Next Article in Journal
The Role of TuACO Gene Family in Response to Biotic and Abiotic Stresses in Triticum urartu
Previous Article in Journal
Genome-Wide Identification and Tissue-Specific Expression Profiling of Goji CER Gene Family
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Artificial Intelligence-Assisted CRISPR/Cas Systems for Targeting Plant Viruses

Rustem Omarov Plant Biotechnology Laboratory, Department of Biotechnology and Microbiology, L.N. Gumilyov Eurasian National University, Astana 010008, Kazakhstan
*
Author to whom correspondence should be addressed.
Genes 2025, 16(11), 1258; https://doi.org/10.3390/genes16111258
Submission received: 29 September 2025 / Revised: 17 October 2025 / Accepted: 23 October 2025 / Published: 24 October 2025
(This article belongs to the Section Plant Genetics and Genomics)

Abstract

Plant viral infections continue to pose a significant and ongoing threat to global food security, especially in the context of climatic instability and intensive agricultural practices. The CRISPR/Cas system has emerged as a powerful tool for developing virus-resistant crops by enabling precise modifications to viral genomes or plant susceptibility factors. Nonetheless, the efficacy and dependability of CRISPR-based antiviral approaches are limited by challenges in guide RNA design, off-target effects, insufficiently annotated datasets, and the intricate biological dynamics of plant–virus interactions. This paper summarizes the latest advancements in the incorporation of artificial intelligence (AI) methodologies, including machine learning and deep learning algorithms, into the CRISPR design and optimization framework. It examines how convolutional and recurrent neural networks, transformer architectures, and generative models like AlphaFold2, RoseTTAFold, and ESMFold can be used to predict protein structures, score sgRNAs, and model host–virus interactions. AI-enhanced methods have been proven to improve target specificity, Cas protein performance, and in silico validation. This paper aims to establish a foundation for next-generation genome editing strategies against plant viruses and promote the adoption of AI-powered CRISPR technologies in sustainable agriculture.

1. Introduction

Despite significant improvements in breeding and agricultural technologies, viral plant diseases remain one of the most devastating biotic factors, severely constraining the productivity and quality of agricultural crops worldwide. The Food and Agriculture Organization of the United Nations estimates that yearly output losses in tropical and subtropical regions from viral infections can exceed 30%, particularly in crops lacking inherent resistance [1]. Viruses such as tomato yellow leaf curl virus (TYLCV), rice tungro bacilliform virus (RTBV), cotton leaf curl virus (CLCuV), and maize streak virus (MSV) pose major threats to important food crops. Host species such as Solanum lycopersicum, Oryza sativa, Gossypium hirsutum and Zea mays are particularly vulnerable due to the high virulence and rapid spread of viral pathogens via vectors, seeds, or mechanical transmission [2,3,4,5,6]. The intricacy and adaptability of viral infections, limited protection strategies, and the deficiency of resistant variations necessitate the advancement of novel, high-precision biotechnological solutions. The CRISPR/Cas system, derived from the adaptive immune response of bacteria, is one of the most promising genome editing platforms, reconfigured as a universal tool for precise manipulation of DNA and RNA [7,8,9,10,11]. In plant science, CRISPR/Cas is employed for the direct annihilation of viral genomes and for the alteration of host plant genes associated with the detection and dissemination of infection [12]. Over the past decade, nucleases like Cas9, Cas12a, and Cas13a, which are efficient against many kinds of viruses, encompassing both DNA and RNA viruses, have been successfully modified and utilized [13,14,15,16]. The efficacy of CRISPR systems in plants is largely contingent upon the appropriate selection of essential components: guide RNA (gRNA) [17,18], a variant of the Cas protein with the requisite biochemical properties [19,20], and delivery mechanisms [21,22]. However, precise targeting is challenged by the rapid mutation rates of plant viruses and limited functional annotation of susceptibility genes, especially in under-studied crop species. These constraints hinder the broad applicability of conventional genome editing strategies.
Given these challenges, the application of artificial intelligence (AI) techniques, encompassing machine learning (ML) [23,24,25] and deep learning (DL) [26,27,28], for the analysis, modeling, and optimization of CRISPR editing is especially significant. Contemporary AI models utilizing convolutional neural networks (CNNs) [29,30,31], recurrent neural networks (RNNs) [31,32,33], and transformer [34,35] architectures exhibit significant precision in forecasting gRNA efficiency and specificity, detecting off-target effects, and examining interactions between viral and plant proteins [36,37,38]. Initial research on the in silico design of Cas proteins exhibiting certain attributes, including enhanced PAM (Protospacer Adjacent Motif) specificity, stability inside plant cells, and modularity of functional domains, is emerging. Nonetheless, the majority of these models exhibit restricted transferability among plant species, particularly between model organisms and agricultural crops characterized by extensive and intricate genomes.
The necessity to analyze and integrate multi-omics data from genomes, transcriptomics, proteomics, and metabolomics adds an extra layer of complexity. This data serves as a crucial resource for discovering resistance variables, modifying targets, and forecasting intervention efficacy. An instance is the transcriptome analysis of A. thaliana infected with tobacco etch virus (TEV), which discovered differentially expressed immune response genes, such as PR1 and EDS1, linked to the salicylate-dependent defense system [39]. These data facilitated high-accuracy classification of resistant and susceptible genotypes only after their incorporation into a machine learning model. An analogous methodology was employed in the examination of Solanum lycopersicum’s reaction to tomato spotted wilt virus (TSWV), wherein gradient boosting discerned pivotal genes (WRKY, MYB, EDS1) with a classification accuracy of 89% [40]. It is important to acknowledge that many models are trained on limited datasets and often fail to include cross-validation or a distinct division between training and testing sets, resulting in overfitting and inflated accuracy estimates. The use of computer vision techniques in combination with deep learning architectures such as convolutional neural networks (CNNs) has enabled automated phenotypic analysis of viral symptoms, thereby advancing disease diagnostics. A hybrid CNN and Random Forest model attained an accuracy of up to 95% in assessing the extent of damage to Nicotiana tabacum leaves infected with tobacco mosaic virus (TMV) in photos subjected to varying lighting conditions and morphologies [41,42]. Moreover, GWAS and pan-genome analysis techniques provide avenues for the precise identification of resistance alleles. A pan-genome investigation of 3000 Oryza sativa genomes revealed polymorphisms in eIF(iso)4G and RYMV1 linked to resistance against RTBV and rice yellow mottle virus (RYMV), which were functionally validated via CRISPR interference [43,44]. Despite these advances, several challenges persist, including the absence of cohesive annotated datasets, the inadequate portability of AI models developed on human-derived data, and the limited interpretability of numerous algorithms. Moreover, regulatory concerns and bioethical evaluations of genome-edited plants, especially those employing AI, necessitate thorough analysis and discourse.
This review paper aims to examine the potential and existing applications of artificial intelligence technologies in CRISPR/Cas systems for the protection of plants against viral infections. It investigates the role of AI in the design of guide RNAs, the engineering of Cas proteins, and the modeling of virus–plant interactions. In addition, the review outlines emerging research directions and discusses regulatory considerations relevant to AI-assisted genetic engineering (Figure 1).

2. AI-Enhanced CRISPR Strategies Against Plant Viruses

2.1. AI/ML Tools and Algorithms for sgRNA Design

CRISPR/Cas technologies have become essential to plant biotechnology, serving as a pivotal instrument for precise genome editing. Nonetheless, a considerable disparity persists between effective experimental applications and the advancement of computational techniques for the systematic design of guide RNAs (sgRNAs) [45,46]. The principal constraint continues to be the exceedingly scarce quantity of specialist models tailored for plant systems. Efforts to use algorithms derived from mammalian or prokaryotic data frequently result in a significant decline in predictive accuracy, underscoring the distinctiveness of plant genomes and the necessity for the creation of indigenous solutions [47,48,49]. This issue is especially evident in crops with extensive and intricate genomes, such as wheat and maize, where repetitive elements and atypical PAM motifs are prevalent [50,51]. The disparity is more obvious when juxtaposed with medical genomic engineering, where numerous precise and advanced machine learning (ML) methods, founded on vast in vivo and in vitro datasets, have been established in recent years [52,53].
In plant biology, a notable effective solution is sgRNACNN [54,55,56], an ensemble of convolutional neural networks trained on in planta data for four crops: A. thaliana, Oryza sativa, Zea mays, and Solanum lycopersicum. The model exhibits a 15–30% enhancement in accuracy for forecasting guideline performance vs. current universal methods, but solely inside the initial training area [57,58]. This illustrates the overarching issue of inadequate transferability of AI models among crops, particularly in the absence of transfer learning, fine-tuning, or few-shot learning. Less complex yet interpretable positional classifiers, exemplified by the model of Das et al. (2023) [59], were utilized on Capsicum annuum and N. benthamiana, yielding moderate accuracy (correlation coefficients of 0.45–0.55) while demanding minimal computational resources, thus appealing to laboratories with constrained infrastructure. The benefit of these models lies in their predictive explainability, enabling the identification of nucleotide locations and factors that affect sgRNA efficiency. Their primary limitation is their failure to consider the spatial (3D) architecture of the genome and epigenetic changes, which significantly influence gene expression in plants.
Commonly utilized general-purpose tools, including DeepCpf1, CRISPR-HNN, sgRNA Scorer 2.0, TIGER, CHOPCHOP, E-CRISP, CRISPR Genome-wide Analysis, and Cas-OFFinder, were predominantly designed for humans and bacteria [60]. For instance, DeepCpf1, a model based on deep neural networks for predicting Cas12a (Cpf1) activity, demonstrates elevated ROC-AUC (>0.8) and R2 (>0.6) in human systems [61]. Nonetheless, when implemented using in planta data in rice, the accuracy diminishes by 25–30%, attributable to both biological discrepancies and diversity in PAM preferences and Cas12a sensitivity to sequence context in plants [30,62]. The CRISPR-HNN model, a hybrid design integrating convolutional and recurrent neural networks, demonstrates exceptional efficacy in extracting both local and global features [63,64]. The absence of plant data during training results in prediction instability when analyzing extremely complex genomes, such as those of wheat and maize, characterized by prevalent repetitions and intricate spatial structures [65]. Nevertheless, the majority of research fails to reveal validation criteria, like the type of cross-validation employed, the proportion of the test sample utilized, or the incorporation of independent datasets. This diminishes the likelihood of reproducibility and objective evaluation of prediction quality.
Among traditional but common approaches, sgRNA Scorer 2.0 is prominent. Originally designed as a general-purpose instrument, it has been predominantly evaluated using human and murine data; however, it has also been effectively employed in gene editing endeavors involving rice and tobacco, notably in the development of resistance to tobacco mosaic virus (TMV) [66,67]. TIGER, CHOPCHOP, and E-CRISP are extensively utilized. These tools offer user-friendly web interfaces and facilitate the swift creation of lists of prospective sgRNAs, including assessments of off-target effects. Nonetheless, the majority of these are founded on scientific principles and statistical models obtained from animal or prokaryotic data. Their prediction capability in plant genomes is constrained. For instance, correlation coefficients with trial outcomes never surpass 0.3–0.4, and the compilation of suggested guidelines frequently encompasses locations that are ineffectual in practice. Approaches to sgRNA design guided by AI must move beyond sequence-level parameters and incorporate a deeper understanding of viral evolutionary constraints. In the context of geminiviruses, direct CRISPR/Cas-mediated interference is most successful when targeting highly conserved genomic regions, such as the intergenic region (IR) and the replication-associated protein gene (Rep). By contrast, targeting more variable open reading frames, particularly those encoding capsid proteins (CPs), frequently results in reduced editing efficiency due to the rapid emergence of escape variants. This phenomenon has been experimentally confirmed. Ali et al. (2021) [68] demonstrated that CRISPR/Cas9 constructs directed at the IR of several geminiviruses resulted in robust and stable viral interference in Nicotiana benthamiana. In the same study, constructs targeting the CP gene permitted partial viral replication and gave rise to escape mutants. Similarly, Tripathi et al. (2021) [69] reported that targeting the Rep gene of African cassava mosaic virus (ACMV) conferred stable resistance in transgenic cassava lines, whereas CP-targeting sgRNAs showed lower efficacy and editing persistence.
Beyond geminiviruses, targeted CRISPR/Cas interventions have also been explored in other plant-infecting DNA and RNA viruses. In tomato, Ali et al. (2015) [70] also successfully reduced TYLCV accumulation by targeting both the IR and ORF C1. These findings reinforce the strategic advantage of multiplex targeting to mitigate viral evasion. Maree et al. (2010) [71] characterized a cluster of subgenomic RNAs encoding ORFs 3–12 in grapevine leafroll-associated virus 3 (GLRaV-3), highlighting the need for precision in selecting target sites in large 3′-terminal ORF arrays. Efforts to develop broadly effective sgRNAs have included the use of AI-informed models trained on viral diversity. Liu et al. (2021) [72] reported the design of low-strain-specificity sgRNAs targeting conserved regions of the ORF1 (RdRP) across multiple Potyvirus species. Similarly, computational strategies have been applied to design sgRNAs targeting ORF6 and ORF9 of beet yellows virus (BYV) [73], providing proof-of-concept for CRISPR-based disruption of functional viral modules. Importantly, these challenges are not limited to DNA viruses. RNA viruses such as tobacco mosaic virus (TMV) and cucumber mosaic virus (CMV), characterized by high mutation rates, pose a unique difficulty for stable CRISPR targeting. Here, ML models offer a valuable solution by identifying evolutionarily constrained and functionally essential regions across viral genomes. As emphasized by Zaidi et al. (2020) [74], conservation-guided sgRNA design through AI can reduce the likelihood of viral escape, particularly when combined with multiplexed editing strategies. Collectively, these findings underscore that effective CRISPR-mediated resistance requires thoughtful selection of target loci prioritizing conserved, replication-related ORFs such as Rep and RdRP over structurally plastic genes like CP. The integration of AI tools into this process enables a more nuanced and predictive approach to sgRNA design, which is essential for achieving durable viral resistance in crops and for advancing our mechanistic understanding of plant–virus interactions.
TIGER provides efficient batch processing of targets; however, its heuristics neglect the distribution of PAM motifs and the prevalence of pseudogenes in plant genomes, resulting in a significant proportion of false positives [75]. CHOPCHOP and E-CRISP are favored for their user-friendliness and compatibility with several Cas systems in rice, tomato, and A. thaliana. Specifically, CHOPCHOP was utilized with tomato leaf curl virus (ToLCV), illustrating the tool’s adaptability [76]. Nonetheless, their algorithms for evaluating on-target activity rely on rudimentary measures such as GC composition and the existence of certain motifs, which are not tailored to plant data. While they may provide a foundation for researchers engaged in preliminary initiatives, their predictions necessitate compulsory experimental validation for essential applications. The Cas-OFFinder tool has been utilized to evaluate off-target effects in genome editing initiatives for soybeans, corn, and tobacco, in addition to the creation of CRISPR-based diagnostic systems targeting viral RNAs [77,78]. This tool remains widely used due to its speed and compatibility with various Cas nucleases; however, it performs only mechanical sequence enumeration and fails to consider the biological context. This may result in an exaggerated assessment of risks and the inundation of experiments with superfluous candidates. Conversely, CRISPR Genome-wide Analysis facilitates extensive screening over the entire genome; nevertheless, it is hindered by an absence of plant-specific filters and training data [79,80,81].
A comparative review of current methods reveals that the primary restriction resides not in the computational architecture, but in the insufficient systemic adaptation to plant data. Even very proficient models like DeepCpf1 or CRISPR-HNN, when provided with high-quality in-plant datasets, might attain correlation values of 0.7 or above. Nevertheless, due to the current scarcity of such data, their predictive accuracy is comparable to random selection of candidates based on marginal or non-informative features.
The existing ecosystem of prediction tools for CRISPR/Cas in plants comprises a limited selection of specialized models and numerous general-purpose solutions with reduced transferability [82] (Table 1). To surmount existing constraints, the following are requisite: Systematic aggregation of extensive in-plant datasets; formulation of adaptive machine learning models derived from this data; and establishment of hybrid solutions that integrate the precision of deep learning with the comprehensibility of conventional methodologies.

2.2. Modeling and Optimization of Cas Proteins Using AI

The engineering of Cas proteins for the suppression of plant viruses is a contemporary focus in plant biotechnology, wherein artificial intelligence techniques, especially deep learning, exhibit significant promise for the rational design of proteins with enhanced specificity, efficacy, and controllability. In contrast to the extensively examined selection of guide RNAs, the creation of Cas proteins necessitates comprehension of their molecular characteristics, the architecture of their interactions with nucleic acids, and their functionality under particular plant cell circumstances [24,83]. Deep learning facilitates the modeling of these factors utilizing extensive biological data, encompassing amino acid sequences, three-dimensional structures, and interaction information with viral genomes [84,85].
The initial systematic application of AI-assisted CRISPR technology to combat plant viral infections was reported by Zhang et al. (2018) [86], who employed a modified FnCas9 endonuclease from Francisella novicida to inhibit cucumber mosaic virus (CMV) and tobacco mosaic virus (TMV) in Nicotiana benthamiana and Arabidopsis thaliana. In this study, the CRISPR/Cas components were delivered using a tobacco rattle virus (TRV)-based vector system, enabling systemic expression in planta. The sgRNAs were designed to specifically target the coat protein (CP) genes of CMV and TMV, which are essential for virion assembly and systemic movement. The selection of effective FnCas9 variants was guided by convolutional neural network models trained to predict RNA-protein interaction interfaces, enhancing binding affinity and cleavage efficiency. This AI-guided optimization contributed to a marked reduction in viral load, and importantly, the antiviral effect was shown to be heritable in subsequent plant generations. Another instance was the SpCas9 initiative targeting the cotton leaf curl Multan virus (CLCuMuV) conducted by Yin et al. (2019) [87]. Algorithms utilizing ensemble decision trees and a neural network classifier were employed to identify two extremely effective Cas9-targeted locations in viral DNA. While a traditional delivery strategy was employed throughout the in-plant phase, the protein selection stage and activity prediction relied on AI analysis of nuclease activity inside the framework of the viral genome. This technique guaranteed total viral resistance in N. benthamiana specimens.
In the case of RNA viruses like TMV, Cas13 family proteins have demonstrated significant utility. Cao et al. (2021) [88] employed the CasRx enzyme (RfxCas13d), characterized by its single-stranded RNA cleavage capability, to target foreign RNA viruses. Attention-based deep learning algorithms were employed to examine the secondary structure of viral RNA and pinpoint areas of greatest accessibility. The application of CasRx to N. benthamiana plants resulted in decreased viral expression and observable symptoms of infection. One significant advantage of CasRx is its diminutive size and autonomy from PAM or PFS, which facilitates engineering. Nonetheless, CasRx activity is contingent upon the cellular environment, and in certain instances, the HEPN domain may induce non-specific RNA cleavage, particularly when expression is aberrant.
Prior research by Ji et al. (2015) [89] employing SpCas9 to target beet severe curly top virus (BSCTV) exemplifies one of the initial instances of genetically engineering viral resistance by direct modification of the viral genome. Artificial intelligence was employed in the selection of the most conserved locations in viral DNA, utilizing techniques such as the positional weight model and logistic regression analysis. The study was conducted in N. benthamiana and A. thaliana, achieving a significant decrease in viral replication. Ghorbani Faal et al. (2020) [90] utilized inducible Cas9 expression to inhibit tomato yellow leaf curl virus (TYLCV) in Solanum lycopersicum. AI was employed to model promoter designs and forecast the appropriate temporal progression of Cas9 activation upon infection. This method facilitated virus-specific activation of editing with few side effects. Nonetheless, the majority of models employed for Cas protein design do not incorporate in-plant data and are restricted to in vitro or in silico simulations. This establishes a substantial disparity between computational forecasts and actual biological efficacy under host plant conditions.
Consequently, deep learning is emerging as a crucial instrument in the design of Cas proteins with enhanced characteristics aimed at specific plant viruses (Table 2). This encompasses activity forecasting, optimization of target interactions, creation of novel protein variations, and adaptation to plant physiology. Principal advantages encompass elevated accuracy, capacity to study hitherto unexamined proteins, and integration with in situ testing. Constraints encompass data prerequisites and model interpretability; however, the swift advancement of structural frameworks and explainable AI algorithms is facilitating the mitigation of these obstacles.
In addition to direct antiviral strategies, AI has shown growing potential in identifying and modifying plant host susceptibility (S) genes and immune regulators to enhance resistance against viral pathogens. One of the most studied targets is the eIF(iso)4G gene, a translation initiation factor exploited by viruses such as rice tungro bacilliform virus (RTBV) and rice yellow mottle virus (RYMV). Using a combination of pan-genome association studies, SNP-effect prediction, and structural modeling via AlphaFold2, researchers successfully identified resistance-associated polymorphisms in eIF(iso)4G, which were functionally validated using CRISPR interference in Oryza sativa [91]. Similarly, the receptor-like kinase NIK1, known to inhibit viral replication through translational shutdown, was targeted by CRISPR/Cas systems guided by AI-driven co-expression network analysis (WGCNA) and motif discovery tools. These approaches enabled the precise editing of NIK1 regulatory regions to enhance antiviral immunity without compromising plant development [87,92]. Another promising target is BAK1 (BRI1-ASSOCIATED KINASE 1), a key component of pattern recognition receptor complexes. AI-based protein–protein interaction predictors, such as DeepInteract and AlphaFold-Multimer, have been employed to map interactions between BAK1 and viral effectors, thereby informing CRISPR-based strategies to alter viral binding sites while preserving core immune function [93]. In the context of RNA metabolism, DEAD-box helicases such as RH20 have been identified as crucial host factors supporting the replication of RNA viruses. Through transcriptomic analysis, Random Forest classifiers, and gene regulatory network modeling (GENIE3), RH20 homologs were recognized as central hubs and subsequently edited using CRISPR/Cas12a to suppress viral proliferation in Nicotiana benthamiana [94,95]. These findings collectively demonstrate how AI-integrated methodologies, including structural prediction, network inference, and machine learning classifiers are becoming instrumental in guiding CRISPR-based interventions aimed at host determinants of viral susceptibility. The strategic targeting of plant factors such as eIF(iso)4G, NIK1, BAK1, and DEAD-box helicases opens up new avenues for durable and broad-spectrum resistance engineering in crops.

2.3. Machine Learning in the Analysis of Virus–Host Interactions

The application of machine learning (ML) techniques in examining molecular interactions between plants and viruses is a very promising and swiftly advancing domain within molecular phytopathology [96,97,98]. These interactions are fundamental to viral pathogenesis and predominantly entail protein–protein interactions between viral effector proteins and host cellular elements that regulate replication, intracellular transport, and immunological suppression [99,100]. Due to the intricacy of these interactions and the scarcity of experimental data, machine learning algorithms serve as a valuable instrument for their systematic study, enhancing the ability to forecast plant resistance and pinpoint targets for precise genome editing.
An illustrative instance of a particular interaction between viral and plant proteins is the binding of the βC1 protein, encoded by the satellite RNA of the cotton leaf curl virus Multan betasatellite (CLCuMB), to the plant E2 conjugase of the UBC3 ubiquitin system in Gossypium hirsutum [101]. This interaction interferes with proteasome-mediated protein degradation and undermines the plant’s antiviral defenses. Kamal et al. (2019) [102] conducted energy modeling of this complex utilizing molecular dynamics and machine learning techniques, enabling the identification of putative inhibitory peptides that can obstruct the interaction.
Contemporary models for predicting protein–protein interactions (PPI) between viruses and plants generally employ vectorization of amino acid sequences (such as AAC, DPC, PAAC descriptors), with structural and physicochemical attributes [103,104,105]. The data serve as input characteristics in supervised models that are trained on restricted interaction sets and subsequently calibrated on new data. Zhang et al. (2024) [106] introduced the CBIL-VHPLI model, which integrates a hybrid convolutional neural network (CNN) with a bidirectional long short-term memory (BiLSTM) architecture, employing transfer learning techniques. The model exhibited excellent accuracy of 91.6% and a precision of around 93% when evaluating the interactions between the βC1 protein and UBC3 in Gossypium hirsutum. Nonetheless, taking sgRNA models, the majority of PPI models are not tailored for plant proteins; they are frequently trained on generalized datasets, which diminishes the transferability and biological validity of predictions when applied to plants. Besides PPI, machine learning is extensively employed to examine transcriptome alterations in response to viral infection. Gradient boosting and support vector machine (SVM) techniques facilitated the classification of expression profiles in Solanum lycopersicum infected with tomato spotted wilt virus (TSWV) and the identification of resistance-associated genes [107,108]. Genes associated with salicylic acid pathways and phytoalexin production, such as PR1 and EDS1, were recognized as crucial indicators. The incorporation of meteorological and physiological characteristics, including humidity and temperature, into the training set enhanced the model’s predictive accuracy to 89% [109,110]. Nevertheless, the validation of such models’ conclusions in planta is infrequent, and the anticipated markers do not consistently demonstrate functional activity in natural settings, particularly in field varieties and under complex stressors.
Convolutional neural networks are extensively employed for the visual diagnosis of viral infections, including CMV, TMV, and TYLCV [111,112]. Ashtagi (2024) [113] employed a hybrid CNN and Random Forest model to assess pictures of N. tabacum leaves infected with TMV. The model exhibited an accuracy of 95% and an F1-measure of 0.94 during testing, indicating resilience to fluctuations in external factors, such as lighting and leaf morphology. The focus is on predicting plant vulnerability to viruses using whole-genome data. In a model applied to the Oryza sativa system involving rice tungro bacilliform virus (RTBV), the Decision Tree and SVM algorithms facilitated the identification of allelic changes in the eIF(iso)4G gene, which were associated with susceptibility to infection [96,114]. This was further validated using CRISPR interference, facilitating the practical application of genetic resistance by altering this region.
Consequently, the utilization of machine learning in the examination of plant–virus systems has novel prospects for the precise discovery of essential molecular connections, including protein complexes and gene expression regulation networks. Nonetheless, the overarching difficulty persists: elevated efficiency in silico is not invariably validated in planta, particularly under circumstances where gene expression and activity are contingent upon environmental factors, soil composition, microbiome interactions, and the developmental stage of the plant. This necessitates the integration of forecasts with multivariate field experiments. As of yet, ML methods have demonstrated their utility in generating pathogenic models, evaluating breeding, and developing resistant types (Table 3). Nonetheless, extensive deployment of these methodologies necessitates additional growth and standardization of validation data, along with a robust combination of bioinformatic predictions and experimental validation.

2.4. Using Generative AI Models in CRISPR Design

The formulation of efficient antiviral methods for agricultural crops is gaining significance due to global climate change and rising biotic stressors [114]. Viral infections such as TYLCV, RTBV, CMV, and CLCuV result in substantial yearly crop losses [115,116,117]. The most effective strategy for addressing these infections is the application of CRISPR/Cas technology. In this respect, generative AI models like AlphaFold2, RoseTTAFold, and ESMFold, which facilitate the prediction of three-dimensional structures of proteins and protein–protein complexes with atomic precision, are notably noteworthy. These technologies are revolutionizing the methodology for creating CRISPR systems, especially for modifying plant genomes to confer resistance to viral infections.
As a result, AlphaFold2 was employed to predict the structure of the TYLCV Rep capsid protein and to pinpoint essential areas of its interaction with N. benthamiana host proteins. This facilitated the creation of efficient target sites for Cas12a, leading to a 70–80% decrease in viral replication in in planta tests [118,119]. These approaches support the rational identification of target regions within both viral genomes and plant susceptibility genes.
Another instance is employing RoseTTAFold and ProteinMPNN to engineer stable and highly specific Cas protein variants with enhanced PAM recognition, tailored for functionality in plant cells [120,121]. Researchers utilized ESMFold to create models of mutant variants of Cas13a that are effective against RNA viruses, including potato virus Y (PVY) and CMV, and tailored to the cellular environment of Solanum tuberosum. The anticipated structures were employed for in vitro assessments of binding and catalytic efficacy, facilitating the identification of variations with improved cytoplasmic stability and nuclear localization. Generative models are employed to examine interactions between viral and plant proteins. AlphaFold2 has been applied to predict the interaction between the VPg protein of rice yellow mottle virus (RYMV) and the eukaryotic translation initiation factor eIF4E in Oryza sativa, as demonstrated in previous studies [122,123]. Based on these structural predictions, targeted gene modifications were introduced via CRISPR/Cas9 to disrupt the interaction interface, resulting in enhanced viral resistance without compromising plant viability [124]. However, structure prediction tools such as AlphaFold2 and related models exhibit inherent limitations, as they do not account for the cellular environment, dynamic expression patterns, post-translational modifications, or the temporal behavior of protein interactions [68].
Notwithstanding these remarkable accomplishments, considerable limits persist. The majority of training data for AlphaFold2 and analogous models is sourced from animal and bacterial research, potentially constraining the precision of predictions for particular plant proteins and viruses. Secondly, the existing structural data on plant viruses remains insufficient, hindering model validation. In addition, the integration of generative models into applied biotechnology necessitates defined protocols, in-plant validation, and an assessment of plant physiological characteristics. The emergence of artificially designed proteins generated entirely de novo by AI-based platforms such as ProGen2 [125,126] raises biosafety concerns, as these molecules lack natural equivalents and may exhibit unpredictable behavior in biological contexts.
The implementation of generative AI models such as AlphaFold2, RoseTTAFold, and ESMFold demonstrates considerable promise for designing CRISPR system components targeting plant viruses (Table 4). These methods facilitate the synthesis of high-fidelity proteins and gRNAs customized for plant cell environments, hence advancing more predictable, efficient, and secure genome editing in future agriculture.

3. Conclusions

The integration of artificial intelligence into CRISPR-based technologies represents a transformative advancement in the management of plant viral infections. Advancements in deep learning, transformers, and generative models like AlphaFold2, RoseTTAFold, ESMFold, and ProGen2 have markedly enhanced the precision of designing guide RNAs, Cas proteins, and delivery vectors customized for specific plant systems and viral pathogens. These interventions have exhibited compelling efficacy in mitigating significant plant diseases, including TYLCV, RTBV, CMV, TMV, and CLCuV, in crops such as Solanum lycopersicum, Oryza sativa, Gossypium hirsutum, and N. benthamiana. AI-enabled approaches now enable the resolution of previously intractable challenges, such as molecular characterization of virus–host interactions, predictive modeling of CRISPR targeting specificity, prediction of off-target effects, and structural optimization of genome editing enzymes. The predictive accuracy of AI models often declines when transitioning from in vitro models to actual plants. Many algorithms fail to consider epigenetic changes, chromatin accessibility, and intertissue variations in gene expression. The application of generative AI models for creating Cas proteins with altered PAM recognition, increased stability in plant environments, and reduced off-target risks is a noteworthy area of scientific exploration. The application of these technologies enables the shift from universal to precise editing, which is essential given the fast evolution of viral infections.
Nonetheless, these potent instruments are linked to several biological and ethical dilemmas. At the biological level, there exists a risk of inadvertent disruption of plant genomes, encompassing off-target consequences, horizontal transmission of changed components, and unanticipated interactions with the microbiome and ecosystem. This is especially crucial for the utilization of generative models, which may generate unverified protein sequences necessitating obligatory multi-tier validation.
Ethical considerations pertain to the transparency of AI model applications, the interpretability of their predictions, and the ownership of algorithms, data, and resultant genetic structures. The development of proprietary AI systems driven by commercial interests may hinder equitable access to innovation for resource-constrained nations, particularly those vulnerable to widespread viral plant outbreaks. Moreover, there is a necessity for global standardization of legislation pertaining to gene-edited species, particularly with the application of AI models, which add a new layer of complexity in assessing the degree of genomic alteration. An interdisciplinary approach, integrating the expertise of professionals in molecular biology, bioinformatics, agronomy, ethics, and legislation, is especially crucial in this setting. Such collaboration is essential for the advancement of sustainable, scientifically robust, and socially acceptable plant editing methodologies utilizing AI-enhanced CRISPR systems. The creation of open and accessible tools is equally crucial to guarantee global equity and participation in scientific and technological advancement.
AI significantly improves CRISPR capabilities and fundamentally alters its promise in plant science, facilitating safer, more efficient, and more adaptable management of viral risks. Harnessing this potential requires not just technological advancements but also a carefully designed scientific, regulatory, and ethical framework that guarantees sustainability and fosters public trust in emerging biotechnologies.

Author Contributions

Conceptualization, N.I.; writing—original draft preparation, N.I.; writing—review and editing, N.I., A.M., K.Z. and Z.M.; supervision, Z.M. All authors have read and agreed to the published version of the manuscript.

Funding

This work was funded by the Science Committee of the Ministry of Science and Higher Education of the Republic of Kazakhstan grant No. BR21882269.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were created or analyzed in this study.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Sharma, S.K.; Gupta, O.P.; Pathaw, N.; Sharma, D.; Maibam, A.; Sharma, P.; Sanasam, J.; Karkute, S.G.; Kumar, S.; Bhattacharjee, B. CRISPR-Cas-Led revolution in diagnosis and management of emerging plant viruses: New avenues toward food and nutritional security. Front. Nutr. 2021, 8, 751512. [Google Scholar] [CrossRef]
  2. Jones, R.A. Global plant virus disease pandemics and epidemics. Plants 2021, 10, 233. [Google Scholar] [CrossRef]
  3. Ong, S.N.; Taheri, S.; Othman, R.Y.; Teo, C.H. Viral disease of tomato crops (Solanum lycopesicum L.): An overview. J. Plant Dis. Prot. 2020, 127, 725–739. [Google Scholar] [CrossRef]
  4. Prasad, A.; Sharma, N.; Hari-Gowthem, G.; Muthamilarasan, M.; Prasad, M. Tomato yellow leaf curl virus: Impact, challenges, and management. Trends Plant Sci. 2020, 25, 897–911. [Google Scholar] [CrossRef]
  5. Brown, J.K.; Khan, Z. Breeding cotton for cotton leaf curl disease resistance. In Cotton Breeding and Biotechnology; CRC Press: Boca Raton, FL, USA, 2020; pp. 171–197. [Google Scholar]
  6. Shepherd, D.N.; Martin, D.P.; Van der Walt, E.; Dent, K.; Varsani, A.; Rybicki, E.P. Maize streak virus: An old and complex ‘emerging’pathogen. Mol. Plant Pathol. 2010, 11, 1–12. [Google Scholar] [CrossRef] [PubMed]
  7. Iksat, N.; Masalimov, Z.; Omarov, R. Plant virus resistance biotechnological approaches: From genes to the CRISPR/Cas gene editing system. J. Water Land Dev. 2023, 57, 147–158. [Google Scholar] [CrossRef]
  8. Marqués, M.-C.; Sánchez-Vicente, J.; Ruiz, R.; Montagud-Martínez, R.; Márquez-Costa, R.; Gómez, G.; Carbonell, A.; Daròs, J.-A.; Rodrigo, G. Diagnostics of infections produced by the plant viruses TMV, TEV, and PVX with CRISPR-Cas12 and CRISPR-Cas13. ACS Synth. Biol. 2022, 11, 2384–2393. [Google Scholar] [CrossRef] [PubMed]
  9. Zaidi, S.S.E.A.; Tashkandi, M.; Mansoor, S.; Mahfouz, M.M. Engineering plant immunity: Using CRISPR/Cas9 to generate virus resistance. Front. Plant Sci. 2016, 7, 1673. [Google Scholar] [CrossRef]
  10. Madirov, A.; Iksat, N.; Masalimov, Z. Tomato Bushy Stunt Virus (TBSV): From a Plant Pathogen to a Multifunctional Biotechnology Platform. Viruses 2025, 17, 1268. [Google Scholar] [CrossRef]
  11. Jinek, M.; Chylinski, K.; Fonfara, I.; Hauer, M.; Doudna, J.; Charpentier, E. A Programmable Dual-RNA–Guided DNA Endonuclease in Adaptive Bacterial Immunity. Science 2012, 337, 816–821. [Google Scholar] [CrossRef]
  12. Tatineni, S.; Hein, G.L. Plant viruses of agricultural importance: Current and future perspectives of virus disease management strategies. Phytopathology 2023, 113, 117–141. [Google Scholar] [CrossRef]
  13. Robertson, G.; Burger, J.; Campa, M. CRISPR/Cas--based tools for the targeted control of plant viruses. Mol. Plant Pathol. 2022, 23, 1701–1718. [Google Scholar] [CrossRef]
  14. Shahid, M.S.; Sattar, M.N.; Iqbal, Z.; Raza, A.; Al-Sadi, A.M. Next-generation sequencing and the CRISPR-Cas nexus: A molecular plant virology perspective. Front. Microbiol. 2021, 11, 609376. [Google Scholar] [CrossRef]
  15. Wolter, F.; Puchta, H. The CRISPR/Cas revolution reaches the RNA world: Cas13, a new Swiss Army knife for plant biologists. Plant J. 2018, 94, 767–775. [Google Scholar] [CrossRef]
  16. Schindele, P.; Wolter, F.; Puchta, H. Transforming plant biology and breeding with CRISPR/Cas9, Cas12 and Cas13. FEBS Lett. 2018, 592, 1954–1967. [Google Scholar] [CrossRef]
  17. Aksoy, E.; Yildirim, K.; Kavas, M.; Kayihan, C.; Yerlikaya, B.A.; Çalik, I.; Sevgen, I.; Demirel, U. General guidelines for CRISPR/Cas-based genome editing in plants. Mol. Biol. Rep. 2022, 49, 12151–12164. [Google Scholar] [CrossRef] [PubMed]
  18. Gerashchenkov, G.A.; Rozhnova, N.A.; Kuluev, B.R.; Kiryanova, O.Y.; Gumerova, G.R.; Knyazev, A.V.; Vershinina, Z.R.; Mikhailova, E.V.; Chemeris, D.A.; Matniyazov, R.T.; et al. Design of guide RNA for CRISPR/Cas plant genome editing. Mol. Biol. 2020, 54, 29–50. [Google Scholar] [CrossRef]
  19. Feng, Z.; Zhang, B.; Ding, W.; Liu, X.; Yang, D.-L.; Wei, P.; Cao, F.; Zhu, S.; Zhang, F.; Mao, Y.; et al. Efficient genome editing in plants using a CRISPR/Cas system. Cell Res. 2013, 23, 1229–1232. [Google Scholar] [CrossRef] [PubMed]
  20. Makarova, K.S.; Aravind, L.; Wolf, Y.I.; Koonin, E.V. Unification of Cas protein families and a simple scenario for the origin and evolution of CRISPR-Cas systems. Biol. Direct 2011, 6, 38. [Google Scholar] [CrossRef]
  21. Sandhya, D.; Jogam, P.; Allini, V.R.; Abbagani, S.; Alok, A. The present and potential future methods for delivering CRISPR/Cas9 components in plants. J. Genet. Eng. Biotechnol. 2020, 18, 25. [Google Scholar] [CrossRef]
  22. Miller, K.; Eggenberger, A.L.; Lee, K.; Liu, F.; Kang, M.; Drent, M.; Ruba, A.; Kirscht, T.; Wang, K.; Jiang, S. An improved biolistic delivery and analysis method for evaluation of DNA and CRISPR-Cas delivery efficacy in plant tissue. Sci. Rep. 2021, 11, 7695. [Google Scholar] [CrossRef]
  23. Dixit, S.; Kumar, A.; Srinivasan, K.; Vincent, P.D.R.; Ramu Krishnan, N. Advancing genome editing with artificial intelligence: Opportunities, challenges, and future directions. Front. Bioeng. Biotechnol. 2024, 11, 1335901. [Google Scholar] [CrossRef]
  24. Fong, J.H.; Wong, A.S. Advancing CRISPR/Cas gene editing with machine learning. Curr. Opin. Biomed. Eng. 2023, 28, 100477. [Google Scholar] [CrossRef]
  25. Sherkatghanad, Z.; Abdar, M.; Charlier, J.; Makarenkov, V. Using traditional machine learning and deep learning methods for on-and off-target prediction in CRISPR/Cas9: A review. Brief. Bioinform. 2023, 24, bbad131. [Google Scholar] [CrossRef] [PubMed]
  26. Kim, M.G.; Go, M.J.; Kang, S.H.; Jeong, S.H.; Lim, K. Revolutionizing CRISPR technology with artificial intelligence. Exp. Mol. Med. 2025, 57, 1419–1431. [Google Scholar] [CrossRef] [PubMed]
  27. Chuai, G.; Ma, H.; Yan, J.; Chen, M.; Hong, N.; Xue, D.; Zhou, C.; Zhu, C.; Chen, K.; Duan, B.; et al. DeepCRISPR: Optimized CRISPR guide RNA design by deep learning. Genome Biol. 2018, 19, 80. [Google Scholar] [CrossRef]
  28. Wang, J.; Zhang, X.; Cheng, L.; Luo, Y. An overview and metanalysis of machine and deep learning-based CRISPR gRNA design tools. RNA Biol. 2020, 17, 13–22. [Google Scholar] [CrossRef]
  29. Xue, L.; Tang, B.; Chen, W.; Luo, J. Prediction of CRISPR sgRNA activity using a deep convolutional neural network. J. Chem. Inf. Model. 2018, 59, 615–624. [Google Scholar] [CrossRef] [PubMed]
  30. Luo, J.; Chen, W.; Xue, L.; Tang, B. Prediction of activity and specificity of CRISPR-Cpf1 using convolutional deep learning neural networks. BMC Bioinform. 2019, 20, 332. [Google Scholar] [CrossRef]
  31. 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, 344–354. [Google Scholar] [CrossRef]
  32. Toufikuzzaman, M.; Hassan Samee, M.A.; Sohel Rahman, M. CRISPR-DIPOFF: An interpretable deep learning approach for CRISPR Cas-9 off-target prediction. Brief. Bioinform. 2024, 25, bbad530. [Google Scholar] [CrossRef] [PubMed]
  33. Niu, R.; Peng, J.; Zhang, Z.; Shang, X. R-CRISPR: A deep learning network to predict off-target activities with mismatch, insertion and deletion in CRISPR-Cas9 system. Genes 2021, 12, 1878. [Google Scholar] [CrossRef]
  34. Wan, Y.; Jiang, Z. TransCrispr: Transformer based hybrid model for predicting CRISPR/Cas9 single guide RNA cleavage efficiency. IEEE/ACM Trans. Comput. Biol. Bioinform. 2021, 20, 1518–1528. [Google Scholar] [CrossRef]
  35. Guan, Z.; Jiang, Z. Transformer-based anti-noise models for CRISPR-Cas9 off-target activities prediction. Brief. Bioinform. 2023, 24, bbad127. [Google Scholar] [CrossRef] [PubMed]
  36. Laxmi, B.; Devi, P.U.M.; Thanjavur, N.; Buddolla, V. The applications of artificial intelligence (AI)-driven tools in virus-like particles (VLPs) research. Curr. Microbiol. 2024, 81, 234. [Google Scholar] [CrossRef]
  37. Yakimovich, A. Machine learning and Artificial Intelligence for the prediction of host–pathogen interactions: A viral case. Infect. Drug Resist. 2021, 14, 3319–3326. [Google Scholar] [CrossRef] [PubMed]
  38. Elste, J.; Saini, A.; Mejia-Alvarez, R.; Mejía, A.; Millán-Pacheco, C.; Swanson-Mungerson, M.; Tiwari, V. Significance of artificial intelligence in the study of virus–host cell interactions. Biomolecules 2024, 14, 911. [Google Scholar] [CrossRef]
  39. Postnikova, O.A.; Nemchinov, L.G. Comparative analysis of microarray data in Arabidopsis transcriptome during compatible interactions with plant viruses. Virol. J. 2021, 9, 101. [Google Scholar] [CrossRef]
  40. Ray, M.; Burman, S.; Meshram, S. A Mini Review on Plant Immune System Dynamics: Modern Insights into Biotic and Abiotic Stress. Phyton 2025, 94, 2285. [Google Scholar] [CrossRef]
  41. Chadoulis, R.T.; Livieratos, I.; Manakos, I.; Spanos, T.; Marouni, Z.; Kalogeropoulos, C.; Kotropoulos, C. 3D-CNN detection of systemic symptoms induced by different Potexvirus infections in four Nicotiana benthamiana genotypes using leaf hyperspectral imaging. Plant Methods 2025, 21, 15. [Google Scholar] [CrossRef]
  42. Chen, H.; Han, Y.; Liu, Y.; Liu, D.; Jiang, L.; Huang, K.; Wang, H.; Guo, L.; Wang, X.; Wang, J.; et al. Classification models for Tobacco Mosaic Virus and Potato Virus Y using hyperspectral and machine learning techniques. Front. Plant Sci. 2023, 14, 1211617. [Google Scholar] [CrossRef]
  43. Zhao, Q.; Feng, Q.; Lu, H.; Li, Y.; Wang, A.; Tian, Q.; Zhan, Q.; Lu, Y.; Zhang, L.; Huang, T.; et al. Pan-genome analysis highlights the extent of genomic variation in cultivated and wild rice. Nat. Genet. 2018, 50, 278–284. [Google Scholar] [CrossRef]
  44. Yang, Y.; Saand, M.A.; Huang, L.; Abdelaal, W.B.; Zhang, J.; Wu, Y.; Li, J.; Sirohi, M.H.; Wang, F. Applications of multi-omics technologies for crop improvement. Front. Plant Sci. 2021, 12, 563953. [Google Scholar] [CrossRef]
  45. Zhang, D.; Zhang, Z.; Unver, T.; Zhang, B. CRISPR/Cas: A powerful tool for gene function study and crop improvement. J. Adv. Res. 2020, 29, 207–221. [Google Scholar] [CrossRef] [PubMed]
  46. Saha, D.; Panda, A.K.; Datta, S. Critical considerations and computational tools in plant genome editing. Heliyon 2024, 11, e41135. [Google Scholar] [CrossRef] [PubMed]
  47. Gao, S.; Yu, T.; Rasheed, A.; Wang, J.; Crossa, J.; Hearne, S.; Li, H. Fast-forwarding plant breeding with deep learning-based genomic prediction. J. Integr. Plant Biol. 2025, 67, 1700–1705. [Google Scholar] [CrossRef]
  48. Kozlov, K.N.; Bankin, M.P.; Semenova, E.A.; Samsonova, M.G. Genomic prediction of plant traits by popular machine learning methods. Vavilov J. Genet. Breed. 2025, 29, 458–466. [Google Scholar] [CrossRef] [PubMed]
  49. Ma, J.; Cheng, Z.; Cao, Y. Artificial Intelligence-Assisted Breeding for Plant Disease Resistance. Int. J. Mol. Sci. 2025, 26, 5324. [Google Scholar] [CrossRef]
  50. Brock, N.; Kaur, N.; Halford, N.G. Advances in genome editing in plants within an evolving regulatory landscape, with a focus on its application in wheat breeding. J. Plant Biochem. Biotechnol. 2025, 34, 599–614. [Google Scholar] [CrossRef]
  51. Arndell, T.; Sharma, N.; Langridge, P.; Baumann, U.; Watson-Haigh, N.S.; Whitford, R. gRNA validation for wheat genome editing with the CRISPR-Cas9 system. BMC Biotechnol. 2019, 19, 71. [Google Scholar] [CrossRef]
  52. Zitnik, M.; Nguyen, F.; Wang, B.; Leskovec, J.; Goldenberg, A.; Hoffman, M.M. Machine Learning for Integrating Data in Biology and Medicine: Principles, Practice, and Opportunities. Inf. Fusion 2019, 50, 71–91. [Google Scholar] [CrossRef]
  53. Uddin, M.; Wang, Y.; Woodbury-Smith, M. Artificial intelligence for precision medicine in neurodevelopmental disorders. Npj Digit. Med. 2019, 2, 112. [Google Scholar] [CrossRef]
  54. Niu, M.; Lin, Y.; Zou, Q. sgRNACNN: Identifying sgRNA on-target activity in four crops using ensembles of convolutional neural networks. Plant Mol. Biol. 2021, 105, 483–495. [Google Scholar] [CrossRef]
  55. Son, H. Harnessing CRISPR/Cas Systems for DNA and RNA Detection: Principles, Techniques, and Challenges. Biosensors 2024, 14, 460. [Google Scholar] [CrossRef] [PubMed]
  56. Yu, H.; Cheng, X.; Chen, C.; Heidari, A.A.; Liu, J.; Cai, Z.; Chen, H. Apple leaf disease recognition method with improved residual network. Multimed. Tools Appl. 2022, 81, 7759–7782. [Google Scholar] [CrossRef]
  57. Zhang, G.; Dai, Z.; Dai, X. A Novel Hybrid CNN-SVR for CRISPR/Cas9 Guide RNA Activity Prediction. Front. Genet. 2020, 10, 1303. [Google Scholar] [CrossRef]
  58. Zhang, G.; Luo, Y.; Dai, X.; Dai, Z. Benchmarking deep learning methods for predicting CRISPR/Cas9 sgRNA on-and off-target activities. Brief. Bioinform. 2023, 24, bbad333. [Google Scholar] [CrossRef] [PubMed]
  59. Das, B.S.; Mishra, B.S.P.; Tiwar, A.K.; Panda, B. Optimal Trained Deep LSTM Model for Detecting Plant Leave Diseases. Int. J. Eng. Res. Technol. (IJERT) 2023, 12. [Google Scholar] [CrossRef]
  60. Jasieniecka, A.; Domingues, I. CRISPR-Cas9 and Its Bioinformatics Tools: A Systematic Review. Curr. Issues Mol. Biol. 2025, 47, 307. [Google Scholar] [CrossRef]
  61. Kim, H.K.; Min, S.; Song, M.; Jung, S.; Choi, J.W.; Kim, Y.; Lee, S.; Yoon, S.; Kim, H. Deep learning improves prediction of CRISPR–Cpf1 guide RNA activity. Nat. Biotechnol. 2018, 36, 239–241. [Google Scholar] [CrossRef]
  62. Chen, P.; Wu, Y.; Wang, H.; Liu, H.; Zhou, J.; Chen, J.; Lei, J.; Sun, Z.; Paek, C.; Yin, L. Highly parallel profiling of the activities and specificities of Cas12a variants in human cells. Nat. Commun. 2025, 16, 3022. [Google Scholar] [CrossRef]
  63. Li, C.; Zou, Q.; Li, J.; Feng, H. Prediction of CRISPR-Cas9 on-target activity based on a hybrid neural network. Comput. Struct. Biotechnol. J. 2025, 27, 2098–2106. [Google Scholar] [CrossRef] [PubMed]
  64. Yang, Y.; Li, J.; Zou, Q.; Ruan, Y.; Feng, H. Prediction of CRISPR-Cas9 off-target activities with mismatches and indels based on hybrid neural network. Comput. Struct. Biotechnol. J. 2023, 21, 5039–5048. [Google Scholar] [CrossRef] [PubMed]
  65. Kumar, A.; Anju, T.; Kumar, S.; Chhapekar, S.S.; Sreedharan, S.; Singh, S.; Choi, S.R.; Ramchiary, N.; Lim, Y.P. Integrating omics and gene editing tools for rapid improvement of traditional food plants for diversified and sustainable food security. Int. J. Mol. Sci. 2021, 22, 8093. [Google Scholar] [CrossRef]
  66. Chari, R.; Yeo, N.C.; Chavez, A.; Church, G.M. sgRNA Scorer 2.0: A Species-Independent Model To Predict CRISPR/Cas9 Activity. ACS Synth. Biol. 2017, 6, 902–904. [Google Scholar] [CrossRef]
  67. Naim, F.; Shand, K.; Hayashi, S.; O’Brien, M.; McGree, J.; Johnson, A.A.T.; Dugdale, B.; Waterhouse, P.M. Are the current gRNA ranking prediction algorithms useful for genome editing in plants? PLoS ONE 2020, 15, e0227994. [Google Scholar] [CrossRef]
  68. Ali, Z.; Mahfouz, M.M. CRISPR/Cas systems versus plant viruses: Engineering plant immunity and beyond. Plant Physiol. 2021, 186, 1770–1785. [Google Scholar] [CrossRef] [PubMed]
  69. Tripathi, L.; Ntui, V.O.; Tripathi, J.N.; Kumar, P.L. Application of CRISPR/Cas for diagnosis and management of viral diseases of banana. Front. Microbiol. 2021, 11, 609784. [Google Scholar] [CrossRef]
  70. Ali, Z.; Abulfaraj, A.; Idris, A.; Ali, S.; Tashkandi, M.; Mahfouz, M.M. CRISPR/Cas9-mediated viral interference in plants. Genome Biol. 2015, 16, 238. [Google Scholar] [CrossRef]
  71. Maree, H.J.; Gardner, H.F.; Freeborough, M.J.; Burger, J.T. Mapping of the 5′ terminal nucleotides of Grapevine leafroll-associated virus 3 sgRNAs. Virus Res. 2010, 151, 252–255. [Google Scholar] [CrossRef]
  72. Liu, J.; Carino, E.; Bera, S.; Gao, F.; May, J.P.; Simon, A.E. Structural analysis and whole genome mapping of a new type of plant virus subviral RNA: Umbravirus-like associated RNAs. Viruses 2021, 13, 646. [Google Scholar] [CrossRef] [PubMed]
  73. Ruiz, L.; Simón, A.; García, C.; Velasco, L.; Janssen, D. First natural crossover recombination between two distinct species of the family Closteroviridae leads to the emergence of a new disease. PLoS ONE 2018, 13, e0198228. [Google Scholar] [CrossRef]
  74. Zaidi, S.S.E.A.; Mahas, A.; Vanderschuren, H.; Mahfouz, M.M. Engineering crops of the future: CRISPR approaches to develop climate-resilient and disease-resistant plants. Genome Biol. 2020, 21, 289. [Google Scholar] [CrossRef] [PubMed]
  75. Wessels, H.-H.; Stirn, A.; Méndez-Mancilla, A.; Kim, E.J.; Hart, S.K.; Knowles, D.A.; Sanjana, N.E. Prediction of on-target and off-target activity of CRISPR–Cas13d guide RNAs using deep learning. Nat. Biotechnol. 2024, 42, 628–637. [Google Scholar] [CrossRef]
  76. Montague, T.G.; Cruz, J.M.; Gagnon, J.A.; Church, G.M.; Valen, E. CHOPCHOP: A CRISPR/Cas9 and TALEN web tool for genome editing. Nucleic Acids Res. 2014, 42, W401–W407. [Google Scholar] [CrossRef]
  77. Shashikala, T.; Nagesha, S.; Asokan, R.; Venkataravanappa, V.; Shyamalamma, S. Designing and Validation of sgRNA for CRISPR/Cas12a-based Diagnosis of Tomato Leaf Curl Virus (ToLCV). Mysore J. Agric. Sci. 2025, 59, 302–310. [Google Scholar]
  78. 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, 1473–1475. [Google Scholar] [CrossRef]
  79. Wang, X.; Tu, M.; Wang, Y.; Yin, W.; Zhang, Y.; Wu, H.; Gu, Y.; Li, Z.; Xi, Z.; Wang, X. Whole-genome sequencing reveals rare off-target mutations in CRISPR/Cas9-edited grapevine. Hortic. Res. 2021, 8, 1–11. [Google Scholar] [CrossRef]
  80. Wu, Y.; Ren, Q.; Zhong, Z.; Liu, G.; Han, Y.; Bao, Y.; Liu, L.; Xiang, S.; Liu, S.; Tang, X.; et al. Genome-wide analyses of PAM-relaxed Cas9 genome editors reveal substantial off-target effects by ABE8e in rice. Plant Biotechnol. J. 2022, 20, 1670–1682. [Google Scholar] [CrossRef]
  81. Xu, W.; Fu, W.; Zhu, P.; Li, Z.; Wang, C.; Wang, C.; Zhang, Y.; Zhu, S. Comprehensive analysis of CRISPR/Cas9-mediated mutagenesis in arabidopsis thaliana by genome-wide sequencing. Int. J. Mol. Sci. 2019, 20, 4125. [Google Scholar] [CrossRef] [PubMed]
  82. Das, J.; Kumar, S.; Mishra, D.C.; Chaturvedi, K.K.; Paul, R.K.; Kairi, A. Machine learning in the estimation of CRISPR-Cas9 cleavage sites for plant system. Front. Genet. 2023, 13, 1085332. [Google Scholar] [CrossRef] [PubMed]
  83. Lee, M. Deep learning in CRISPR-Cas systems: A review of recent studies. Front. Bioeng. Biotechnol. 2023, 11, 1226182. [Google Scholar] [CrossRef] [PubMed]
  84. Murmu, S.; Chaurasia, H.; Guha Majumdar, S.; Rao, A.R.; Rai, A.; Archak, S. Prediction of protein–protein interactions between anti-CRISPR and CRISPR-Cas using machine learning technique. J. Plant Biochem. Biotechnol. 2023, 32, 818–830. [Google Scholar] [CrossRef]
  85. Fang, Z.; Feng, T.; Zhou, H.; Chen, M. DeePVP: Identification and classification of phage virion proteins using deep learning. GigaScience 2022, 11, giac076. [Google Scholar] [CrossRef]
  86. Zhang, T.; Zheng, Q.; Yi, X.; An, H.; Zhao, Y.; Ma, S.; Zhou, G. Establishing RNA virus resistance in plants by harnessing CRISPR immune system. Plant Biotechnol. J. 2018, 16, 1415–1423. [Google Scholar] [CrossRef]
  87. Yin, K.; Han, T.; Xie, K.; Zhao, J.; Song, J.; Liu, Y. Engineer complete resistance to Cotton Leaf Curl Multan virus by the CRISPR/Cas9 system in Nicotiana benthamiana. Phytopathol. Res. 2019, 1, 9. [Google Scholar] [CrossRef]
  88. Cao, Y.; Zhou, H.; Zhou, X.; Li, F. Conferring resistance to plant RNA viruses with the CRISPR/CasRx system. Virol. Sin. 2021, 36, 814–817. [Google Scholar] [CrossRef]
  89. Ji, X.; Zhang, H.; Zhang, Y.; Wang, Y.; Gao, C. Establishing a CRISPR–Cas-like immune system conferring DNA virus resistance in plants. Nat. Plants 2015, 1, 15144. [Google Scholar] [CrossRef]
  90. Ghorbani Faal, P.; Farsi, M.; Seifi, A.; Mirshamsi Kakhki, A. Virus-induced CRISPR-Cas9 system improved resistance against tomato yellow leaf curl virus. Mol. Biol. Rep. 2020, 47, 3369–3376. [Google Scholar] [CrossRef]
  91. Xiang, Y.; Dong, X. Translational Regulation of Plant Stress Responses: Mechanisms, Pathways, and Applications in Bioengineering. Annu. Rev. Phytopathol. 2025, 63, 117–146. [Google Scholar] [CrossRef] [PubMed]
  92. Zorzatto, C.; Machado, J.P.B.; Lopes, K.V.G.; Nascimento, K.J.T.; Pereira, W.A.; Brustolini, O.J.B.; Reis, P.A.B.; Calil, I.P.; Deguchi, M.; Sachetto-Martins, G.; et al. NIK1-mediated translation suppression functions as a plant antiviral immunity mechanism. Nature 2015, 520, 679–682. [Google Scholar] [CrossRef]
  93. Liu, F.; Zeng, M.; Sun, Y.; Chen, Z.; Chen, Z.; Wang, L.; Cui, J.-R.; Zhang, F.; Lv, D.; Chen, X.; et al. protects the receptor-like kinase BIR2 from SNIPER2a/b-mediated degradation to promote pattern-triggered immunity in Nicotiana benthamiana. Plant Cell 2023, 35, 3566–3584. [Google Scholar] [CrossRef]
  94. Wen, Z.; Hu, R.; Pi, Q.; Zhang, D.; Duan, J.; Li, Z.; Li, Q.; Zhao, X.; Yang, M.; Zhao, X.; et al. DEAD-box RNA helicase RH20 positively regulates RNAi-based antiviral immunity in plants by associating with SGS3/RDR6 bodies. Plant Biotechnol. J. 2024, 22, 3295–3311. [Google Scholar] [CrossRef] [PubMed]
  95. Uranga, M.; Daròs, J.A. Tools and targets: The dual role of plant viruses in CRISPR–Cas genome editing. Plant Genome 2023, 16, e20220. [Google Scholar] [CrossRef]
  96. Ghosh, D.; Chakraborty, S.; Kodamana, H.; Chakraborty, S. Application of machine learning in understanding plant virus pathogenesis: Trends and perspectives on emergence, diagnosis, host-virus interplay and management. Virol. J. 2022, 19, 42. [Google Scholar] [CrossRef]
  97. Peng, Y.; Dallas, M.M.; Ascencio-Ibáñez, J.T.; Hoyer, J.S.; Legg, J.; Hanley-Bowdoin, L.; Bruce, G.; Yin, H. Early detection of plant virus infection using multispectral imaging and spatial–spectral machine learning. Sci. Rep. 2022, 12, 3113. [Google Scholar] [CrossRef] [PubMed]
  98. Breves, S.S.; Silva, F.A.; Euclydes, N.C.; Saia, T.F.; Jean-Baptiste, J.; Andrade Neto, E.R.; Fontes, E.P. Begomovirus–host interactions: Viral proteins orchestrating intra and intercellular transport of viral DNA while suppressing host defense mechanisms. Viruses 2023, 15, 1593. [Google Scholar] [CrossRef]
  99. Yadav, S.; Chhibbar, A.K. Plant–virus interactions. In Molecular Aspects of Plant-Pathogen Interaction; Springer: Berlin/Heidelberg, Germany, 2018; pp. 43–77. [Google Scholar]
  100. Hipper, C.; Brault, V.; Ziegler-Graff, V.; Revers, F. Viral and cellular factors involved in phloem transport of plant viruses. Front. Plant Sci. 2013, 4, 154. [Google Scholar] [CrossRef] [PubMed]
  101. Iksat, N.; Madirov, A.; Artykbayeva, D.; Shevchenko, O.; Zhanassova, K.; Baikarayev, Z.; Masalimov, Z. Heat Stress Induces Partial Resistance to Tomato Bushy Stunt Virus in Nicotiana benthamiana Via Combined Stress Pathways. Viruses 2025, 17, 1250. [Google Scholar] [CrossRef]
  102. Kamal, H.; Minhas, F.A.; Tripathi, D.; Abbasi, W.A.; Hamza, M.; Mustafa, R.; Khan, M.Z.; Mansoor, S.; Pappu, H.R.; Amin, I. βC1, pathogenicity determinant encoded by Cotton leaf curl Multan betasatellite, interacts with calmodulin-like protein 11 (Gh-CML11) in Gossypium hirsutum. PLoS ONE 2019, 14, e0225876. [Google Scholar] [CrossRef]
  103. Shukla, D.; Alanazi, A.M.; Panda, S.P.; Dwivedi, V.D.; Kamal, M.A. Unveiling the antiviral potential of Plant compounds from the Meliaceae family against the Zika virus through QSAR modeling and MD simulation analysis. J. Biomol. Struct. Dyn. 2024, 42, 11064–11079. [Google Scholar] [CrossRef] [PubMed]
  104. Asim, M.N.; Fazeel, A.; Ibrahim, M.A.; Dengel, A.; Ahmed, S. MP-VHPPI: Meta predictor for viral host protein-protein interaction prediction in multiple hosts and viruses. Front. Med. 2022, 9, 1025887. [Google Scholar] [CrossRef] [PubMed]
  105. Murmu, S.; Chaurasia, H.; Rao, A.; Rai, A.; Jaiswal, S.; Bharadwaj, A.; Yadav, R.; Archak, S. PlantPathoPPI: An Ensemble-based Machine Learning Architecture for Prediction of Protein-Protein Interactions between Plants and Pathogens. J. Mol. Biol. 2025, 437, 169093. [Google Scholar] [CrossRef]
  106. Zhang, M.; Zhang, L.; Liu, T.; Feng, H.; He, Z.; Li, F.; Zhao, J.; Liu, H. CBIL-VHPLI: A model for predicting viral-host protein-lncRNA interactions based on machine learning and transfer learning. Sci. Rep. 2024, 14, 17549. [Google Scholar] [CrossRef]
  107. Gu, Q.; Sheng, L.; Zhang, T.; Lu, Y.; Zhang, Z.; Zheng, K.; Hu, H.; Zhou, H. Early detection of tomato spotted wilt virus infection in tobacco using the hyperspectral imaging technique and machine learning algorithms. Comput. Electron. Agric. 2019, 167, 105066. [Google Scholar] [CrossRef]
  108. Gao, Z.; Huang, J.; Chen, J.; Shao, T.; Ni, H.; Cai, H. Deep transfer learning-based computer vision for real-time harvest period classification and impurity detection of Porphyra haitnensis. Aquac. Int. 2024, 32, 5171–5198. [Google Scholar] [CrossRef]
  109. Bhaskar, N.; Tupe-Waghmare, P.; Shetty, P.; Shetty, S.S.; Rai, T. A deep learning hybrid approach for automated leaf disease identification in paddy crops. In Proceedings of the 2024 International Conference on Distributed Computing and Optimization Techniques (ICDCOT), Bangalore, India, 15–16 March 2024; pp. 1–5. [Google Scholar]
  110. Soni, A.; Kushvaha, R.P.; Snehi, S.K. Current strategies for management of plant viruses and future perspectives: Enhancing crop health, yield and productivity. Asian J. Biochem. Genet. Mol. Biol. 2021, 16, 21–34. [Google Scholar] [CrossRef]
  111. Kasi Viswanath, K.; Hamid, A.; Ateka, E.; Pappu, H.R. CRISPR/Cas, multiomics, and RNA interference in virus disease management. Phytopathology 2023, 113, 1661–1676. [Google Scholar] [CrossRef]
  112. Tugrul, B.; Elfatimi, E.; Eryigit, R. Convolutional neural networks in detection of plant leaf diseases: A review. Agriculture 2022, 12, 1192. [Google Scholar] [CrossRef]
  113. Ashtagi, R.; Mane, D.; Deore, M.; Maranur, J.R.; Hosmani, S. Combined deep learning and machine learning models for the prediction of stages of melanoma. J. Auton. Intell. 2024, 7. [Google Scholar] [CrossRef]
  114. Ghosh, D.; Malavika, M.; Chakraborty, S. Impact of viral silencing suppressors on plant viral synergism: A global agro-economic concern. Appl. Microbiol. Biotechnol. 2021, 105, 6301–6313. [Google Scholar] [CrossRef] [PubMed]
  115. Zhanassova, K.; Satkanov, M.; Samat, A.; Iksat, N.; Bekturova, A.; Zhamanbayeva, M.; Kurmanbayeva, A.; Masalimov, Z. Short-term high temperature stress in plants: Stress markers and cell signaling. Casp. J. Environ. Sci. 2025, 23, 805–844. [Google Scholar]
  116. Jumper, J.; Evans, R.; Pritzel, A.; Green, T.; Figurnov, M.; Ronneberger, O.; Tunyasuvunakool, K.; Bates, R.; Žídek, A.; Potapenko, A.; et al. Applying and improving AlphaFold at CASP14. Proteins Struct. Funct. Bioinform. 2021, 89, 1711–1721. [Google Scholar] [CrossRef] [PubMed]
  117. Baek, M.; Anishchenko, I.; Humphreys, I.R.; Cong, Q.; Baker, D.; DiMaio, F. Efficient and accurate prediction of protein structure using RoseTTAFold2. BioRxiv 2023. preprint. [Google Scholar] [CrossRef]
  118. Lin, Z.; Akin, H.; Rao, R.; Hie, B.; Zhu, Z.; Lu, W.; Smetanin, N.; Verkuil, R.; Kabeli, O.; Shmueli, Y.; et al. Evolutionary-scale prediction of atomic-level protein structure with a language model. Science 2023, 379, 1123–1130. [Google Scholar] [CrossRef] [PubMed]
  119. Wen, Y.; Wei, H.; You, H.; Liu, J.; Wu, H.; Liu, Y. Engineering CRISPR/Cas Systems for High-Specific Nucleic Acid Detection: Innovations, Challenges and Opportunities. Anal. Sens. 2025. [Google Scholar] [CrossRef]
  120. Tang, N.; Ji, Q. Miniature CRISPR-Cas12 systems: Mechanisms, engineering, and genome editing applications. ACS Chem. Biol. 2024, 19, 1399–1408. [Google Scholar] [CrossRef]
  121. Jin, S.; Wu, Q.; Fu, G.; Lu, D.; Wang, F.; Deng, L.; Nie, K. Breaking Evolution’s Ceiling: AI-Powered Protein Engineering. Catalysts 2025, 15, 842. [Google Scholar] [CrossRef]
  122. Chen, G.; Hou, L.; Li, Z.; Xie, B.; Liu, Y. A new strategy for Cas protein recognition based on graph neural networks and SMILES encoding. Sci. Rep. 2025, 15, 15236. [Google Scholar] [CrossRef]
  123. Burman, N.; Belukhina, S.; Depardieu, F.; Wilkinson, R.A.; Skutel, M.; Santiago-Frangos, A.; Graham, A.B.; Livenskyi, A.; Chechenina, A.; Morozova, N.; et al. A virally encoded tRNA neutralizes the PARIS antiviral defence system. Nature 2024, 634, 424–431. [Google Scholar] [CrossRef]
  124. Karimi, M.; Ghorbani, A.; Niazi, A.; Rostami, M.; Tahmasebi, A. CRISPR-Cas13a as a next-generation tool for rapid and precise plant RNA virus diagnostics. Plant Methods 2025, 21, 83. [Google Scholar] [CrossRef] [PubMed]
  125. Roy, B.G.; Choi, J.; Fuchs, M.F. Predictive modeling of proteins encoded by a plant virus sheds a new light on their structure and inherent multifunctionality. Biomolecules 2024, 14, 62. [Google Scholar] [CrossRef] [PubMed]
  126. Yao, J.; Wang, X. Artificial intelligence in de novo protein design. Med. Nov. Technol. Devices 2025, 26, 100366. [Google Scholar] [CrossRef]
Figure 1. Schematic overview of the application of artificial intelligence (AI) and machine learning (ML) in the development of virus-resistant plants using CRISPR-based genome editing. The process begins with the identification of the virus and proceeds to comprehensive data collection and analysis, including the viral genome, host plant genome/transcriptome, multi-omics data, and cultivation conditions. AI and deep learning (DL) methods are employed for Cas protein optimization, modeling virus–plant protein–protein interactions, and designing sgRNAs. These models are validated through train/test splitting and cross-validation techniques, using performance metrics such as accuracy, F1 score, and ROC-AUC. The selected sgRNAs and optimized CRISPR constructs are delivered to plants, followed by confirmation of successful genome editing. The final step involves the creation of resistant plants and ethical/regulatory analysis to ensure compliance with biosafety and bioethics standards.
Figure 1. Schematic overview of the application of artificial intelligence (AI) and machine learning (ML) in the development of virus-resistant plants using CRISPR-based genome editing. The process begins with the identification of the virus and proceeds to comprehensive data collection and analysis, including the viral genome, host plant genome/transcriptome, multi-omics data, and cultivation conditions. AI and deep learning (DL) methods are employed for Cas protein optimization, modeling virus–plant protein–protein interactions, and designing sgRNAs. These models are validated through train/test splitting and cross-validation techniques, using performance metrics such as accuracy, F1 score, and ROC-AUC. The selected sgRNAs and optimized CRISPR constructs are delivered to plants, followed by confirmation of successful genome editing. The final step involves the creation of resistant plants and ethical/regulatory analysis to ensure compliance with biosafety and bioethics standards.
Genes 16 01258 g001
Table 1. Comparison of sgRNA prediction tools for CRISPR/Cas in plants.
Table 1. Comparison of sgRNA prediction tools for CRISPR/Cas in plants.
NameGoalModelLearning Type+OrganismsMetricsMetrics (Plants)
sgRNACNNPrediction of sgRNA efficiencyHybrid CNNDeep Learning (Keras/TensorFlow)Trained on plant data, high accuracyDomain sensitive, requires transfer learningA. thaliana, O. sativa,
Z. mays, S. lycopersicum
ROC-AUC ~0.85, Spearman ~0.70ROC-AUC ~0.85, Spearman ~0.70
Positional classifiersOn-target activityGradient BoostingClassical MLInterpretability, low computational requirementsThey do not take into account 3D chromatin and epigenetics.O. sativa, Z. mays,
A. thaliana
Spearman ~0.55Spearman ~0.45–0.55
DeepCpf1Prediction of Cas12a activityCNN + FC LayersDeep Learning (TensorFlow)Accuracy for Cas12aNo plant data, reduced accuracyO. sativaROC-AUC ~0.82, R2 ~0.65AUC ~0.55–0.60
sgRNA Scorer 2.0Predicting gRNA efficiency for Cas9 in different genomesGradient BoostingClassical MLSimplicity, accessibilityHeuristic model, no plant dataO. sativa,
N. benthamiana
Correlation ~0.70Correlation ~0.30–0.40
E-CRISPsgRNA generation and scoringRule-basedRule-basedSimplicity, speedDoes not take epigenetics into accountA. thaliana, O. sativaCorr ~0.35~0.30–0.35
CHOPCHOPA universal web tool for sgRNARule-basedRule-basedSupport for many Cas, simplicityPrimitive on-target metricsN. benthamiana,
S. lycopersicum,
O. sativa
Corr ~0.30Correlation ~0.30
Cas-OFFinderOff-target searchExhaustive search algorithmno MLFlexibility, PAM support, speedNo consideration of the biocontextZ. mays, G. max,
N. benthamiana
-Greatly overestimates the risks
CRISPR Genome-wide analysisGenomic screeningPipeline-basedno MLGenomic coverageNo ML, not adapted to plantsC. annuum L.,
O. Sativa, A. thaliana
--
Table 2. Application of AI to design Cas proteins against plant viruses.
Table 2. Application of AI to design Cas proteins against plant viruses.
Cas ProteinTarget VirusPlantAIEfficiency+
FnCas9CMV, TMVN. benthamiana,
A. thaliana
CNN for assessing FnCas9-RNA interactionsDecreased viral RNA levels, inheritedRNA viruses, no need for DNA editingPotential off-target for RNA
SpCas9CLCuMuVN. benthamiana,
A. thaliana
Deep-RPA (1D Convolutional Neural Network)99% accuracy in predicting off-target sitesTargeting multiple sitesRisk of mosaic mutation, Limited to model plants
SaCas9
(modified)
TYLCV, TMVS. lycopersicum,
N. benthamiana
GUIDE-seq + Deep Learning (CNN for off-target)Off-target reduction without loss of activity (~80–90%)Improved specificity; suitable for vectorsNot tested in plants; obtained in animal models
CasRx (RfxCas13d)ssRNA virusesN. benthamianaAttention network for RNA structureSuppression of viral expression by more than 80%High specificity, small sizeHEPN domain activity
Cas13a
(codon-optimized)
TuMVN. benthamianaDeepCodon (DL for codon optimization)Cas13a expression increased 2.3-fold; viral load decreasedEnhanced expression through DL optimizationLimited to plants with PVX infection
Cas12a
(modified)
TYLCVS.lycopersicumCRISPR-GAN (DL for generating Cas12a variants)Cas12a activity increased by 40% compared to wild typeExtended PAM profilesHigh computational costs
Table 3. Tools for analyzing virus–plant interactions.
Table 3. Tools for analyzing virus–plant interactions.
NameType of LearningApplicationPlanteTarget Virus+Metrics (Accuracy)
CBIL-VHPLICNN + BiLSTM + Transfer LearningPrediction of viral protein PPIs from lncRNAG. hirsutumCLCuMB High accuracy,
generalizability,
adaptability to new data
Requires large training samples, high computational loadAccuracy: 91.6%, Precision: ~93%
SVM + Gradient BoostingML, transcriptome classificationDetermination of resistance markers at the expression levelS.lycopersicumTSWVBiomarker detection,
integration with climate factors
Sensitive to sampling,
requires annotated data
Accuracy: ~89%
CNN + Random ForestHybrid Deep Learning + MLClassification of images of viral infection symptomsN. tabacumTMVResistance to visual noise, high F1 metricDependence on image quality and domainAccuracy: 95%, F1: ~0.94
Decision Tree + SVMML on genomic SNP dataDetection of susceptibility alleles (eIF(iso)4G)O.sativaRTBVDetection of functionally significant mutationsFurther validation is needed (CRISPR)F1: ~0.88
Table 4. Application of generative AI models for designing Cas proteins against plant viruses.
Table 4. Application of generative AI models for designing Cas proteins against plant viruses.
AI ModelGoalVirus/PlantEfficiency+
AlphaFold2Prediction of 3D structures of Cas proteins (Cas9, Cas12a) and TYLCV virus proteins for the selection of interaction interfacesTYLCV/S. lycopersicumRMSD < 2Å, high structural accuracy; used to create stable complexes with targeted mutationsAllows for clarification of Cas protein interactions with viral DNA, speeding up the design cycleDoes not predict time-scale dynamics; subsequent molecular dynamics simulations are required
RoseTTAFoldEngineering Cas13a for RNA viruses, modeling the complex structure of Cas13-viral RNARTBV/O. sativaRMSD 1.8–2.3Å; the accuracy of the complex prediction has been
confirmed experimentally
Accounting for intermolecular interfaces, support for multi-chain modelingHigh computational load, limited training set of plant viruses
ProGen2Generation of new Cas protein variants with altered PAM recognitionCLCuV/G. hirsutum~30% of generated sequences
retained functionality in vitro
The possibility of creating non-standard Cas proteins with new specificitiesLow accuracy without filtering; further structure validation required
ESMFold (Meta AI)Rapid structure prediction of modified Cas proteins to improve stability in plant cytoplasmTMV/N. tabacumMSD ~2.1Å; models were used to screen proteins with high resistance to degradationhigh speed, no alignment requiredNo accurate accounting of interactions with DNA/RNA; requires supplementation with molecular dynamics
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Iksat, N.; Madirov, A.; Zhanassova, K.; Masalimov, Z. Artificial Intelligence-Assisted CRISPR/Cas Systems for Targeting Plant Viruses. Genes 2025, 16, 1258. https://doi.org/10.3390/genes16111258

AMA Style

Iksat N, Madirov A, Zhanassova K, Masalimov Z. Artificial Intelligence-Assisted CRISPR/Cas Systems for Targeting Plant Viruses. Genes. 2025; 16(11):1258. https://doi.org/10.3390/genes16111258

Chicago/Turabian Style

Iksat, Nurgul, Almas Madirov, Kuralay Zhanassova, and Zhaksylyk Masalimov. 2025. "Artificial Intelligence-Assisted CRISPR/Cas Systems for Targeting Plant Viruses" Genes 16, no. 11: 1258. https://doi.org/10.3390/genes16111258

APA Style

Iksat, N., Madirov, A., Zhanassova, K., & Masalimov, Z. (2025). Artificial Intelligence-Assisted CRISPR/Cas Systems for Targeting Plant Viruses. Genes, 16(11), 1258. https://doi.org/10.3390/genes16111258

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop