (EMNLP), pp. 1532–43, Doha, Qatar. Association for Computational Linguistics. 2014. 10.3115/v1/D14-1162 [DOI]
- 197. Lin J, Zhang Z, Zhang S. et al. CRISPR-net: a recurrent convolutional network quantifies CRISPR off-target activities with mismatches and Indels. Advanced Science 2020;7:1903562. 10.1002/advs.201903562 [DOI] [Google Scholar]
- 198. Zhang G, Dai Z, Dai X. A novel hybrid CNN-SVR for CRISPR/Cas9 guide RNA activity prediction. Front Genet 2020;10:1303. 10.3389/fgene.2019.01303 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 199. Zhang G, Dai Z, Dai X. C-RNNCrispr: prediction of CRISPR/Cas9 sgRNA activity using convolutional and recurrent neural networks. Comput Struct Biotechnol J 2020;18:344–54. 10.1016/j.csbj.2020.01.013 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 200. Konstantakos V, Nentidis A, Krithara A. et al. CRISPRedict: a CRISPR-Cas9 web tool for interpretable efficiency predictions. Nucleic Acids Res 2022;50:W191–8. 10.1093/nar/gkac466 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 201. Das J, Kumar S, Mishra DC. et al. Machine learning in the estimation of CRISPR-Cas9 cleavage sites for plant system. Front Genet 2023;13:1085332. 10.3389/fgene.2022.1085332 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 202. Karmakar S, Panda D, Panda S. et al. A miniature alternative to Cas9 and Cas12: transposon-associated TnpB mediates targeted genome editing in plants. Plant Biotechnol J 2024;22:2950–3. 10.1111/pbi.14416 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 203. Karvelis T, Druteika G, Bigelyte G. et al. Transposon-associated TnpB is a programmable RNA-guided DNA endonuclease. Nature 2021;599:692–6. 10.1038/s41586-021-04058-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 204. Marquart KF, Mathis N, Mollaysa A. et al. Effective genome editing with an enhanced ISDra2 TnpB system and deep learning-predicted ωRNAs. Nat Methods 2024;21:2084–93. 10.1038/s41592-024-02418-z [DOI] [PMC free article] [PubMed] [Google Scholar]
- 205. Chakrabarti AM, Henser-Brownhill T, Monserrat J. et al. Target-specific precision of CRISPR-mediated genome editing. Mol Cell 2019;73:699–713.e6. 10.1016/j.molcel.2018.11.031 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 206. Molla KA, Yang Y. CRISPR/Cas-Mediated Base editing: technical considerations and practical applications. Trends Biotechnol 2019;37:1121–42. 10.1016/j.tibtech.2019.03.008 [DOI] [PubMed] [Google Scholar]
- 207. Biswas S, Zhang D, Shi J. CRISPR/Cas systems: opportunities and challenges for crop breeding. Plant Cell Rep 2021;40:979–98. 10.1007/s00299-021-02708-2 [DOI] [PubMed] [Google Scholar]
- 208. Liu X, Wang S, Ai D. Predicting CRISPR/Cas9 repair outcomes by attention-based deep learning framework. Cells 2022;11:1847. 10.3390/cells11111847 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 209. Pallaseni A, Peets EM, Girling G. et al. The interplay of DNA repair context with target sequence predictably biases Cas9-generated mutations. Nat Commun 2024;15:10271. 10.1038/s41467-024-54566-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 210. Molla KA, Shih J, Yang Y. Single-nucleotide editing for zebra3 and wsl5 phenotypes in rice using CRISPR/Cas9-mediated adenine base editors. aBIOTECH 2020;1:106–18. 10.1007/s42994-020-00018-x [DOI] [PMC free article] [PubMed] [Google Scholar]
- 211. Molla KA, Sretenovic S, Bansal KC. et al. Precise plant genome editing using base editors and prime editors. Nat Plants 2021;7:1166–87. 10.1038/s41477-021-00991-1 [DOI] [PubMed] [Google Scholar]
- 212. Tong Y, Whitford CM, Robertsen HL. et al. Highly efficient DSB-free base editing for streptomycetes with CRISPR-BEST. Proc Natl Acad Sci 2019;116:20366–75. 10.1073/pnas.1913493116 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 213. Blin K, Shaw S, Tong Y. et al. Designing sgRNAs for CRISPR-BEST base editing applications with CRISPy-web 2.0. Synth Syst. Biotechnol 2020;5:99–102. 10.1016/j.synbio.2020.05.005 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 214. Wang Y, Gao R, Wu J. et al. Comparison of cytosine base editors and development of the BEable-GPS database for targeting pathogenic SNVs. Genome Biol 2019;20:218. 10.1186/s13059-019-1839-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 215. Rabinowitz R, Abadi S, Almog S. et al. Prediction of synonymous corrections by the BE-FF computational tool expands the targeting scope of base editing. Nucleic Acids Res 2020;48:W340–7. 10.1093/nar/gkaa215 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 216. Billon P, Bryant EE, Joseph SA. et al. CRISPR-Mediated Base editing enables efficient disruption of eukaryotic genes through induction of STOP codons. Mol Cell 2017;67:1068–1079.e4. 10.1016/j.molcel.2017.08.008 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 217. Siegner SM, Karasu ME, Schröder MS. et al. PnB designer: a web application to design prime and base editor guide RNAs for animals and plants. BMC Bioinformatics 2021;22:101. 10.1186/s12859-021-04034-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 218. Xie X, Li F, Tan X. et al. BEtarget: a versatile web-based tool to design guide RNAs for base editing in plants. Comput Struct Biotechnol J 2022;20:4009–14. 10.1016/j.csbj.2022.07.046 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 219. Wu Y, He Y, Sretenovic S. et al. CRISPR-BETS: a base-editing design tool for generating stop codons. Plant Biotechnol J 2022;20:499–510. 10.1111/pbi.13732 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 220. Fan J, Shi L, Liu Q. et al. Annotation and evaluation of base editing outcomes in multiple cell types using CRISPRbase. Nucleic Acids Res 2023;51:D1249–56. 10.1093/nar/gkac967 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 221. Wang Y, Liu Y, Li J. et al. Expanding targeting scope, editing window, and base transition capability of base editing in Corynebacterium glutamicum. Biotechnol Bioeng 2019;116:3016–29. 10.1002/bit.27121 [DOI] [PubMed] [Google Scholar]
- 222. Yu H, Wu Z, Chen X. et al. CRISPR-CBEI: a designing and Analyzing tool kit for Cytosine Base editor-mediated gene inactivation. mSystems 2020;5:5. 10.1128/mSystems.00350-20 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 223. Maxim DS, Wu DW, Johnson NS. et al. EditABLE: a simple web application for designing genome editing experiments. Res Sq 2024;rs-3. 10.21203/rs.3.rs-4775705/v1 [DOI] [Google Scholar]
- 224. Schneider PG, Liu S, Bullinger L. et al. BEscreen: a versatile toolkit to design base editing libraries. Nucleic Acids Res 2025;53:W68–72. 10.1093/nar/gkaf406 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 225. Song M, Kim HK, Lee S. et al. Sequence-specific prediction of the efficiencies of adenine and cytosine base editors. Nat Biotechnol 2020;38:1037–43. 10.1038/s41587-020-0573-5 [DOI] [PubMed] [Google Scholar]
- 226. Arbab M, Shen MW, Mok B. et al. Determinants of base editing outcomes from target library analysis and machine learning. Cell 2020;182:463–480.e30. 10.1016/j.cell.2020.05.037 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 227. Koblan LW, Arbab M, Shen MW. et al. Efficient C•G-to-G•C base editors developed using CRISPRi screens, target-library analysis, and machine learning. Nat Biotechnol 2021;39:1414–25. 10.1038/s41587-021-00938-z [DOI] [PMC free article] [PubMed] [Google Scholar]
- 228. Yuan T, Yan N, Fei T. et al. Optimization of C-to-G base editors with sequence context preference predictable by machine learning methods. Nat Commun 2021;12:4902. 10.1038/s41467-021-25217-y [DOI] [PMC free article] [PubMed] [Google Scholar]
- 229. Marquart KF, Allam A, Janjuha S. et al. Predicting base editing outcomes with an attention-based deep learning algorithm trained on high-throughput target library screens. Nat Commun 2021;12:5114. 10.1038/s41467-021-25375-z [DOI] [PMC free article] [PubMed] [Google Scholar]
- 230. Zhang C, Yu Z, Wang D. et al. BEdeepon: an in silico tool for prediction of base editor efficiencies and outcomes. bioRxiv 2021. 10.1101/2021.03.14.435303 [DOI] [Google Scholar]
- 231. Pallaseni A, Peets EM, Koeppel J. et al. Predicting base editing outcomes using position-specific sequence determinants. Nucleic Acids Res 2022;50:3551–64. 10.1093/nar/gkac161 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 232. Zhang C, Yang Y, Qi T. et al. Prediction of base editor off-targets by deep learning. Nat Commun 2023;14:5358. 10.1038/s41467-023-41004-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 233. Kim N, Choi S, Kim S. et al. Deep learning models to predict the editing efficiencies and outcomes of diverse base editors. Nat Biotechnol 2024;42:484–97. 10.1038/s41587-023-01792-x [DOI] [PubMed] [Google Scholar]
- 234. Molla KA, Qi Y, Karmakar S. et al. Base editing landscape extends to perform Transversion mutation. Trends Genet 2020;36:899–901. 10.1016/j.tig.2020.09.001 [DOI] [PubMed] [Google Scholar]
- 235. Kim DY, Bin MS, Ko J-H. et al. Unbiased investigation of specificities of prime editing systems in human cells. Nucleic Acids Res 2020;48:10576–89. 10.1093/nar/gkaa764 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 236. Li H, Li J, Chen J. et al. Precise modifications of both exogenous and endogenous genes in Rice by prime editing. Mol Plant 2020;13:671–4. 10.1016/j.molp.2020.03.011 [DOI] [PubMed] [Google Scholar]
- 237. Sürün D, Schneider A, Mircetic J. et al. Efficient generation and correction of mutations in human iPS cells utilizing mRNAs of CRISPR Base editors and prime editors. Genes (Basel) 2020;11:511. 10.3390/genes11050511 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 238. Xu W, Zhang C, Yang Y. et al. Versatile nucleotides substitution in plant using an improved prime editing system. Mol Plant 2020;13:675–8. 10.1016/j.molp.2020.03.012 [DOI] [PubMed] [Google Scholar]
- 239. Nelson JW, Randolph PB, Shen SP. et al. Engineered pegRNAs improve prime editing efficiency. Nat Biotechnol 2022;40:402–10. 10.1038/s41587-021-01039-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 240. Liu Y, Li X, He S. et al. Efficient generation of mouse models with the prime editing system. Cell Discov 2020;6:27. 10.1038/s41421-020-0165-z [DOI] [PMC free article] [PubMed] [Google Scholar]
- 241. Lin Q, Jin S, Zong Y. et al. High-efficiency prime editing with optimized, paired pegRNAs in plants. Nat Biotechnol 2021;39:923–7. 10.1038/s41587-021-00868-w [DOI] [PubMed] [Google Scholar]
- 242. Chow RD, Chen JS, Shen J. et al. A web tool for the design of prime-editing guide RNAs. Nat Biomed Eng 2020;5:190–4. 10.1038/s41551-020-00622-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 243. Hsu JY, Grünewald J, Szalay R. et al. PrimeDesign software for rapid and simplified design of prime editing guide RNAs. Nat Commun 2021;12:1034. 10.1038/s41467-021-21337-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 244. Morris JA, Rahman JA, Guo X. et al. Automated design of CRISPR prime editors for 56,000 human pathogenic variants. iScience 2021;24:103380. 10.1016/j.isci.2021.103380 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 245. Hwang G-H, Jeong YK, Habib O. et al. PE-designer and PE-Analyzer: web-based design and analysis tools for CRISPR prime editing. Nucleic Acids Res 2021;49:W499–504. 10.1093/nar/gkab319 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 246. Li Y, Chen J, Tsai SQ. et al. Easy-prime: a machine learning–based prime editor design tool. Genome Biol 2021;22:235. 10.1186/s13059-021-02458-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 247. Bhagwat AM, Graumann J, Wiegandt R. et al. multicrispr: gRNA design for prime editing and parallel targeting of thousands of targets. Life Sci Alliance 2020;3:e202000757. 10.26508/lsa.202000757 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 248. Standage-Beier K, Tekel SJ, Brafman DA. et al. Prime editing guide RNA design automation using PINE-CONE. ACS Synth Biol 2021;10:422–7. 10.1021/acssynbio.0c00445 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 249. Adikusuma F, Lushington C, Arudkumar J. et al. Optimized nickase- and nuclease-based prime editing in human and mouse cells. Nucleic Acids Res 2021;49:10785–95. 10.1093/nar/gkab792 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 250. Choi J, Chen W, Suiter CC. et al. Precise genomic deletions using paired prime editing. Nat Biotechnol 2022;40:218–26. 10.1038/s41587-021-01025-z [DOI] [PMC free article] [PubMed] [Google Scholar]
- 251. Gould SI, Wuest AN, Dong K. et al. High-throughput evaluation of genetic variants with prime editing sensor libraries. Nat Biotechnol 2024;1–15. 10.1038/s41587-024-02172-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 252. Kim HK, Yu G, Park J. et al. Predicting the efficiency of prime editing guide RNAs in human cells. Nat Biotechnol 2021;39:198–206. 10.1038/s41587-020-0677-y [DOI] [PubMed] [Google Scholar]
- 253. Yu G, Kim HK, Park J. et al. Prediction of efficiencies for diverse prime editing systems in multiple cell types. Cell 2023;186:2256–2272.e23. 10.1016/j.cell.2023.03.034 [DOI] [PubMed] [Google Scholar]
- 254. Koeppel J, Weller J, Peets EM. et al. Prediction of prime editing insertion efficiencies using sequence features and DNA repair determinants. Nat Biotechnol 2023;41:1446–56. 10.1038/s41587-023-01678-y [DOI] [PMC free article] [PubMed] [Google Scholar]
- 255. Mathis N, Allam A, Kissling L. et al. Predicting prime editing efficiency and product purity by deep learning. Nat Biotechnol 2023;41:1151–9. 10.1038/s41587-022-01613-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 256. Mathis N, Allam A, Tálas A. et al. Machine learning prediction of prime editing efficiency across diverse chromatin contexts. Nat Biotechnol 2025;43:712–9. 10.1038/s41587-024-02268-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 257. Jiang W, Bikard D, Cox D. et al. RNA-guided editing of bacterial genomes using CRISPR-Cas systems. Nat Biotechnol 2013;31:233–9. 10.1038/nbt.2508 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 258. Mali P, Yang L, Esvelt KM. et al. RNA-guided human genome engineering via Cas9. Science 1979;339:823–6. 10.1126/science.1232033 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 259. Xie K, Yang Y. RNA-guided genome editing in plants using a CRISPR–Cas system. Mol Plant 2013;6:1975–83. 10.1093/mp/sst119 [DOI] [PubMed] [Google Scholar]
- 260. Lei Y, Lu L, Liu H-Y. et al. CRISPR-P: a web tool for synthetic single-guide RNA design of CRISPR-system in plants. Mol Plant 2014;7:1494–6. 10.1093/mp/ssu044 [DOI] [PubMed] [Google Scholar]
- 261. Xie K, Zhang J, Yang Y. Genome-wide prediction of highly specific guide RNA spacers for CRISPR–Cas9-mediated genome editing in model plants and major crops. Mol Plant 2014;7:923–6. 10.1093/mp/ssu009 [DOI] [PubMed] [Google Scholar]
- 262. Brazelton VA, Zarecor S, Wright DA. et al. A quick guide to CRISPR sgRNA design tools. GM Crops Food 2015;6:266–76. 10.1080/21645698.2015.1137690 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 263. Liu H, Ding Y, Zhou Y. et al. CRISPR-P 2.0: An improved CRISPR-Cas9 tool for genome editing in plants. Mol Plant 2017;10:530–2. 10.1016/j.molp.2017.01.003 [DOI] [PubMed] [Google Scholar]
- 264. Minkenberg B, Zhang J, Xie K. et al. CRISPR-PLANT v2: an online resource for highly specific guide RNA spacers based on improved off-target analysis. Plant Biotechnol J 2019;17:5–8. 10.1111/pbi.13025 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 265. Cram D, Kulkarni M, Buchwaldt M. et al. WheatCRISPR: a web-based guide RNA design tool for CRISPR/Cas9-mediated genome editing in wheat. BMC Plant Biol 2019;19:474. 10.1186/s12870-019-2097-z [DOI] [PMC free article] [PubMed] [Google Scholar]
- 266. Sun J, Liu H, Liu J. et al. CRISPR-local: a local single-guide RNA (sgRNA) design tool for non-reference plant genomes. Bioinformatics 2019;35:2501–3. 10.1093/bioinformatics/bty970 [DOI] [PubMed] [Google Scholar]
- 267. Naim F, Shand K, Hayashi S. et al. Are the current gRNA ranking prediction algorithms useful for genome editing in plants? PloS One 2020;15:e0227994. 10.1371/journal.pone.0227994 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 268. He C, Liu H, Chen D. et al. CRISPR-cereal: a guide RNA design tool integrating regulome and genomic variation for wheat, maize and rice. Plant Biotechnol J 2021;19:2141–3. 10.1111/pbi.13675 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 269. Vazquez-Vilar M, Bernabé-Orts JM, Fernandez-del-Carmen A. et al. A modular toolbox for gRNA–Cas9 genome engineering in plants based on the GoldenBraid standard. Plant Methods 2016;12:10. 10.1186/s13007-016-0101-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 270. Vazquez-Vilar M, Garcia-Carpintero V, Selma S. et al. The GB4.0 platform, an all-In-one tool for CRISPR/Cas-based multiplex genome engineering in plants. Front Plant Sci 2021;12:12. 10.3389/fpls.2021.689937 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 271. Sarrion-Perdigones A, Falconi EE, Zandalinas SI. et al. GoldenBraid: An iterative cloning system for standardized assembly of reusable genetic modules. PloS One 2011;6:e21622. 10.1371/journal.pone.0021622 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 272. Feng Z, Mao Y, Xu N. et al. Multigeneration analysis reveals the inheritance, specificity, and patterns of CRISPR/Cas-induced gene modifications in Arabidopsis. Proc Natl Acad Sci 2014;111:4632–7. 10.1073/pnas.1400822111 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 273. Li J-F, Norville JE, Aach J. et al. Multiplex and homologous recombination–mediated genome editing in Arabidopsis and Nicotiana benthamiana using guide RNA and Cas9. Nat Biotechnol 2013;31:688–91. 10.1038/nbt.2654 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 274. Pinello L, Canver MC, Hoban MD. et al. Analyzing CRISPR genome-editing experiments with CRISPResso. Nat Biotechnol 2016;34:695–7. 10.1038/nbt.3583 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 275. Brinkman EK, Kousholt AN, Harmsen T. et al. Easy quantification of template-directed CRISPR/Cas9 editing. Nucleic Acids Res 2018;46:e58–8. 10.1093/nar/gky164 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 276. Lindsay H, Burger A, Biyong B. et al. CrispRVariants charts the mutation spectrum of genome engineering experiments. Nat Biotechnol 2016;34:701–2. 10.1038/nbt.3628 [DOI] [PubMed] [Google Scholar]
- 277. Boel A, Steyaert W, De Rocker N. et al. BATCH-GE: batch analysis of next-generation sequencing data for genome editing assessment. Sci Rep 2016;6:30330. 10.1038/srep30330 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 278. Wang X, Tilford C, Neuhaus I. et al. CRISPR-DAV: CRISPR NGS data analysis and visualization pipeline. Bioinformatics 2017;33:3811–2. 10.1093/bioinformatics/btx518 [DOI] [PubMed] [Google Scholar]
- 279. Liu W, Xie X, Ma X. et al. DSDecode: a web-based tool for decoding of sequencing chromatograms for genotyping of targeted mutations. Mol Plant 2015;8:1431–3. 10.1016/j.molp.2015.05.009 [DOI] [PubMed] [Google Scholar]
- 280. Park J, Lim K, Kim J-S. et al. Cas-analyzer: an online tool for assessing genome editing results using NGS data. Bioinformatics 2017;33:286–8. 10.1093/bioinformatics/btw561 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 281. Liu Q, Wang C, Jiao X. et al. Hi-TOM: a platform for high-throughput tracking of mutations induced by CRISPR/Cas systems. Sci China Life Sci 2019;62:1–7. 10.1007/s11427-018-9402-9 [DOI] [PubMed] [Google Scholar]
- 282. Connelly JP, Pruett-Miller SM. CRIS.py: a versatile and high-throughput analysis program for CRISPR-based genome editing. Sci Rep 2019;9:4194. 10.1038/s41598-019-40896-w [DOI] [PMC free article] [PubMed] [Google Scholar]
- 283. Lee H, Chang HY, Cho SW. et al. CRISPRpic: fast and precise analysis for CRISPR-induced mutations via prefixed index counting. NAR Genom Bioinform 2020;2:2. 10.1093/nargab/lqaa012 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 284. Bloh K, Kanchana R, Bialk P. et al. Deconvolution of complex DNA repair (DECODR): establishing a novel deconvolution algorithm for comprehensive analysis of CRISPR-edited sanger sequencing data. CRISPR J 2021;4:120–31. 10.1089/crispr.2020.0022 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 285. Kurgan G, Turk R, Li H. et al. CRISPAltRations: a validated cloud-based approach for interrogation of double-strand break repair mediated by CRISPR genome editing. Mol Ther Methods Clin Dev 2021;21:478–91. 10.1016/j.omtm.2021.03.024 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 286. Conant D, Hsiau T, Rossi N. et al. Inference of CRISPR edits from sanger trace data. CRISPR J 2022;5:123–30. 10.1089/crispr.2021.0113 [DOI] [PubMed] [Google Scholar]
- 287. Corsi GI, Gadekar VP, Gorodkin J. et al. CRISPRroots: on-and off-target assessment of RNA-seq data in CRISPR-Cas9 edited cells. Nucleic Acids Res 2022;50:e20. 10.1093/nar/gkab1131 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 288. Fu H, Shan C, Kang F. et al. CRISPR-GRANT: a cross-platform graphical analysis tool for high-throughput CRISPR-based genome editing evaluation. BMC Bioinformatics 2023;24:219. 10.1186/s12859-023-05333-w [DOI] [PMC free article] [PubMed] [Google Scholar]
- 289. Han Y, Liu G, Wu Y. et al. CrisprStitch: fast evaluation of the efficiency of CRISPR editing systems. Plant Commun 2024;5:100783. 10.1016/j.xplc.2023.100783 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 290. Xue L-J, Tsai C-J. AGEseq: analysis of genome editing by sequencing. Mol Plant 2015;8:1428–30. 10.1016/j.molp.2015.06.001 [DOI] [PubMed] [Google Scholar]
- 291. Varshney GK, Pei W, LaFave MC. et al. High-throughput gene targeting and phenotyping in zebrafish using CRISPR/Cas9. Genome Res 2015;25:1030–42. 10.1101/gr.186379.114 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 292. You Q, Zhong Z, Ren Q. et al. CRISPRMatch: An automatic calculation and visualization tool for high-throughput CRISPR genome-editing data analysis. Int J Biol Sci 2018;14:858–62. 10.7150/ijbs.24581 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 293. Labun K, Guo X, Chavez A. et al. Accurate analysis of genuine CRISPR editing events with ampliCan. Genome Res 2019;29:843–7. 10.1101/gr.244293.118 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 294. Bruyneel AAN, Colas AR, Karakikes I. et al. AlleleProfileR: a versatile tool to identify and profile sequence variants in edited genomes. PloS One 2019;14:e0226694. 10.1371/journal.pone.0226694 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 295. Hwang G-H, Yu J, Yang S. et al. CRISPR-sub: analysis of DNA substitution mutations caused by CRISPR-Cas9 in human cells. Comput Struct Biotechnol J 2020;18:1686–94. 10.1016/j.csbj.2020.06.026 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 296. Chatterjee P, Jakimo N, Jacobson JM. Minimal PAM specificity of a highly similar SpCas9 ortholog. Sci Adv 2018;4:4. 10.1126/sciadv.aau0766 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 297. Kluesner MG, Nedveck DA, Lahr WS. et al. EditR: a method to Quantify Base editing from sanger sequencing. CRISPR J 2018;1:239–50. 10.1089/crispr.2018.0014 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 298. Xu L, Liu Y, Han R. BEAT: a python program to Quantify Base editing from sanger sequencing. CRISPR J 2019;2:223–9. 10.1089/crispr.2019.0017 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 299. Spahn PN, Bath T, Weiss RJ. et al. PinAPL-Py: a comprehensive web-application for the analysis of CRISPR/Cas9 screens. Sci Rep 2017;7:15854. 10.1038/s41598-017-16193-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 300. Hsu JY, Fulco CP, Cole MA. et al. CRISPR-SURF: discovering regulatory elements by deconvolution of CRISPR tiling screen data. Nat Methods 2018;15:992–3. 10.1038/s41592-018-0225-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 301. Jeong H-H, Kim SY, Rousseaux MWC. et al. CRISPRcloud: a secure cloud-based pipeline for CRISPR pooled screen deconvolution. Bioinformatics 2017;33:2963–5. 10.1093/bioinformatics/btx335 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 302. Jeong H-H, Kim SY, Rousseaux MWC. et al. Beta-binomial modeling of CRISPR pooled screen data identifies target genes with greater sensitivity and fewer false negatives. Genome Res 2019;29:999–1008. 10.1101/gr.245571.118 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 303. Li W, Xu H, Xiao T. et al. MAGeCK enables robust identification of essential genes from genome-scale CRISPR/Cas9 knockout screens. Genome Biol 2014;15:554. 10.1186/s13059-014-0554-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 304. Hart T, Moffat J. BAGEL: a computational framework for identifying essential genes from pooled library screens. BMC Bioinformatics 2016;17:164. 10.1186/s12859-016-1015-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 305. Allen F, Behan F, Khodak A. et al. JACKS: joint analysis of CRISPR/Cas9 knockout screens. Genome Res 2019;29:464–71. 10.1101/gr.238923.118 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 306. Schoonenberg VAC, Cole MA, Yao Q. et al. CRISPRO: identification of functional protein coding sequences based on genome editing dense mutagenesis. Genome Biol 2018;19:169. 10.1186/s13059-018-1563-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 307. Kim E, Hart T. Improved analysis of CRISPR fitness screens and reduced off-target effects with the BAGEL2 gene essentiality classifier. Genome Med 2021;13:2. 10.1186/s13073-020-00809-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 308. Li W, Köster J, Xu H. et al. Quality control, modeling, and visualization of CRISPR screens with MAGeCK-VISPR. Genome Biol 2015;16:281. 10.1186/s13059-015-0843-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 309. Wang B, Wang M, Zhang W. et al. Integrative analysis of pooled CRISPR genetic screens using MAGeCKFlute. Nat Protoc 2019;14:756–80. 10.1038/s41596-018-0113-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 310. Fiaux PC, Chen HV, Chen PB. et al. Discovering functional sequences with RELICS, an analysis method for CRISPR screens. PLoS Comput Biol 2020;16:e1008194. 10.1371/journal.pcbi.1008194 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 311. de Boer CG, Ray JP, Hacohen N. et al. MAUDE: inferring expression changes in sorting-based CRISPR screens. Genome Biol 2020;21:134. 10.1186/s13059-020-02046-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Supplementary Materials
Data Availability Statement
Data presented in the study are included as tables in the Supplementary material. All supplementary materials are available via Zenodo at https://doi.org/10.5281/zenodo.15254840.










