Next Article in Journal
Nutrient-Element-Mediated Alleviation of Cadmium Stress in Plants: Mechanistic Insights and Practical Implications
Previous Article in Journal
Morphological Acclimation of Durum Wheat Spikes in Response to Foliar Micronutrient Applications
Previous Article in Special Issue
A Protoplast System for CRISPR-Cas Ribonucleoprotein Delivery in Pinus taeda and Abies fraseri
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Turning Susceptibility into Strength: A New Era of Durable Resistance in Plants Through Genome Editing

Tropical Research and Education Centre, Horticultural Sciences Department, University of Florida, IFAS, Homestead, FL 33031, USA
*
Author to whom correspondence should be addressed.
Plants 2025, 14(19), 3080; https://doi.org/10.3390/plants14193080
Submission received: 31 July 2025 / Revised: 3 September 2025 / Accepted: 28 September 2025 / Published: 5 October 2025

Abstract

In plants, resistance genes (R) are key players in combatting diseases caused by various phytopathogens. Typically, resistance relies on detecting a single pathogen-derived molecular pattern. However, R-gene-mediated resistance is often race specific, follows the gene-for-gene hypothesis, and can be overcome in field conditions as pathogens evolve. On the contrary, altering plant susceptibility genes (S-genes) facilitates compatibility and results in broad and durable resistance. S-genes are negative regulators present in plants and exploited by pathogens to facilitate their growth and cause infection. Several studies across crop species have reported manipulation of S-genes using genome editing to confer broad spectrum resistance. This review focuses on the plant defense mechanism against biotic stress, R-genes vs. S-genes, different types/classes of S-genes, different tools for S-gene discovery, and the use of gene editing technologies to target S-genes in addition to their applications, challenges, and future perspectives.

1. Introduction

Crop yields are reduced by 40% each year due to biotic stressors such as insect pests and pathogens [1]. Estimates indicate a need to increase food productivity by 70% to feed an exponentially growing global population of 10 billion by 2050 [2]. Achieving this ambitious goal will require not only boosting agricultural production but also minimizing the losses caused by various biotic stresses [1]. With rising global temperatures, the frequency of disease outbreaks is expected to increase, which could hamper productivity and pose major challenges to food security [3]. Therefore, there is a pressing need to explore ways to enhance plants’ tolerance to various biotic stresses.
Biotic stresses in plants are caused by living organisms such as bacteria, viruses, fungi, nematodes, insects, arachnids, and weeds [4]. Plants have naturally evolved to defend themselves against invading pathogens by developing efficient defense mechanisms [5,6,7]. One of the most effective strategies to manage phytopathogens is the development and use of disease-resistant crop varieties. Developing resistant plant varieties usually involves introducing naturally occurring dominant resistance (R) genes into crop cultivars. However, resistance sources are available in the wild germplasm, limiting their applicability [8,9]. Furthermore, R-gene-mediated resistance is frequently overcome by the emergence of new races that evade recognition by altering their cognate effector proteins [10].
Inactivating susceptibility (S) genes, which facilitate pathogen infection and compatibility, offers a promising strategy for durable plant disease resistance [11,12]. Unlike R-gene-mediated resistance, S-gene inactivation provides broad-spectrum and more reliable resistance. In the year 2002, the concept of an S-gene was first explored after the identification of pmr6 (PM resistance) in Arabidopsis. PMR6 was described as “… a novel form of disease resistance based on the loss of a gene required during a compatible interaction …” [13], after which the term “susceptibility gene” was proposed [14]. Inactivation, mutation, or loss of S-gene function leads to disease-resistant plants by disrupting compatible plant–pathogen interactions and slowing pathogen growth and disease progression [11,15]. Many studies have demonstrated the exploitation of S-genes in conferring disease resistance against various biotic stresses [16]. Among S-genes, Mildew Resistance Locus O (MLO) is the most extensively studied, with its role in plant immunity conserved across species. To date, approximately 200 MLO genes have been identified, grouped into seven conserved clades, with Clade IV (monocots) and Clade V (dicots) most associated with powdery mildew (PM) susceptibility. In barley, loss of function mlo alleles have shown durable broad-spectrum resistance against PM for more than five decades, with few pleiotropic effects [17,18]. In addition, similar gene orthologs have been mutated in several crops to confer resistance to PM, including Arabidopsis, wheat, tomato, pea, pepper, grapevine, and strawberry [19,20,21,22,23,24]. This has led to the discovery and characterization of many other MLO orthologs in several plant species, such as Arabidopsis (AtMLO2, AtMLO6, and AtMLO12), barley (TaMlo-A1, TaMlo-B1, and TaMlo-D1), cucumber (CsaMLO8), melon (CmMLO3, CmMLO5, and CmMLO12), C. moschata (CmaMLO1, CmoMLO2, CmoMLO3, CmoMLO4, and CmoMLO5), and C. maxima (CmaMLO1, CmaMLO2, CmaMLO3, CmaMLO4, and CmaMLO5), in addition to tomato (SlMLO1) and pea (Er1/PsMLO1) [19,25,26,27,28].
Another well-characterized S-gene is the eukaryotic initiation factor 4E (eIF4E), which plays a crucial role in viral RNA replication [29]. Inactivation of this gene (eIF4E) leads to resistance against potyvirus, and loss-of-function eIF4E mutants confer resistance in tomato, rice, barley, lettuce, melon, pea, and pepper [30]. Mutating either eIF4E or eIF(iso)4E is often enough to provide potyvirus resistance with minimal or no pleiotropic effects [31]. The eIF4E and MLO examples highlight that the dependency of pathogens on host factors for infection or replication runs the risk of reaching a dead end in the evolutionary arms race. To overcome this, they must either revert and regain a previously lost function or shift to a different host altogether. This concept highlights the potential of S-genes as important targets for resistance breeding.
Previously, RNA interference (RNA-i) and Transcription Activator-Like Effector Nuclease (TALEN)-induced mutagenesis were used to inactivate S-genes [22,23,32,33,34]. With advancements in genome editing (GE) technology, clustered regularly interspaced short palindromic repeats (CRISPR)-associated nucleases (Cas9) have been used successfully to modify S-genes to obtain effective disease-resistant crops. Owing to its specificity, adaptability, ease of use, and cost and labor effectiveness, this transgene-free technology has the potential to create resistant plants against a plethora of microbes by identifying and targeting S-genes in various crop species to tackle the challenge of food security and environmental sustainability [35,36]. This review summarizes plant defense strategies against biotic stress, R-genes vs. S-genes, different types/classes of S-genes, different tools for S-gene discovery, and different S-genes targeted using CRISPR/Cas9 technology, along with their applications, challenges, and future perspectives.

2. Defense Mechanism in Plants Against Biotic Stress

Plants defend themselves via two tiers of receptors against invading pathogens [37,38]. The first layer consists of transmembrane pattern-recognition receptors (PRRs), which are responsible for detecting conserved pathogen-associated molecular patterns (PAMPs) from microbes and damage-associated molecular patterns (DAMPs) from plants [39]. The first line of defense in plants against pathogens is known as pathogen-triggered immunity (PTI) [40]. The activation of PTI is the essential component of plant innate immunity. The recognition of PAMPs by PRRs triggers a cascade of downstream signaling pathways that activate plant defense responses. As a result, a pathogen’s virulence potential depends on its ability to suppress PTI using effector molecules [41]. Activation of PTI initiates signaling cascades involving mitogen-activated protein kinases (MAPKs), transcriptional reprogramming through transcription factors such as WRKY, and the production of various reactive oxygen species (ROS) in the host plant [42]. These signaling cascades also involve key hormones such as salicylic acid (SA) for biotrophs, and jasmonic acid (JA) and ethylene for necrotrophs to enhance resistance and trigger localized hypersensitive responses (HRs), characterized by programmed cell death at infection sites (Figure 1).
In contrast, effector-triggered immunity (ETI) is activated by specific pathogenic effectors, with their corresponding plant receptors showing high specificity and undergoing strong diversifying selection. Most ETI-associated receptors inside host cells are detected by nucleotide-binding/leucine-rich repeat (NLR) receptors and other cytoplasmic proteins [43,44]. ETI responses are typically stronger and more long lasting than PTI and often lead to HR and programmed cell death [45,46]. In ETI, pathogen effectors, known as avirulence (Avr) proteins, are detected by host NLRs, also referred to as R proteins. In simple terms, Avr proteins function as ligands that specifically bind to corresponding NLRs. Activation of ETI triggers an HR and programmed cell death in infected and surrounding cells, helping to limit pathogen spread. Disease occurs when there is a compatible interaction between plant and pathogen. In many cases, plants achieve durable resistance by modifying genes that are essential for compatibility. These genes, which enable pathogen infection and support compatibility, are known as susceptibility (S) genes [11]. Despite these defense mechanisms, a few host susceptibility (S) genes follow an inverse gene-for-gene model. The necrotrophic pathogen utilizes effectors to coopt host programmed cell death known as effector-triggered susceptibility (ETS) [37]. The effectors or host-selective toxins (HSTs) directly interact with sensitivity genes, activate plant defense responses including cell death, and lead to susceptibility. ETS is therefore regarded as a pathogen-driven manipulation of host genes and metabolic pathways, representing the opposite of R-gene-mediated resistance.

3. R-Genes vs. S-Genes

Plant R-genes function by directly or indirectly recognizing conserved pathogen effectors, also known as Avr proteins. Most R-genes encode either surface immune receptors such as receptor-like kinases (RLKs) or intracellular immune receptors [47]. Among these, the nucleotide-binding leucine-rich repeat (NB-LRR or NLR) family represents the largest class of R-genes, characterized by a nucleotide-binding (NB) domain and a leucine-rich repeat (LRR) domain. Based on their N-terminal structural motifs, NLRs are further categorized into Toll/interleukin-1 receptor (TIR)-NB-LRR and coiled-coil (CC)-NB-LRR subclasses [48]. In traditional breeding, disease resistance in plants has been introduced through the incorporation of R-genes. This involves transferring R-genes from closely related wild relatives or compatible species into cultivated varieties [49]. Over the years single or multiple R-genes have been successfully introduced into different crops [49]. However, this strategy has limitations, as pathogen Avr effector proteins frequently mutate, enabling them to evade recognition by host R-gene receptors [50,51,52]. Consequently, plant breeders continually search for new R-gene sources to combat newly emerging pathogens. In this context, CRISPR/Cas-mediated modification or engineering of known NLRs has emerged as a promising strategy for enhancing disease resistance. Editing regulatory elements within NLRs can improve their activation potential, thereby boosting plant immunity [53]. Additionally, modifying the effector recognition sites of NLRs can confer broad-spectrum disease resistance [53]. For example, targeted mutagenesis of the tomato I2 immune receptor successfully expanded effector recognition and provided broad-spectrum resistance [54,55]. Similarly, precise editing of the LRR domain of NLRs using CRISPR/Cas9 can generate novel resistance traits, as the LRR domain plays a critical role in pathogen recognition specificity [56,57,58] (Figure 2).
S-gene editing offers an alternative and effective approach for developing crop cultivars with durable disease resistance. Plant S-genes function as negative regulators of immunity and are frequently exploited by pathogens to enhance their proliferation and initiate infection [59]. Since S-genes are naturally present in the host plant, introgression of foreign genetic material is not required to develop resistance. Knocking out S-genes can confer broad-spectrum disease resistance and prevent pathogens from successfully infecting the host (Figure 2). However, resistance conferred by S-gene loss or modification is usually recessive and may come with some fitness costs [12]. In plants, the balance between R-genes and S-genes largely determines whether a plant is susceptible or resistant to disease. Recent advances in genome editing (GE) technologies have transformed plant science, enabling improvements not only in disease resistance but also in nutritional value and fruit quality traits. Tools such as transcription activator-like effector nucleases (TALENs), zinc-finger nucleases (ZFNs), and especially CRISPR/Cas9 allow precise manipulation of target DNA sequences [60,61,62]. Among these, CRISPR/Cas9 is widely used to study S-gene function and develop disease-resistant cultivars. Although recessive inheritance makes S-gene-based resistance challenging to incorporate through conventional breeding, GE provides a powerful approach to create durable and broad-spectrum resistance, similar to non-host resistance [63].

4. Methods for Identification of S-Genes in Plants

Identifying S-genes, deciphering their functions, and mapping their location within the plant genome are essential steps toward engineering broad-spectrum disease resistance and developing improved crop cultivars [64]. As research into plant–pathogen interactions advances, a range of methodologies have been used to identify and characterize these elusive S-genes. Here, a detailed description of molecular and computational approaches used for the identification of S-genes is presented (Figure 3).

4.1. Quantitative Trait Locus (QTL)

QTL mapping is a process of identifying genomic regions linked to variations in quantitative traits, including disease susceptibility [65]. This process involves the development of suitable mapping populations by crossing plants with contrasting phenotypes (e.g., resistant vs. susceptible) followed by genotyping. Finally, by using appropriate statistical packages to perform linkage analysis by using genotyping and phenotypic information, researchers can identify genomic regions responsible for susceptibility [66]. For example, a single major QTL flanked by two AFLP markers (200011_345Dr and 200011_208Br) governing susceptibility to PM in hop (Humulus lupulus L) has been identified in co-segregating populations [67]. In Lathyrus sativus and L. cicero, the MLO1 gene was recently identified using QTL mapping [68]. Further phylogenetic analysis confirmed that LsMLO1 and LciMLO1 belong to Clade V (associated with susceptibility) [68]. Similarly, QTL mapping and RT-PCR revealed CsGy5G015660 as a strong candidate gene conferring PM resistance in cucumber [66].

4.2. Genome-Wide Association Studies (GWASs)

The GWAS approach is a genetic approach used to identify the genetic variants associated with a specific trait across large natural populations [69,70]. GWASs typically identify broad genomic regions linked to susceptibility and fine mapping techniques are then used to narrow these regions and locate the most likely candidate S-genes [71]. For instance, a recent GWAS in tea (Camellia sinensis) identified the CsNCED1 gene, which encodes 9-cis-epoxycarotenoid dioxygenase, a key enzyme in abscisic acid (ABA) biosynthesis, as a susceptibility factor for biotic stress. Overexpression of CsNCED1 led to reduced salicylic acid levels and downregulation of immune-related genes (e.g., NPR1, PR1, and WRKY18), resulting in greater susceptibility to Spodoptera litura [72].

4.3. Transcriptomic Analysis

Transcriptomics studies are widely used to study gene expression patterns in response to pathogen infection. The transcriptomics profiles of resistant and susceptible genotypes at different time intervals were compared to identify differentially expressed genes, including potential S-genes, that play a role in plant defense mechanisms [73,74,75].
For instance, nine S-gene homologs of CAMTA3, CNGC4, PMR4, PMR5, and PUX2 (S-genes in Arabidopsis have been identified in tobacco using RNA-seq analysis of resistant (BH) and susceptible (XHJ) tobacco cultivars infected with Phytophthora nicotianae [76]. Similarly, CsaMLO1, CsaDMR6-2, and PMR/DMR homologs in cucumber have been identified based on transcriptomic data [77].

4.4. Homology-Based Search

Initially, an extensive literature review was performed to identify genes implicated in plant–pathogen interactions [78,79,80]. With the increasing availability of extensive genomic resources for a wide range of crop species, identifying homologous or functionally similar genes in the genome of interest has become more feasible. Identifying genes through this approach helps narrow down the pool of potential candidate genes [81]. The BLAST sequence similarity search is a major platform for screening high-level similarity or conserved coding regions. Moreover, various in-silico bioinformatics tools are available with many statistical algorithms or computational methods, for instance, data-mining analysis, hidden Markov analysis, cluster analysis, kernel-based data fusion analysis, machine learning, K-Nearest Neighbors (KNN) classification algorithm, and others. To further investigate their roles in disease resistance, various molecular approaches such as gene expression analysis, knockout (KO) or overexpression studies, and protein–protein interaction assays can be employed. Several tools and databases are now available to facilitate the prediction of protein–protein interactions between candidate S-genes and pathogen effectors (Table 1). These platforms primarily incorporate machine learning (ML) and deep learning (DL) models to improve prediction accuracy and efficiency. Using similar approaches, S-genes has been identified in arabidopsis, barley, cucumber, tomato, chestnut, cotton, rubber, poplar, apple, grapevines, and many more [19,23,25,26,27,81,82,83]. Together, these approaches offer an effective way to identify and characterize S-genes, allowing targeted modifications to strengthen plant resistance and improve disease management strategies.

5. Classification and Mechanisms of S-Genes

There are three major classes of S-genes based on their mechanism of action (Figure 4).

5.1. Facilitating Host Recognition and Entry

S-genes enable pathogen recognition, penetration, colonization and initiation of infection through various mechanisms including modifying structural barriers or immune responses. Class I genes are active during the early stages of infection, facilitating pathogen entry and establishing infection through various entry points [11]. For instance, mutants of the Glossy II gene in corn have shown reduced susceptibility to PM due to lower levels of very long-chain aldehydes in the leaf cuticles [110]. Similarly, the ram2 mutant in Medicago truncatula has exhibited decreased susceptibility to Phytophthora palmivora because of altered cutin composition [111]. In another study, the irg1 mutant in Medicago exhibited reduced epicuticular wax levels on the leaf surface, and the mutants showed decreased susceptibility to fungal pathogens including Phakopsora pachyrhizi, Puccinia emaculata, and Colletotrichum trifolii [112]. Similarly, mutants in Arabidopsis genes bre1/lacs2, which are defective in cutin biosynthesis, display increased cuticle permeability. This alteration facilitates the release of antifungal compounds, leading to enhanced resistance against Botrytis cinerea [113,114]. The rat4 mutants in Arabidopsis, with disrupted CSLA9 genes encoding a cellulose-like protein, show reduced susceptibility to Agrobacterium infection, highlighting the role of cell wall components in pathogen recognition [115,116]. Pathogens also exploit natural openings like stomata for entry, a process regulated by genes such as LecRK, RIN4, and AHA1. Mutants in these genes exhibit reduced bacterial entry [117,118,119]. Membrane genes like MLO and BAX inhibitor-1 (BI-1) are essential for haustoria formation in PM pathogens, and their loss of function confers broad resistance [111,120,121]. Similarly, small GTPases like HvRACB in barley and ROP6 in Arabidopsis support susceptibility to adapted pathogens while resisting non-adapted ones [122,123]. In rice, genes such as OsRAC4, OsRAC5, and OsRACB are associated with susceptibility to Magnaporthe oryzae [124,125]. In Arabidopsis thaliana, the ARF-GAP protein AGD5 functions as a susceptibility factor during infection by Hyaloperonospora arabidopsidis [126]. These examples collectively highlight the diverse roles of S-genes in modulating plant structures and physiological processes to facilitate pathogen infection [11].

5.2. Modulating Host Immune Responses

S-genes act as negative regulators in plant immunity that suppress host defense signaling, allowing pathogens to evade immune responses. In plant immunity, S-genes that play a crucial role in modulating defense responses belong to class 2 genes. These genes, while not directly involved in pathogen resistance, influence the plant’s ability to respond effectively to infections. For instance, in Arabidopsis, the PMR4 gene encodes callose synthase, which is involved in cell wall reinforcement during pathogen attack. Mutations in PMR4 lead to enhanced resistance against PM by promoting callose deposition at infection sites, thereby restricting pathogen entry [127]. Similarly, the WRKY45 transcription factor in rice has been identified as an S-gene and the overexpression of WRKY45-1 increases susceptibility to Xanthomonas oryzae, while WRKY45-2 enhances resistance, highlighting the complex roles of WRKY genes in disease susceptibility [128]. In Arabidopsis, the DMR6 gene functions as an S-gene for Downy Mildew (DM). It promotes pathogen growth by degrading SA, a crucial signaling molecule that plays an essential role in enhancing plant defense mechanisms [129,130]. Likewise, begomovirus infection triggers the production of a calmodulin-like protein called rgs-CaM, which acts as a negative regulator of gene silencing. The disruption of RNA silencing machinery by rgs-CaM reduces the transcription of RNA-dependent RNA polymerase 6 (RDR6) and promotes the degradation of suppressor of gene silencing 3 (SGS3) [131,132]. RDR6 plays a key role in amplifying RNA silencing signals, while SGS3 helps maintain their stability and distribution. RDR6 suppression and SGS3 degradation by rgs-CaM weaken the RNA silencing pathway, resulting in reduced signal amplification, increased viral RNA accumulation, and more severe infections [131]. These examples highlight the functions of class 2 S-genes in plant defense, where their modulation can tilt the balance between susceptibility and resistance.

5.3. Sustaining Pathogen Growth Post-Invasion

In plant–pathogen interactions, S-genes that facilitate pathogen establishment by providing essential resources belong to class 3 genes. These genes are often recessive and are exploited by pathogens to enhance their virulence through specialized effectors. These effectors modify plant cellular processes to create a more favorable environment for pathogen growth. Plant–pathogen interactions often hinge on the exploitation of host S-genes, which pathogens hijack to facilitate their invasion and proliferation. For example, in rice, the SWEET family of sugar transporters, such as SWEET11 and SWEET13, are targeted by Xanthomonas oryzae to export sugars into the apoplast, providing essential nutrients for the pathogen’s growth [124]. Similarly, in Arabidopsis, the DMR1 gene, encoding homoserine kinase, is exploited by Hyaloperonospora arabidopsidis, which relies on host-derived amino acids for its development [133,134]. In maize, the lipoxygenase gene ZmLOX3 is involved in the production of oxylipins that promote fungal virulence, however, mutations in ZmLOX3 can increase resistance to certain pathogens [135]. Barley alcohol dehydrogenase (ADH) gene expression is triggered by PM infection, promoting pathogen growth by enhancing anaerobic glycolytic metabolism. In contrast, plants with silenced ADH genes show reduced susceptibility to PM. [136]. PM susceptibility genes like PMR5 and PMR6 encode for pectate lyases, and mutations in these alleles result in accumulation of pectin in cell walls and reduced pathogen proliferation at later infection stages [13,137]. Plant viruses often hijack host translation machinery for replication; for instance, eIF4E and eIF(iso)4E play a key role in potyvirus infection by interacting with the viral protein (VPg). Mutations in these factors can confer resistance to potyviruses, but viruses may adapt by acquiring mutations in VPg to interact with alternative eIF4E isoforms. In Arabidopsis, a mutated eIF4 gene restricted the viral movement within the host and resulted in reduced susceptibility to cucumber mosaic virus (CMV) and turnip crinkle virus (TCV) [138]. Additionally, tonoplast membrane proteins TOM1, TOM2, and TOM3 in Arabidopsis are required for efficient multiplication of tobamoviruses, indicating their essential role in the viral life cycle [139,140,141]. The nine necrotrophic effectors (HSTs) are secreted by Parastagonospora nodorum, the causal agent of P. nodorum leaf blotch in wheat, that target specific host susceptibility loci resulting in host cell death and promoting the necrotrophic lifestyle of the fungus [142,143,144]. The SnTox1 interacts with Snn1 in a light-dependent manner, inducing programmed cell death and enhancing pathogen colonization [145,146]. Notably, SnTox1 also contains a chitin-binding domain that protects fungal cell walls from plant chitinases and suppresses PRR-mediated immune activation, whereas Snn1 encodes a wall-associated kinase involved in resistance to biotrophic pathogens, demonstrating how necrotrophic pathogens hijack these pathways for susceptibility [147,148]. Another effector, SnToxA, is nearly identical to PtrToxA from Pyrenophora tritici-repentis and utilizes the same S-gene, Tsn1, suggesting horizontal gene transfer between pathogens [149]. Additional effectors (SnTox2SnTox8) and their corresponding loci (Snn2Snn8) have also been identified, several of which trigger light-dependent programmed cell death, further subverting host defenses [149,150,151,152,153,154]. Transcription activator-like (TAL) effectors from Xanthomonas, delivered via the type III secretion system, bind specific DNA sequences in host promoters, activating genes that facilitate infection. For instance, TAL effectors such as AvrBs3 and PthXo1 induce expression of the sugar transporter gene OsSWEET14 in rice, promoting bacterial growth by increasing nutrient availability. The TaNCED gene in wheat is activated by TAL effector of Xanthomonas translucens and results in elevated ABA production and alteration of plant stress responses and water flow, creating conditions that favor pathogen survival and spread [155,156]. Similarly, the victorin toxin from the fungus Cochliobolus activates the Arabidopsis gene LOV1, converting it from a resistance gene to a susceptibility factor under certain conditions [157]. Additionally, Pseudomonas effector HopZ2 targets the Arabidopsis gene MLO2, a homolog of the Mlo gene in barley, highlighting the role of effector-targeted class 3 S-genes in pathogen virulence [158]. Understanding how pathogens exploit S-genes is essential for designing effective strategies to improve plant resistance against pathogens.

6. Introgression of S-Gene for Durable Resistance

Classical Breeding Approaches

In many crops, over the years classical mutation breeding has been used for the inactivation of S-genes [59]. In classical breeding, introgression of these mutated S-genes requires multiple backcrosses with the elite parent followed by selfing and selection for resistance and desirable agronomic traits. Such approaches are time consuming, labor-intensive, and even inefficient in eliminating some undesirable traits due to linkage drag. Resistance in plants is commonly assessed through symptom observation in addition to molecular and biochemical analysis, as demonstrated in the case of pea resistance to pea seed-borne mosaic virus (PSbMV) [158]. Traditional breeding for S-genes offers the advantage of broad public acceptance. The most well-studied form of durable recessive resistance (S-gene mutation) is eIF4E-based resistance against potyviruses. The pepper pvr1/2 gene was the first identified source of recessive resistance to potato virus Y (PVY) and has been successfully utilized for many years. [159,160]. Numerous studies have reported the breakdown of eIF4E-based resistance against potyviruses due to frequent mutations in viral proteins (VPs) [161,162,163,164]. Virus replication generally relies on the physical interaction between its VPg and plant eIF4E factors [31,165]. Natural variants of eIF4 factors have been identified in several plants, including lettuce (Lactuca sativa), barley (Hordeum vulgare), pea (Pisum sativum), and pepper (Capsicum annuum) and have been successfully introduced into various genotypes [166,167,168,169].

7. Targeting S-Genes for Durable Resistance

7.1. Targeting eIF4 Genes

Potyviruses can overcome inactivated S-gene resistance by acquiring new specificity for an alternative eIF4 isoform or by completely bypassing the need for eIF4 binding [156]. Only a handful of instances have reported VPs other than VPg being responsible for overcoming the requirement for eIF4E [170,171]. Thus, based on the crop and the specific eIF4E gene used, resistance to certain potyviruses can be achieved. Chandrasekaran et al. (2016) used CRISPR/Cas9 technology to KOeIF4E in cucumber. The edited lines showed resistance to Ipomovirus (CVYV) and potyviruses (ZYMV and PRSV-W) [172]. In another study, eIF(iso)4E in A. thaliana was targeted using CRISPR/Cas9, resulting in transgene-free plants that showed resistance to turnip mosaic virus (TuMV) [173]. Similarly, the eIF4G gene in the susceptible rice variety Oryza sativa cv. IR64 was mutated using CRISPR to create new alleles, leading to the development of a resistant variety against rice tungro spherical virus (RTSV) [74,174]. Editing the eIF4E gene in wheat, particularly in TaeiF4E-aabbdd mutants, provided resistance to wheat yellow mosaic virus (WYMV) without affecting growth, resulting in taller plants, delayed heading, and no yield reduction [175]. Base editing of eIF4E1 by CRISPR/Cas9-cytidine deaminase conferred resistance to clover yellow vein virus (ClYVV) in Arabidopsis [176]. In tomato (cv. M82), eIF4E KO conferred resistance to PVYN, but it remained susceptible to PVYO [177]. Similarly, eIF4E1 editing in the tomato cv. Micro-Tom confers resistance to pepper mottle virus (PepMoV) but not to tobacco etch virus (TEV) [178]. These two above-mentioned studies in tomato demonstrated that CRISPR-mediated inactivation of eIF4E1 resulted in a narrow resistance spectrum against potyviruses [177,178]. In addition to eIF4E1, eIF4E2 mutants were developed in cherry tomato, showing complete resistance to one PVMV isolate, partial resistance to another, and full susceptibility to both tested Potato virus Y (PVY) isolates [179]. Editing cassava nCBP1 and nCBP2 (alternative eIFE-like proteins) was shown to reduce the severity and incidence of cassava brown streak (CBS) disease symptoms [180]. CRISPR/Cas9 was employed to mutate the eIF4E in the elite tetraploid potato cultivar ‘Kruda’, resulting in enhanced resistance to PVY. The edited lines exhibited broad-spectrum resistance [181]. Resistance has been developed in melon against Moroccan watermelon mosaic virus (MWMV) by targeting the gene encoding the cap-binding protein eIF4E [182,183]. CRISPR-mediated mutations created in eIF4E and cucumber plants resistant to watermelon mosaic virus (WMV), papaya ringspot virus (PRSV), and zucchini yellow mosaic virus (ZYMV) have been developed [184]. In another study, KOof CleIF4E1 in watermelon provided resistance to ZYMV, however, the mutants showed developmental defects, including altered plant growth, abnormal leaf morphology, and reduced yield [185].

7.2. Targeting Mlo Genes

The Mlo gene family encodes membrane-associated proteins that act as negative regulators of plant immunity, making them ideal targets for S-gene editing. Loss-of-function mutations in Mlo genes confer durable and broad-spectrum resistance to PM, a widespread fungal disease that severely affects many crops [11,186]. Although natural resistance to PM is typically polygenic, modern gene-editing tools like CRISPR/Cas9 enable the precise editing of Mlo genes, offering a targeted approach for developing resistant cultivars. Tek et al. used CRISPR/Cas9 to investigate the function of genes responsible for mlo resistance to PM [187]. CRISPR/Cas9 GE of the Mlo gene has been successfully implemented in wheat, barley, grapevine, cucumber, tomato, and soybean [22,188,189,190,191,192,193]. In 2014, Wang et al. used the CRISPR/Cas9 system to target the TaMLOA1 allele in the wheat cultivar Kenong 199. The mutants exhibited robust and broad-spectrum resistance to PM along with undesirable pleiotropic effects [22]. Similarly, Nekrasov et al. targeted the SlMlo1 loci and developed a variety named “Tomelo” (genome-edited tomato), which conferred resistance to the PM pathogen Oidium neolycopersici [191]. In addition, two susceptibility genes SlPelo and SlMlo1 have been targeted in tomato (BN-86 cultivar) using multiple sgRNAs and KOsgenerated for SlMlo1 showed complete resistance to PM [192]. In 2022, a wheat variety, Tamlo-R32, having targeted deletion (304 Kb) in the MLO-B1 locus, has been developed using CRISPR, which confers strong resistance to PM without compromising plant growth or yield [188]. In grapevine, four VvMLO3-edited lines generated using CRISPR/Cas9 exhibited enhanced resistance to PM along with cell wall appositions in edited lines compared to WT lines [189]. In another study, CRISPR/Cas9 was used to develop PM-resistant cucumber by targeting CsaMLO1, CsaMLO8, and CsaMLO11 genes. Mutation in CsaMLO8 and CsaMLO1/CsaMLO11 corresponded to pre-invasion and post-invasion defense responses (HRs), respectively [187]. Recently, Shnaider et al. (2023) targeted CsaMLO8 using CRISPR technology to develop PM resistance in the susceptible cucumber cultivar Ilan [190]. In another study, researchers used a dual gRNA CRISPR construct to simultaneously KO four MLO homologs (GmMLO02, GmMLO19, GmMLO20, and GmMLO23) in the Vietnamese soybean elite cultivar DT26, which conferred increased resistance to PM [193].

7.3. Targeting SWEET Family Genes

Targeting SWEET genes has proven effective in controlling bacterial infections in crops, particularly bacterial blight in rice [194]. Several studies have reported CRISPR/Cas9-mediated editing of the OsSWEET gene promoters to enhance resistance against Xanthomonas oryzae pv. oryzae (Xoo), the causative agent of bacterial blight [195,196]. In a notable study, Zafar et al. (2020) edited the effector binding elements (EBEs) in the OsSWEET14 promoter that are recognized by three TALEs (AvrXa7, PthXo3, and TalF). This promoter editing was performed in four elite super basmati rice lines, resulting in enhanced resistance to Xoo [197]. In another study, CRISPR mediated KOof OsSWEET14 in rice cultivar Zhonghua 11 conferred strong resistance to both African and Asian Xoo strains [198]. CRISPR/Cas9-mediated editing of the OsSWEET11 promoter in Mentik Wangi rice (a local variety of central Java) conferred bacterial blight resistance without altering key agronomic traits [199]. Recently, cytidine and adenine base editors based on CRISPR/Cas9 were employed to modify the TalC effector-binding element in the OsSWEET14 promoter of rice cultivars IR24, Kitaake, and Zhonghua 11. C-to-T edits disrupted OsSWEET14 induction, conferring resistance to African Xoo strains, while A-to-G edits maintained susceptibility without any off-targets effects [200].

7.4. Targeting Other S-Genes

In tomatoes, StDMR6-1 ortholog has been mutated using CRISPR/Cas9 and mutated lines showed increased resistance to P. capsici and P. syringae pv. tomato, indicating the broad-spectrum disease resistance role of DMR6-1 [201]. CRISPR/Cas9-mediated multiplex GE of the BnWRKY70 gene in B. napus resulted in mutants with enhanced resistance to Sclerotinia sclerotiorum [33]. In Arabidopsis, mutants of enhanced disease resistance 1 (EDR1) gene provide resistance to Erysiphe cichoracearum, a causal agent of PM [202]. Therefore, EDR1 serves as a promising target S-gene for enhancing resistance to PM. Using CRISPR/Cas9 to simultaneously edit all homologs of wheat, TaEDR1 produced mutant plants resistant to Blumeria graminis f. sp. tritici (Bgt). Interestingly, these edited plants showed no adverse effects on growth or development [203]. In apple, the MdDIPM4 gene, a member of the DIPM family associated with susceptibility to fire blight, was targeted using CRISPR technology, resulting in edited cultivars with enhanced resistance to the disease [204]. In addition to MLO, powdery mildew resistance 4 (AtPMR4) has been identified as another S-gene in Arabidopsis. Recently, Martinez et al. used CRISPR/Cas9 to edit the SlPMR4 gene (AtPMR4 ortholog) in tomato. The edited T2 mutant plants exhibited partial resistance to Oidium neolycopersici with growth penalties [205]. In potato, CRISPR/Cas9 was used to generate functional KOs of StDND1, StCHL1, and StDMR6-1, resulting in increased resistance to late blight. Among these, StDMR6-1 and StCHL1 mutants showed no adverse growth effects, making them promising candidates for disease-resistant crop development [206]. In addition, simultaneous silencing of miR482b and miR482c provided enhanced resistance to P. infestans [207]. In sweet basil, Zhang et al. (2021) used CRISPR/Cas9 to edit the orthologous ObHSK gene, generating transgene-free plants. The edited lines exhibited normal growth under greenhouse conditions and showed enhanced resistance to Peronospora belbahrii compared to WT [208]. Cha et al. recently targeted the nucleoredoxin (SlNRX1) gene in tomato through CRISPR/Cas9 technology, and the mutants displayed enhanced resistance against the fungal pathogen Alternaria brassicicola. [209]. Similarly, multiplex CRISPR/Cas9 editing of miR482b and miR482c (acting as susceptibility factors) in tomato exhibited reduced disease symptoms and increased yield [210,211,212]. A new susceptibility gene, StPM1, was identified in Solanum tuberosum by Bi et al. (2023), and CRISPR/Cas9-mediated KOof StPM1 conferred resistance to Phytophthora without impacting plant growth or development [213]. CRISPR/Cas9-mediated KOof the rice S-gene OsHPP04 enhanced resistance to Meloidogyne graminicola without affecting plant growth. The transgene-free mutants showed stronger immune responses, showing the feasibility of S-gene editing for developing nematode-resistant crops [214]. A summary of successful editing events targeting S-genes in various crops is provided in Table 2.

8. Global Policies and Regulatory Approaches for GE Development and Commercialization

GE stands out as the breakthrough technology of the decade in agriculture for crop improvement. Unlike traditional genetically modified organisms (GMOs), genome-edited crops may not contain foreign DNA, blurring the lines for regulatory frameworks. As technology use expands, so does the need for robust governance to ensure biosafety, transparency, and equitable benefit sharing. In many countries, GMOs and gene-edited crops are often governed by the same sets of rules and regulations, despite their fundamental differences [239]. Globally, regulatory frameworks for GMOs generally follow two primary approaches, product-based regulations (focus is on the safety and characteristics of the final product, regardless of how it was developed) and process-based regulations (focusing on the techniques used to create GMOs, especially when introducing new traits). Differences in these regulations partly reflect varying national priorities and the fact that some countries have not signed the Cartagena Protocol on Biosafety (CPB). For instance, the EU’s definition of GMOs as “not naturally altered” impacted the public perception as a whole towards GE technologies [240]. In 2018, the European Court of Justice ruled that genome-edited organisms are subject to the GMO Directive, Callaway, 2018 (Confédération paysanne and others v. Premier ministre and Ministre de l’Agriculture de l’Agroalimentaire et de la Forêt, 2018) [241]. The European Union follows a process-based approach, classifying most GE crops as GMOs. This results in extensive labeling and traceability requirements, effectively silently negating rapid adoption, despite ongoing debates about revising the framework for new genomic techniques (NGTs). Likewise, Malaysia and several African nations have strict GMO style regulations, resulting in delayed approvals for GE crops [242].
In contrast to the EU, the U.S. has adopted a product-based regulatory approach. The USDA does not subject certain genome-edited crops (SDN 1, SDN 2) to the same scrutiny as GMOs, provided they do not contain foreign DNA and could have been developed through conventional breeding [242]. In December 2024, a federal judge overturned the SECURE Rule, restoring pre-2020 regulations. While previously approved crops remain unaffected, the ruling adds complexity and cost to new products, posing challenges for smaller developers. A CRISPR/Cas9-edited white button mushroom resistant to browning was the first genome-edited crop to be exempt from USDA regulations [243]. The USDA has consistently supported research on CRISPR/Cas9 technology across various crops [244,245]. The US adopts a product-based approach, where crops with minor edits (SDN 1, SDN 2) are exempt from GMO regulations, enabling faster approvals and lower costs.
Similar regulation to the USA has been implemented by Canada and four South American countries that classified genome-edited crops (SDN-1, -2) as equivalent to conventional breeds [242]. Similarly, India’s regulatory framework for genome-edited crops was updated in 2022, exempting plants without foreign gene insertions from being classified as transgenic provided they undergo a safety assessment (Ministry of Environment, Forest and Climate Change MoEFCC, Govt. of India, 2022). India exempts genome-edited plants developed through SDN 1 and SDN 2 techniques (Guidelines for the Safety Assessment of Genome Edited Plants, 2022). In May 2025, India approved its first genome-edited rice varieties, DRR Dhan 100 (Kamala) and Pusa DST Rice 1. In Australia, the 2019 amendments specify that organisms modified using SDN 1 technique are generally exempt from regulation under the Act, as such changes are considered comparable to those occurring naturally or through conventional breeding. The African Union’s Agenda 2063 emphasizes the use of GE to enhance agricultural productivity and crop resilience. So far, two countries (Nigeria and Kenya) have implemented regulations for a case-by-case review of genome-edited crops [246] (Figure 5).
Countries like Argentina, Brazil, and Japan, have adopted more flexible regulatory frameworks that distinguish GE crops from traditional GMOs where no foreign genes are introduced [234,235]. In 2022, the Ministry of Agriculture and Rural Affairs in China released safety guidelines for the evaluation of genome-edited plants for agriculture. Recently, in May 2024, the Chinese government approved gene-edited wheat for the first time, marking a significant step toward the adoption of genome-editing food crops. In summer 2024, the Philippines’ Department of Agriculture and Bureau of Plant Industry classified genome-edited bananas as non-GMO, approving them for import and cultivation (www.isaaa.org). However, such varying national approaches pose challenges for international trade and innovation. Generally, crops produced by CRISPR/Cas9 and other gene editing methods are challenging traditional definitions of GMOs and pushing regulatory bodies to reconsider existing frameworks. Regulatory bodies worldwide are still adjusting to the fast-paced advancements in this technology. Therefore, despite existing legal hurdles, researchers, investors, and consumers should continue to support and prioritize the development and study of improved and beneficial crops. NBTs have the potential to create more resilient, nutritious, and high-yielding plants that can meet the growing global food demand and address challenges like climate change and disease pressures.

9. Challenges and Future Perspectives

While targeting S-genes via GE has proven to be a promising strategy for enhancing disease resistance, there are several challenges that hinder its universal application. First, many S-genes play dual roles in plant physiology, and their inactivation may lead to unintended pleiotropic effects. For example, the eIF4E gene, essential for potyvirus replication, is also involved in basic cellular processes, and its modification can result in growth penalties. Another challenge is the potential for resistance breakdown due to the adaptive evolution of pathogens. For instance, potyviruses have been shown to evolve new VPg variants capable of overcoming eIF4E-based resistance. Moreover, while S-gene resistance tends to be durable and broad-spectrum, its recessive nature complicates its integration into elite lines through conventional breeding. Another major challenge resides in the widespread public opinion that fails to distinguish between genetically modified organisms (GMOs) and genome-edited crops, often viewing both as equally harmful to the environment and human health. This misconception is compounded by the lack of a unified global regulatory framework that clearly differentiates genome-edited crops from GMOs and facilitates their production and commercialization [247]. Regulatory frameworks also remain inconsistent globally, for example, the European Union and New Zealand currently regulates gene-edited crops under strict GMO legislation, in contrast to product-based approaches in North and South America (such as the USA, Colombia, Canada, and Argentina), along with several in Asia (including India, China, and Japan), and Australia [248]. This disparity creates obstacles for global commercialization and international trade.
Overall, the manipulation of plant S-genes presents a promising avenue for enhancing disease resistance in crops, but it also carries the risk of negatively impacting vital plant processes due to the native functions these genes often perform. Many S-genes are integral to essential physiological functions like sugar allocation, mediated by SWEET transporters, or the regulation of defense signaling by various kinases. Therefore, altering these genes can lead to undesirable trade-offs, including reduced yield, impaired growth, or decreased tolerance to environmental stresses. To circumvent trade-offs it is important to understand the roles of S-genes in physiology and development [59]. Looking ahead, several emerging innovations offer a pathway to overcome these hurdles and further harness the potential of S-gene editing. Recent advancements in GE technologies such as base editing and prime editing provide powerful tools to precisely modify S-genes, minimizing unintended negative consequences on plant fitness [59,249]. These technologies allow researchers to make targeted genetic alterations that can confer disease resistance while preserving the essential native functions of the genes. Strategies such as cis-regulatory editing, modifying promoter regions instead of coding sequences, offer a way to attenuate gene expression rather than abolish function entirely [250]. The integration of multi-omics approaches, such as transcriptomics, proteomics, and metabolomics, with machine learning techniques is significantly accelerating the identification and prioritization of candidate S-genes. These advanced computational methods can analyze vast datasets to uncover complex gene regulatory networks and predict the functional impact of S-gene modifications. This allows researchers to more accurately identify S-genes whose perturbation would lead to beneficial resistance traits while minimizing undesirable trade-offs. By leveraging these integrated approaches, plant breeders can make more informed decisions, leading to the development of more resilient and sustainable crops.
Additionally, multiplex CRISPR platforms are enabling the simultaneous editing of several susceptibility loci, which can significantly enhance resistance durability. Finally, harmonization of international regulatory policies and increased public engagement are essential to reduce barriers to market entry and foster societal trust in genome-edited crops. Lastly, the combination of S-gene editing with sustainable agricultural practices, can provide a holistic strategy to achieve resilient, productive, and environmentally friendly food systems in the face of climate change and increasing disease pressures.

Author Contributions

Conceptualization, S.T. and G.M.; literature search and analysis, S.T. and G.M. original draft preparation, S.T., S.K., S.A., P.S., Y.F., and G.M.; writing—review and editing, S.T., S.K., S.A., P.S., Y.F., and G.M., supervision, G.M.; project administration, S.T. All authors have read and agreed to the published version of the manuscript.

Funding

The authors acknowledge financial support from the U.S. Department of Agriculture and the Florida Department of Agriculture and Consumer Services (FDACS) through contract grant number 29295.

Data Availability Statement

No new data were generated or analyzed in this study. All data referenced are publicly available in the cited literature.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Savary, S.; Willocquet, L.; Pethybridge, S.J.; Esker, P.; McRoberts, N.; Nelson, A. The global burden of pathogens and pests on major food crops. Nat. Ecol. Evol. 2019, 3, 430–439. [Google Scholar] [CrossRef]
  2. FAO. How to Feed the World in 2050; Food and Agriculture Organization: Rome, Italy, 2009. [Google Scholar]
  3. Zayan, S.A. Impact of climate change on plant diseases and IPM strategies. In Plant Diseases—Current Threats and Management Trends; IntechOpen: London, UK, 2019. [Google Scholar]
  4. Fahad, S.; Bajwa, A.A.; Nazir, U.; Anjum, S.A.; Farooq, A.; Zohaib, A. Crop production under drought and heat stress: Plant responses and management options. Front. Plant Sci. 2017, 8, 1147. [Google Scholar] [CrossRef]
  5. Dodds, P.N.; Rathjen, J.P. Plant immunity: Towards an integrated view of plant–pathogen interactions. Nat. Rev. Genet. 2010, 11, 539–548. [Google Scholar] [CrossRef]
  6. Couto, D.; Zipfel, C. Regulation of pattern recognition receptor signalling in plants. Nat. Rev. Immunol. 2016, 16, 537–552. [Google Scholar] [CrossRef]
  7. Jones, N.T.; Gilbert, B. Biotic forcing: The push–pull of plant ranges. Plant Ecol. 2016, 217, 1331–1344. [Google Scholar] [CrossRef]
  8. Bailey-Serres, J.; Parker, J.E.; Ainsworth, E.A.; Oldroyd, G.E.; Schroeder, J.I. Genetic strategies for improving crop yields. Nature 2019, 575, 109–118. [Google Scholar] [CrossRef] [PubMed]
  9. Kourelis, J.; Van der Hoorn, R.A.L. Defended to the nines: 25 years of resistance gene cloning identifies nine mechanisms for R protein function. Plant Cell 2018, 30, 285–299. [Google Scholar] [CrossRef]
  10. Dangl, J.L.; Horvath, D.M.; Staskawicz, B.J. Pivoting the plant immune system from dissection to deployment. Science 2013, 341, 746–751. [Google Scholar] [CrossRef] [PubMed]
  11. van Schie, C.N.; Takken, F.L.W. Susceptibility genes 101: How to be a good host. Annu. Rev. Phytopathol. 2014, 52, 551–581. [Google Scholar] [CrossRef] [PubMed]
  12. Zaidi, S.S.E.A.; Mukhtar, M.S.; Mansoor, S. Genome editing: Targeting susceptibility genes for plant disease resistance. Trends Biotechnol. 2018, 36, 898–906. [Google Scholar] [CrossRef]
  13. Vogel, J.P.; Raab, T.K.; Schiff, C.; Somerville, S.C. PMR6, a pectate lyase-like gene required for powdery mildew susceptibility in Arabidopsis. Plant Cell 2002, 14, 2095–2106. [Google Scholar] [CrossRef]
  14. Eckardt, N.A. Plant disease susceptibility genes? Plant Cell 2002, 14, 1983–1986. [Google Scholar] [CrossRef] [PubMed]
  15. Moniruzzaman, M.; Zhong, Y.; Yan, H.; Yuanda, L.; Jiang, B.; Zhong, G. Exploration of susceptible genes with clustered regularly interspaced short palindromic repeats–tissue-specific knockout (CRISPR-TSKO) to enhance host resistance. Crit. Rev. Plant Sci. 2020, 39, 387–417. [Google Scholar] [CrossRef]
  16. Das, A.; Sharma, N.; Prasad, M. CRISPR/Cas9: A novel weapon in the arsenal to combat plant diseases. Front. Plant Sci. 2019, 9, 2008. [Google Scholar] [CrossRef] [PubMed]
  17. Freisleben, R.; Lein, A. Über die Auffindung einer mehltauresistenten Mutante nach Röntgenbestrahlung einer anfälligen reinen Linie von Sommergerste. Naturwissenschaften 1942, 30, 608. [Google Scholar] [CrossRef]
  18. Kusch, S.; Panstruga, R. mlo-based resistance: An apparently universal “weapon” to defeat powdery mildew disease. Mol. Plant Microbe Interact. 2017, 30, 179–189. [Google Scholar] [CrossRef]
  19. Humphry, M.; Reinstädler, A.; Ivanov, S.; Bisseling, T.; Panstruga, R. Durable broad-spectrum powdery mildew resistance in pea er1 plants is conferred by natural loss-of-function mutations in PsMLO1. Mol. Plant Pathol. 2011, 12, 866–878. [Google Scholar] [CrossRef]
  20. Pavan, S.; Schiavulli, A.; Appiano, M.; Marcotrigiano, A.R.; Cillo, F.; Visser, R.G.; Bai, Y.; Lotti, C.; Ricciardi, L. Pea powdery mildew er1 resistance is associated with loss-of-function mutations at a MLO homologous locus. Theor. Appl. Genet. 2011, 123, 1425–1431. [Google Scholar] [CrossRef]
  21. Zheng, Z.; Nonomura, T.; Appiano, M.; Pavan, S.; Matsuda, Y.; Toyoda, H.; Wolters, A.M.A.; Visser, R.G.; Bai, Y. Loss of function in Mlo orthologs reduces susceptibility of pepper and tomato to powdery mildew disease caused by Leveillula taurica. PLoS ONE 2013, 8, e70723. [Google Scholar] [CrossRef]
  22. Wang, Y.; Cheng, X.; Shan, Q.; Zhang, Y.; Liu, J.; Gao, C.; Qiu, J.L. Simultaneous editing of three homoeoalleles in hexaploid bread wheat confers heritable resistance to powdery mildew. Nat. Biotechnol. 2014, 32, 947–951. [Google Scholar] [CrossRef]
  23. Pessina, S.; Angeli, D.; Martens, S.; Visser, R.G.F.; Bai, Y.; Salamini, F.; Velasco, R.; Schouten, H.J.; Malnoy, M. The knock-down of the expression of MdMLO19 reduces susceptibility to powdery mildew (Podosphaera leucotricha) in apple (Malus domestica). Plant Biotechnol. J. 2016, 14, 2033–2044. [Google Scholar] [CrossRef]
  24. Acevedo-Garcia, J.; Spencer, D.; Thieron, H.; Reinstädler, A.; Hammond-Kosack, K.; Phillips, A.L.; Panstruga, R. mlo-based powdery mildew resistance in hexaploid bread wheat generated by a non-transgenic TILLING approach. Plant Biotechnol. J. 2017, 15, 367–378. [Google Scholar] [CrossRef]
  25. Consonni, C.; Humphry, M.E.; Hartmann, H.A.; Livaja, M.; Durner, J.; Westphal, L.; Vogel, J.P.; Lipka, V.; Kemmerling, B.; Schulze-Lefert, P. Conserved requirement for a plant host cell protein in powdery mildew pathogenesis. Nat. Genet. 2006, 38, 716–720. [Google Scholar] [CrossRef]
  26. Berg, J.A.; Appiano, M.; Santillán Martínez, M.; Hermans, F.W.K.; Vriezen, W.H.; Visser, R.G.F.; Bai, Y.; Schouten, H.J. A transposable element insertion in the susceptibility gene CsaMLO8 results in hypocotyl resistance to powdery mildew in cucumber. BMC Plant Biol. 2015, 15, 243. [Google Scholar] [CrossRef]
  27. Iovieno, P.; Andolfo, G.; Schiavulli, A.; Catalano, D.; Ricciardi, L.; Frusciante, L.; Pavan, S. Structure, evolution and functional inference on the MLO gene family in three cultivated Cucurbitaceae spp. BMC Genom. 2015, 16, 1112. [Google Scholar] [CrossRef] [PubMed]
  28. Porterfield, R.; Meru, G. Candidate Susceptibility Genes for Powdery and Downy Mildew in Watermelon and Squash. J. Phylogenet. Evol. Biol. 2017, 5, 186. [Google Scholar] [CrossRef]
  29. Sonenberg, N.; Morgan, M.A.; Merrick, W.C.; Shatkin, A.J. A polypeptide in eukaryotic initiation factors that crosslinks specifically to the 5’-terminal cap in mRNA. Proc. Natl. Acad. Sci. USA 1978, 75, 4843–4847. [Google Scholar] [CrossRef] [PubMed]
  30. Wang, A.; Krishnaswamy, S. Eukaryotic translation initiation factor 4E-mediated recessive resistance to plant viruses and its utility in crop improvement. Mol. Plant Pathol. 2012, 13, 795–803. [Google Scholar] [CrossRef] [PubMed]
  31. Charron, C.; Nicolaï, M.; Gallois, J.L.; Robaglia, C.; Moury, B.; Palloix, A.; Caranta, C. Natural variation and functional analyses provide evidence for co-evolution between plant eIF4E and potyviral VPg. Plant J. 2008, 54, 56–68. [Google Scholar] [CrossRef]
  32. Sun, J.; Li, N.; Oh, K.S.; Dutta, B.; Vayttaden, S.J.; Lin, B.; Fraser, I.D. Comprehensive RNAi-based screening of human and mouse TLR pathways identifies species-specific preferences in signaling protein use. Sci. Signal. 2016, 9, ra3. [Google Scholar] [CrossRef]
  33. Sun, Q.; Lin, L.; Liu, D.; Wu, D.; Fang, Y.; Wu, J.; Wang, Y. CRISPR/Cas9-mediated multiplex genome editing of the BnWRKY11 and BnWRKY70 genes in Brassica napus L. Int. J. Mol. Sci. 2018, 19, 2716. [Google Scholar] [CrossRef]
  34. Li, T.; Liu, B.; Spalding, M.H.; Weeks, D.P.; Yang, B. High-efficiency TALEN-based gene editing produces disease-resistant rice. Nat. Biotechnol. 2012, 30, 390–392. [Google Scholar] [CrossRef] [PubMed]
  35. 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]
  36. Tyagi, S.; Kumar, R.; Kumar, V.; Won, S.Y.; Shukla, P. Engineering disease resistant plants through CRISPR-Cas9 technology. GM Crops Food 2021, 12, 125–144. [Google Scholar] [CrossRef] [PubMed]
  37. Jones, J.D.; Dangl, J.L. The plant immune system. Nature 2006, 444, 323–329. [Google Scholar] [CrossRef]
  38. Boller, T.; He, S.Y. Innate immunity in plants: An arms race between pattern recognition receptors in plants and effectors in microbial pathogens. Science 2009, 324, 742–744. [Google Scholar] [CrossRef]
  39. Tanaka, K.; Heil, M. Damage-associated molecular patterns (DAMPs) in plant innate immunity: Applying the danger model and evolutionary perspectives. Annu. Rev. Phytopathol. 2021, 59, 53–75. [Google Scholar] [CrossRef]
  40. Yuan, M.; Jiang, Z.; Bi, G.; Nomura, K.; Liu, M. Pattern-recognition receptors are required for NLR-mediated plant immunity. Nature 2021, 592, 105–109. [Google Scholar] [CrossRef] [PubMed]
  41. Speth, E.B.; Lee, Y.N.; He, S.Y. Pathogen virulence factors as molecular probes of basic plant cellular functions. Curr. Opin. Plant Biol. 2007, 10, 580–586. [Google Scholar] [CrossRef]
  42. Nuernberger, T.; Kemmerling, B. PAMP-triggered basal immunity in plants. Adv. Bot. Res. 2009, 51, 823937. [Google Scholar]
  43. Bentham, A.R.; De la Concepcion, J.C.; Mukhi, N.; Zdrzałek, R.R.; Draeger, M.; Gorenkin, D.; Hughes, R.K.; Banfield, M.J. A molecular roadmap to the plant immune system. J. Biol. Chem. 2020, 295, 14916–14935. [Google Scholar] [CrossRef]
  44. Xie, S.S.; Duan, C.G. Epigenetic regulation of plant immunity: From chromatin codes to plant disease resistance. aBIOTECH 2023, 4, 124–139. [Google Scholar] [CrossRef]
  45. Kourelis, J.; Adachi, H. Activation and regulation of NLR immune receptor networks. Plant Cell Physiol. 2022, 63, 1366–1377. [Google Scholar] [CrossRef]
  46. Chiang, Y.; Coaker, G. Effector triggered immunity: NLR immune perception and downstream defense responses. Arab. Book 2015, 13, e0183. [Google Scholar] [CrossRef]
  47. Deng, Y.; Ning, Y.; Yang, D.L.; Zhai, K.; Wang, G.L.; He, Z. Molecular basis of disease resistance and perspectives on breeding strategies for resistance improvement in crops. Mol. Plant 2020, 13, 1402–1419. [Google Scholar] [CrossRef]
  48. Dangl, J.L.; Jones, J.D. Plant pathogens and integrated defence responses to infection. Nature 2001, 411, 826–833. [Google Scholar] [CrossRef]
  49. Langner, T.; Kamoun, S.; Belhaj, K. CRISPR Crops: Plant genome editing toward disease resistance. Annu. Rev. Phytopathol. 2018, 56, 479–512. [Google Scholar] [CrossRef] [PubMed]
  50. Sánchez-Vallet, A.; Fouché, S.; Fudal, I.; Hartmann, F.E.; Soyer, J.L.; Tellier, A.; Croll, D. The genome biology of effector gene evolution in filamentous plant pathogens. Annu. Rev. Phytopathol. 2018, 56, 21–40. [Google Scholar] [CrossRef] [PubMed]
  51. Dong, O.X.; Ronald, P.C. Genetic engineering for disease resistance in plants: Recent progress and future perspectives. Plant Physiol. 2019, 180, 26–38. [Google Scholar] [CrossRef] [PubMed]
  52. Amezrou, R.; Audéon, C.; Compain, J.; Gélisse, S.; Ducasse, A.; Saintenac, C.; Lapalu, N. A secreted protease-like protein in Zymoseptoria tritici is responsible for avirulence on Stb9 resistance gene in wheat. PLoS Pathog. 2023, 19, e1011376. [Google Scholar] [CrossRef]
  53. Veillet, F.; Durand, M.; Kroj, T.; Cesari, S.; Gallois, J.L. Precision breeding made real with CRISPR: Illustration through genetic resistance to pathogens. Plant Commun. 2020, 1, 100027. [Google Scholar] [CrossRef]
  54. van Wersch, S.; Tian, L.; Hoy, R.; Li, X. Plant NLRs: The whistleblowers of plant immunity. Plant Commun. 2020, 1, 100016. [Google Scholar] [CrossRef]
  55. Giannakopoulou, A.; Steele, J.F.; Segretin, M.E.; Bozkurt, T.O.; Zhou, J.; Robatzek, S. Tomato I2 immune receptor can be engineered to confer partial resistance to the oomycete Phytophthora infestans in addition to the fungus Fusarium oxysporum. Mol. Plant Microbe Interact. 2015, 28, 1316–1329. [Google Scholar] [CrossRef]
  56. Andolfo, G.; Iovieno, P.; Frusciante, L.; Ercolano, M.R. Genome-editing technologies for enhancing plant disease resistance. Front. Plant Sci. 2016, 7, 1813. [Google Scholar] [CrossRef]
  57. Jacob, P.; Avni, A.; Bendahmane, A. Translational research: Exploring and creating genetic diversity. Trends Plant Sci. 2018, 23, 42–52. [Google Scholar] [CrossRef] [PubMed]
  58. Kapos, P.; Devendrakumar, K.T.; Li, X. Plant NLRs: From discovery to application. Plant Sci. 2019, 279, 3–18. [Google Scholar] [CrossRef] [PubMed]
  59. Garcia-Ruiz, H.; Szurek, B.; Van den Ackerveken, G. Stop helping pathogens: Engineering plant susceptibility genes for durable resistance. Curr. Opin. Biotechnol. 2021, 70, 187–195. [Google Scholar] [CrossRef] [PubMed]
  60. Gaj, T.; Sirk, S.J.; Shui, S.L.; Liu, J. Genome-editing technologies: Principles and applications. Cold Spring Harb. Perspect. Biol. 2016, 8, a023754. [Google Scholar] [CrossRef]
  61. Sufyan, M.; Daraz, U.; Hyder, S.; Zulfiqar, U.; Iqbal, R.; Eldin, S.M.; Rafiq, F. An overview of genome engineering in plants, including its scope, technologies, progress and grand challenges. Funct. Integr. Genom. 2023, 23, 119. [Google Scholar] [CrossRef]
  62. Choudry, M.W.; Riaz, R.; Nawaz, P.; Ashraf, M.; Ijaz, B.; Bakhsh, A. CRISPR-Cas9 mediated understanding of plants’ abiotic stress-responsive genes to combat changing climatic patterns. Funct. Integr. Genom. 2024, 24, 132. [Google Scholar] [CrossRef]
  63. Pavan, S.; Jacobsen, E.; Visser, R.G.; Bai, Y. Loss of susceptibility as a novel breeding strategy for durable and broad-spectrum resistance. Mol. Breed. 2010, 25, 1–12. [Google Scholar] [CrossRef]
  64. Engelhardt, S.; Stam, R.; Hückelhoven, R. Good riddance? Breaking disease susceptibility in the era of new breeding technologies. Agronomy 2018, 8, 114. [Google Scholar] [CrossRef]
  65. Lou, X.-Y.; Ma, J.Z.; Yang, M.C.K.; Zhu, J.; Liu, P.-Y.; Deng, H.-W.; Elston, R.C.; Li, M.D. Improvement of mapping accuracy by unifying linkage and association analysis. Genetics 2006, 172, 647–661. [Google Scholar] [CrossRef] [PubMed]
  66. Zhang, R.; Chen, S.; Meng, X.; Chai, Z.; Wang, D.; Yuan, Y.; Chen, K.; Jiang, L.; Li, J.; Gao, C. Generating broad-spectrum tolerance to ALS-inhibiting herbicides in rice by base editing. Sci. China Life Sci. 2021, 64, 1624–1633. [Google Scholar] [CrossRef] [PubMed]
  67. Henning, J.A.; Townsend, M.S.; Gent, D.H. QTL mapping of powdery mildew susceptibility in hop (Humulus lupulus L.). Euphytica 2011, 180, 411–420. [Google Scholar] [CrossRef]
  68. Santos, C.; Polanco, C.; Rubiales, D.; Vaz Patto, M.C. The MLO1 powdery mildew susceptibility gene in Lathyrus species: The power of high-density linkage maps in comparative mapping and synteny analysis. Plant Genome 2021, 14, e20090. [Google Scholar] [CrossRef]
  69. Alseekh, S.; Kostova, D.; Bulut, M.; Fernie, A.R. Genome-wide association studies: Assessing trait characteristics in model and crop plants. Cell. Mol. Life Sci. 2021, 78, 5743–5754. [Google Scholar] [CrossRef]
  70. Tibbs Cortes, L.; Zhang, Z.; Yu, J. Status and prospects of genome-wide association studies in plants. Plant Genome 2021, 14, e20077. [Google Scholar] [CrossRef]
  71. van de Bunt, M.; Cortes, A.; IGAS Consortium; Brown, M.A.; Morris, A.P.; McCarthy, M.I. Evaluating the performance of fine-mapping strategies at common variant GWAS loci. PLoS Genet. 2015, 11, e1005535. [Google Scholar] [CrossRef]
  72. Deng, X.; Huang, D.; Wang, Y.; An, H.; Bai, D.; Wang, X.; Niu, S.; Song, X. Genome-wide association study of salicylic acid provides genetic insights for tea plant selective breeding. Hortic. Res. 2025, 12, uhae362. [Google Scholar] [CrossRef]
  73. Chittem, K.; Yajima, W.R.; Goswami, R.S.; del Río Mendoza, L.E. Transcriptome analysis of the plant pathogen Sclerotinia sclerotiorum interaction with resistant and susceptible canola (Brassica napus) lines. PLoS ONE 2020, 15, e0229844. [Google Scholar] [CrossRef]
  74. Cao, Y.; Zhou, H.; Zhou, X.; Li, F. Control of plant viruses by CRISPR/Cas system-mediated adaptive immunity. Front. Microbiol. 2020, 11, 593700. [Google Scholar] [CrossRef]
  75. Ju, X.; Li, F.; Li, J.; Wu, C.; Xiang, G.; Zhao, X.; Nan, Y.; Zhao, D.; Ding, Q. Genome-wide transcriptomic analysis of highly virulent African swine fever virus infection reveals complex and unique virus host interaction. Vet. Microbiol. 2021, 261, 109211. [Google Scholar] [CrossRef]
  76. Meng, H.; Sun, M.; Jiang, Z.; Liu, Y.; Sun, Y.; Liu, D.; Jiang, C. Comparative transcriptome analysis reveals resistant and susceptible genes in tobacco cultivars in response to infection by Phytophthora nicotianae. Sci. Rep. 2021, 11, 809. [Google Scholar] [CrossRef]
  77. Schouten, H.J.; Krauskopf, J.; Visser, R.G.F. Identification of candidate genes required for susceptibility to powdery or downy mildew in cucumber. Euphytica 2014, 200, 475–486. [Google Scholar] [CrossRef]
  78. Tian, D.; Wang, P.; Tang, B.; Teng, X.; Li, C.; Liu, X.; Zou, D.; Song, S.; Zhang, Z. GWAS Atlas: A curated resource of genome-wide variant-trait associations in plants and animals. Nucleic Acids Res. 2020, 48, D927–D932. [Google Scholar] [CrossRef] [PubMed]
  79. Zahid, G.; Kaçar, Y.A.; Dönmez, D.; Küden, A.; Giordani, T. Perspectives and recent progress of genome-wide association studies (GWAS) in fruits. Mol. Biol. Rep. 2022, 49, 5341–5352. [Google Scholar] [CrossRef] [PubMed]
  80. Gupta, P.K. GWAS for genetics of complex quantitative traits: Genome to pangenome and SNPs to SVs and k-mers. BioEssays 2021, 43, 2100109. [Google Scholar] [CrossRef]
  81. Morales, J.; Pujar, S.; Loveland, J.E.; Astashyn, A.; Bennett, R.; Berry, A.; Cox, E. A joint NCBI and EMBL-EBI transcript set for clinical genomics and research. Nature 2022, 604, 310–315. [Google Scholar] [CrossRef] [PubMed]
  82. Liyanage, K.K.; Khan, S.; Herath, V.; Brooks, S.; Mortimer, P.E.; Nadir, S.; Hyde, K.D.; Xu, J. Genome wide identification of the MLO gene family associated with powdery mildew resistance in rubber trees (Hevea brasiliensis). Trop. Plant Biol. 2020, 13, 331–342. [Google Scholar] [CrossRef]
  83. Pessina, S.; Pavan, S.; Catalano, D.; Gallotta, A.; Visser, R.G.F.; Bai, Y.; Malnoy, M. Characterization of the MLO gene family in Rosaceae and gene expression analysis in Malus domestica. BMC Genom. 2014, 15, 618. [Google Scholar] [CrossRef]
  84. Calle Garcia, J.; Guadagno, A.; Paytuvi-Gallart, A.; Saera-Vila, A.; Amoroso, C.G.; D’Esposito, D.; Andolfo, G.; Aiese Cigliano, R.; Sanseverino, W.; Ercolano, M.R. PRGdb 4.0: An updated database dedicated to genes involved in plant disease resistance process. Nucleic Acids Res. 2022, 50, D1483–D1490. [Google Scholar] [CrossRef] [PubMed]
  85. Cao, C.; Wang, J.; Kwok, D.; Cui, F.; Zhang, Z.; Zhao, D.; Li, M.J.; Zou, Q. webTWAS: A resource for disease candidate susceptibility genes identified by transcriptome-wide association study. Nucleic Acids Res. 2022, 50, D1123–D1130. [Google Scholar] [CrossRef] [PubMed]
  86. Finn, R.D.; Clements, J.; Eddy, S.R. HMMER web server: Interactive sequence similarity searching. Nucleic Acids Res. 2011, 39, W29–W37. [Google Scholar] [CrossRef] [PubMed]
  87. Altschul, S.F.; Madden, T.L.; Schäffer, A.A.; Zhang, J.; Zhang, Z.; Miller, W.; Lipman, D.J. Gapped BLAST and PSI-BLAST: A new generation of protein database search programs. Nucleic Acids Res. 1997, 25, 3389–3402. [Google Scholar] [CrossRef]
  88. Remmert, M.; Biegert, A.; Hauser, A.; Söding, J. HHblits: Lightning-fast iterative protein sequence searching by HMM-HMM alignment. Nat. Methods 2012, 9, 173–175. [Google Scholar] [CrossRef]
  89. Teufel, F.; Almagro Armenteros, J.J.; Johansen, A.R.; Gíslason, M.H.; Pihl, S.I.; Tsirigos, K.D.; Nielsen, H. SignalP 6.0 predicts all five types of signal peptides using protein language models. Nat. Biotechnol. 2022, 40, 1023–1025. [Google Scholar] [CrossRef]
  90. Emanuelsson, O.; Brunak, S.; von Heijne, G.; Nielsen, H. Locating proteins in the cell using TargetP, SignalP and related tools. Nat. Protoc. 2007, 2, 953–971. [Google Scholar] [CrossRef]
  91. Chen, Y.; Yu, P.; Luo, J.; Jiang, Y. Secreted protein prediction system combining CJ-SPHMM, TMHMM, and PSORT. Mamm. Genome 2003, 14, 859–865. [Google Scholar] [CrossRef]
  92. Sperschneider, J.; Catanzariti, A.M.; DeBoer, K.; Petre, B.; Gardiner, D.M.; Singh, K.B.; Taylor, J.M. LOCALIZER: Subcellular localization prediction of both plant and effector proteins in the plant cell. Sci. Rep. 2017, 7, 44598. [Google Scholar] [CrossRef]
  93. Sperschneider, J.; Dodds, P.N.; Singh, K.B.; Taylor, J.M. ApoplastP: Prediction of effectors and plant proteins in the apoplast using machine learning. New Phytol. 2018, 217, 1764–1778. [Google Scholar] [CrossRef]
  94. Gupta, A.; Sankararamakrishnan, R. dbSWEET: An integrated resource for SWEET superfamily to understand, analyze and predict the function of sugar transporters in prokaryotes and eukaryotes. J. Mol. Biol. 2018, 430, 2203–2211. [Google Scholar] [CrossRef] [PubMed]
  95. Sperschneider, J.; Dodds, P.N. EffectorP 3.0: Prediction of apoplastic and cytoplasmic effectors in fungi and oomycetes. Mol. Plant Microbe Interact. 2021, 34, 146–156. [Google Scholar] [CrossRef] [PubMed]
  96. Kulmanov, M.; Khan, M.A.; Hoehndorf, R. DeepGO: Predicting protein functions from sequence and interactions using a deep ontology-aware classifier, Bioinformatics. Bioinformatics 2018, 34, 660–668. [Google Scholar] [CrossRef]
  97. Warde-Farley, D.; Donaldson, S.L.; Comes, O.; Zuberi, K.; Badrawi, R.; Chao, P. The GeneMANIA prediction server: Biological network integration for gene prioritization and predicting gene function. Nucleic Acids Res. 2010, 38, W214–W220. [Google Scholar] [CrossRef]
  98. Altschul, S.F.; Gish, W.; Miller, W.; Myers, E.W.; Lipman, D.J. Basic local alignment search tool. J. Mol. Biol. 1990, 215, 403–410. [Google Scholar] [CrossRef]
  99. Blum, M.; Andreeva, A.; Florentino, L.C.; Chuguransky, S.R.; Grego, T.; Hobbs, E.; Pinto, B.L. InterPro: The protein sequence classification resource in 2025. Nucleic Acids Res. 2025, 53, D444–D456. [Google Scholar] [CrossRef]
  100. Carreón-Anguiano, K.G.; Islas-Flores, I.; Vega-Arreguín, J.; Sáenz-Carbonell, L.; Canto-Canché, B. EffHunter: A tool for prediction of effector protein candidates in fungal proteomic databases. Biomolecules 2020, 10, 712. [Google Scholar] [CrossRef]
  101. Bülow, L.; Schindler, M.; Choi, C.; Hehl, R. PathoPlant: A database on plant-pathogen interactions. Silico Biol. 2004, 4, 529–536. [Google Scholar] [CrossRef]
  102. Lei, C.; Zhou, K.; Zheng, J.; Zhao, M.; Huang, Y.; He, H.; Zhang, Z. Arapathogen2.0: An improved prediction of plant–pathogen protein–protein interactions empowered by the natural language processing technique. J. Proteome Res. 2023, 23, 494–499. [Google Scholar] [CrossRef] [PubMed]
  103. Ammari, M.G.; Gresham, C.R.; McCarthy, F.M.; Nanduri, B. HPIDB 2.0: A curated database for host–pathogen interactions. Database 2016, 2016, baw103. [Google Scholar] [CrossRef]
  104. Urban, M.; Cuzick, A.; Seager, J.; Wood, V.; Rutherford, K.; Venkatesh, S.Y.; Hammond-Kosack, K.E. PHI-base in 2022: A multi-species phenotype database for pathogen–host interactions. Nucleic Acids Res. 2022, 50, D837–D847. [Google Scholar] [CrossRef]
  105. Goodstein, D.M.; Shu, S.; Howson, R.; Neupane, R.; Hayes, R.D.; Fazo, J. Phytozome: A comparative platform for green plant genomics. Nucleic Acids Res. 2012, 40, D1178–D1186. [Google Scholar] [CrossRef]
  106. Bolser, D.M.; Staines, D.M.; Perry, E.; Kersey, P.J. Ensembl Plants: Integrating tools for visualizing, mining, and analyzing plant genomic data. In Plant Genomics Databases; Van Dijk, A., Ed.; Methods in Molecular Biology; Humana Press: New York, NY, USA, 2017; Volume 1533, pp. 1–31. [Google Scholar] [CrossRef]
  107. Reiser, L.; Bakker, E.; Subramaniam, S.; Chen, X.; Sawant, S.; Khosa, K.; Prithvi, T.; Berardini, T.Z. The Arabidopsis Information Resource in 2024. Genetics 2024, 227, iyae027. [Google Scholar] [CrossRef] [PubMed]
  108. Li, L.; Stoeckert, C.J., Jr.; Roos, D.S. OrthoMCL: Identification of Ortholog Groups for Eukaryotic Genomes. Genome Res. 2003, 13, 2178–2189. [Google Scholar] [CrossRef] [PubMed]
  109. Barrett, T.; Wilhite, S.E.; Ledoux, P.; Evangelista, C.; Kim, I.F.; Tomashevsky, M.; Marshall, K.A.; Phillippy, K.H.; Sherman, P.M.; Holko, M. NCBI GEO: Archive for functional genomics data sets—Update. Nucleic Acids Res. 2013, 41, D991–D995. [Google Scholar] [CrossRef] [PubMed]
  110. Hansjakob, A.; Riederer, M.; Hildebrandt, U. Wax matters: Absence of very-long-chain aldehydes from the leaf cuticular wax of the glossy11 mutant of maize compromises the prepenetration processes of Blumeria graminis. Plant Pathol. 2011, 60, 1151–1161. [Google Scholar] [CrossRef]
  111. Wang, E.; Schornack, S.; Marsh, J.F.; Gobbato, E.; Schwessinger, B.; Eastmond, P. A common signaling process that promotes mycorrhizal and oomycete colonization of plants. Curr. Biol. 2012, 22, 2242–2246. [Google Scholar] [CrossRef]
  112. Uppalapati, S.R.; Ishiga, Y.; Doraiswamy, V.; Bedair, M.; Mittal, S.; Chen, J.; Nakashima, J.; Tang, Y.; Tadege, M.; Ratet, P. Loss of abaxial leaf epicuticular wax in Medicago truncatula irg1/palm1 mutants results in reduced spore differentiation of anthracnose and nonhost rust pathogens. Plant Cell 2012, 24, 353–370. [Google Scholar] [CrossRef]
  113. Chassot, C.; Nawrath, C.; Metraux, J.P. The cuticle: Not only a barrier for plant defence—A novel defence syndrome in plants with cuticular defects. Plant Signal Behav. 2008, 3, 142–144. [Google Scholar] [CrossRef]
  114. L’Haridon, F.; Besson-Bard, A.; Binda, M.; Serrano, M.; Abou-Mansour, E. A permeable cuticle is associated with the release of reactive oxygen species and induction of innate immunity. PLoS Pathog. 2011, 7, e1002148. [Google Scholar] [CrossRef]
  115. Zhu, Y.; Nam, J.; Carpita, N.C.; Matthysse, A.G.; Gelvin, S.B. Agrobacterium-mediated root transformation is inhibited by mutation of an Arabidopsis cellulose synthase-like gene. Plant Physiol. 2003, 133, 1000–1010. [Google Scholar] [CrossRef]
  116. Zhu, C.; Perry, S.E. Control of expression and autoregulation of AGL15, a member of the MADS-box family. Plant J. 2005, 41, 583–594. [Google Scholar] [CrossRef]
  117. Desclos-Theveniau, M.; Arnaud, D.; Huang, T.Y.; Lin, G.J.C.; Chen, W.Y.; Lin, Y.C.; Zimmerli, L. The Arabidopsis lectin receptor kinase LecRK-V. 5 represses stomatal immunity induced by Pseudomonas syringae pv. tomato DC3000. PLoS Pathog. 2012, 8, e1002513. [Google Scholar] [CrossRef]
  118. Sawinski, K.; Mersmann, S.; Robatzek, S.; Böhmer, M. Guarding the green: Pathways to stomatal immunity. Mol. Plant-Microbe Interact. 2013, 26, 626–632. [Google Scholar] [CrossRef] [PubMed]
  119. Liu, J.; Elmore, J.M.; Coaker, G. Investigating the functions of the RIN4 protein complex during plant innate immune responses. Plant Signal. Behav. 2009, 4, 1107–1110. [Google Scholar] [CrossRef] [PubMed]
  120. Eichmann, R.; Bischof, M.; Weis, C.; Shaw, J.; Lacomme, C.; Schweizer, P.; Duchkov, D.; Hensel, G.; Kumlehn, J.; Hückelhoven, R. BAX INHIBITOR-1 is required for full susceptibility of barley to powdery mildew. Mol. Plant Microbe Interact. 2010, 23, 1217–1227. [Google Scholar] [CrossRef]
  121. Kim, H.; Kim, J.S. A guide to genome engineering with programmable nucleases. Nat. Rev. Genet. 2014, 15, 321–334. [Google Scholar] [CrossRef]
  122. Schultheiss, H.; Dechert, C.; Kogel, K.H.; Huckelhoven, R. A small GTP-binding host protein is required for entry of powdery mildew fungus into epidermal cells of barley. Plant Physiol. 2002, 128, 1447–1454. [Google Scholar] [CrossRef]
  123. Poraty-Gavra, L.; Zimmermann, P.; Haigis, S.; Bednarek, P.; Hazak, O.; Stelmakh, O.R.; Sadot, E.; Schulze-Lefert, P.; Gruissem, W.; Yalovsky, S. The Arabidopsis Rho of plants GTPase AtROP6 functions in developmental and pathogen response pathways. Plant Physiol. 2013, 161, 1172–1188. [Google Scholar] [CrossRef] [PubMed]
  124. Chen, L.; Shiotani, K.; Togashi, T.; Miki, D.; Aoyama, M.; Wong, H.L.; Kawasaki, T.; Shimamoto, K. Analysis of the Rac/Rop small GTPase family in rice: Expression, subcellular localization and role in disease resistance. Plant Cell Physiol. 2010, 51, 585–595. [Google Scholar] [CrossRef]
  125. Jung, Y.H.; Agrawal, G.K.; Rakwal, R.; Kim, J.A.; Lee, M.O.; Choi, P.G.; Kim, Y.J. Functional characterization of OsRacB GTPase–a potentially negative regulator of basal disease resistance in rice. Plant Physiol. Biochem. 2006, 44, 68–77. [Google Scholar] [CrossRef]
  126. Kessler, S.A.; Shimosato-Asano, H.; Keinath, N.F.; Wuest, S.E.; Ingram, G. Conserved molecular components for pollen tube reception and fungal invasion. Science 2010, 330, 968–971. [Google Scholar] [CrossRef] [PubMed]
  127. Ellinger, D.; Naumann, M.; Falter, C.; Zwikowics, C.; Jamrow, T.; Manisseri, C.; Somerville, S.C.; Voigt, C.A. Elevated early callose deposition results in complete penetration resistance to powdery mildew in Arabidopsis. Plant Physiol. 2013, 161, 1433–1444. [Google Scholar] [CrossRef] [PubMed]
  128. Tao, Z.; Liu, H.; Qiu, D.; Zhou, Y.; Li, X.; Xu, C.; Wang, S. A pair of allelic WRKY genes play opposite roles in rice-bacteria interactions. Plant Physiol. 2009, 151, 936–948. [Google Scholar] [CrossRef] [PubMed]
  129. Pirrello, C.; Malacarne, G.; Moretto, M.; Lenzi, L.; Perazzolli, M.; Zeilmaker, T.; Van den Ackerveken, G.; Pilati, S.; Moser, C.; Giacomelli, L. Grapevine DMR6-1 is a candidate gene for susceptibility to downy mildew. Biomolecules 2022, 12, 182. [Google Scholar] [CrossRef]
  130. Enebe, M.C.; Babalola, O.O. The impact of microbes in the orchestration of plants’ resistance to biotic stress: A disease management approach. Appl. Microbiol. Biotechnol. 2019, 103, 9–25. [Google Scholar] [CrossRef]
  131. Guerrero, J.; Regedanz, E.; Lu, L.; Ruan, J.; Bisaro, D.M.; Sunter, G. Manipulation of the plant host by the geminivirus AC2/C2 protein, a central player in the infection cycle. Front. Plant Sci. 2020, 11, 59. [Google Scholar] [CrossRef]
  132. 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]
  133. van Damme, M.; Zeilmaker, T.; Elberse, J.; Andel, A.; de Sain-van der Velden, M.; van den Ackerveken, G. Downy mildew resistance in Arabidopsis by mutation of HOMOSERINE KINASE. Plant Cell 2009, 21, 2179–2189. [Google Scholar] [CrossRef]
  134. Stuthman, D.D.; Leonard, K.J.; Miller-Garvin, J. Breeding crops for durable resistance to disease. Adv. Agron. 2007, 95, 319–367. [Google Scholar]
  135. Gao, X.; Brodhagen, M.; Isakeit, T.; Brown, S.H.; Göbel, C.; Betran, J.; Feussner, I.; Keller, N.P.; Kolomiets, M.V. Inactivation of the lipoxygenase ZmLOX3 increases susceptibility of maize to Aspergillus spp. Mol. Plant Microbe Interact. 2009, 22, 222–231. [Google Scholar] [CrossRef]
  136. Pathuri, I.P.; Reitberger, I.E.; Hückelhoven, R.; Proels, R.K. Alcohol dehydrogenase 1 of barley modulates susceptibility to the parasitic fungus Blumeria graminis f. sp. hordei. J. Exp. Bot. 2011, 62, 3449–3457. [Google Scholar] [CrossRef]
  137. Vogel, J.P.; Raab, T.K.; Somerville, C.R.; Somerville, S.C. Mutations in PMR5 result in powdery mildew resistance and altered cell wall composition. Plant J. 2004, 40, 968–978. [Google Scholar] [CrossRef]
  138. Callot, C.; Gallois, J.L. Pyramiding resistances based on translation initiation factors in Arabidopsis is impaired by male gametophyte lethality. Plant Signal. Behav. 2014, 9, e27940. [Google Scholar] [CrossRef]
  139. Tsujimoto, Y.; Numaga, T.; Ohshima, K.; Yano, M.; Ohsawa, R. Arabidopsis TOBAMOVIRUS MULTIPLICATION (TOM) 2 locus encodes a transmembrane protein that interacts with TOM1. EMBO J. 2003, 22, 335–343. [Google Scholar] [CrossRef] [PubMed]
  140. Yamanaka, T.; Imai, T.; Satoh, R.; Kawashima, A.; Takahashi, M. Complete inhibition of tobamovirus multiplication by simultaneous mutations in two homologous host genes. J. Virol. 2002, 76, 2491–2497. [Google Scholar] [CrossRef] [PubMed]
  141. Yamanaka, T.; Ohta, T.; Takahashi, M.; Meshi, T.; Schmidt, R. TOM1, an Arabidopsis gene required for efficient multiplication of a tobamovirus, encodes a putative transmembrane protein. Proc. Natl. Acad. Sci. USA 2000, 97, 10107–10112. [Google Scholar] [CrossRef] [PubMed]
  142. Solomon, P.S.; Lowe, R.G.; Tan, K.C.; Waters, O.D.; Oliver, R.P. Stagonospora nodorum: Cause of Stagonospora nodorum blotch of wheat. Mol. Plant Pathol. 2006, 7, 147–156. [Google Scholar] [CrossRef]
  143. Cowger, C.; Ward, B.; Brown-Guedira, G.; Brown, J. Role of effector-sensitivity gene interactions and durability of quantitative resistance to Septoria nodorum blotch in eastern U.S. wheat. Front. Plant Sci. 2020, 11, 155. [Google Scholar] [CrossRef]
  144. Kariyawasam, G.; Nelson, A.C.; Williams, S.; Solomon, P.S.; Faris, J.D.; Friesen, T.L. The necrotrophic pathogen Parastagonospora nodorum is a master manipulator of wheat defense. Mol. Plant Microbe Interact. 2023, 36, 764–773. [Google Scholar] [CrossRef] [PubMed]
  145. Liu, Z.H.; Faris, J.D.; Meinhardt, S.W.; Ali, S.; Rasmussen, J.B.; Friesen, T.L. Genetic and physical mapping of a gene conditioning sensitivity in wheat to a partially purified host-selective toxin produced by Stagonospora nodorum. Phytopathology 2004, 94, 1056–1060. [Google Scholar] [CrossRef] [PubMed]
  146. Liu, Z.; Zhang, Z.; Faris, J.D.; Oliver, R.P.; Syme, R.; McDonald, M.C.; A McDonald, B.; Solomon, P.S.; Lu, S.; Shelver, W.L.; et al. The cysteine-rich necrotrophic effector SnTox1 produced by Stagonospora nodorum triggers susceptibility of wheat lines harboring Snn1. PLoS Pathog. 2012, 8, e1002467. [Google Scholar] [CrossRef]
  147. Liu, Z.; Gao, Y.; Kim, Y.M.; Faris, J.D.; Shelver, W.L.; de Wit, P.J.G.M.; Xu, S.S.; Friesen, T.L. SnTox1, a Parastagonospora nodorum necrotrophic effector, is a dual-function protein that facilitates infection while protecting from wheat-produced chitinases. New Phytol. 2016, 211, 1052–1064. [Google Scholar] [CrossRef]
  148. Shi, G.; Friesen, T.L.; Saini, J.; Xu, S.S.; Rasmussen, J.B.; Faris, J.D. The wheat Snn7 gene confers susceptibility on recognition of the Parastagonospora nodorum necrotrophic effector SnTox7. Plant Genome 2015, 8, 1–10. [Google Scholar] [CrossRef]
  149. Liu, Z.; Faris, J.D.; Oliver, R.P.; Tan, K.C.; Solomon, P.S.; McDonald, M.C.; McDonald, B.A.; Nunez, A.; Lu, S.; Rasmussen, J.B.; et al. SnTox3 acts in effector-triggered susceptibility to induce disease on wheat carrying the Snn3 gene. PLoS Pathog. 2009, 5, e1000581. [Google Scholar] [CrossRef]
  150. Friesen, T.L.; Stukenbrock, E.H.; Liu, Z.; Meinhardt, S.; Ling, H.; Faris, J.D.; Rasmussen, J.B.; Solomon, P.S.; McDonald, B.A.; Oliver, R.P. Emergence of a new disease as a result of interspecific virulence gene transfer. Nature 2006, 38, 953–956. [Google Scholar] [CrossRef]
  151. Friesen, T.L.; Meinhardt, S.W.; Faris, J.D. The Stagonospora nodorum–wheat pathosystem involves multiple proteinaceous host-selective toxins and corresponding host sensitivity genes that interact in an inverse gene-for-gene manner. Plant J. 2007, 51, 681–692. [Google Scholar] [CrossRef]
  152. Abeysekara, N.S.; Friesen, T.L.; Keller, B.; Faris, J.D. Identification and characterization of a novel host-toxin interaction in the wheat–Stagonospora nodorum pathosystem. Theor. Appl. Genet. 2009, 120, 117–126. [Google Scholar] [CrossRef]
  153. Gao, Y.; Faris, J.D.; Liu, Z.; Kim, Y.M.; Syme, R.A.; Oliver, R.P.; Xu, S.S.; Friesen, T.L. Identification and characterization of the SnTox6-Snn6 interaction in the Parastagonospora nodorum–wheat pathosystem. Mol. Plant Microbe Interact. 2015, 28, 615–625. [Google Scholar] [CrossRef] [PubMed]
  154. Shao, D.; Smith, D.L.; Kabbage, M.; Roth, M.G. Effectors of plant necrotrophic fungi. Front. Plant Sci. 2021, 12, 687713. [Google Scholar] [CrossRef]
  155. Peng, Z.; Hu, Y.; Zhang, J.; Huguet-Tapia, J.C.; Block, A.K.; Park, S.; Sapkota, S.; Liu, Z.; Liu, S.; White, F.F. Xanthomonas translucens commandeers the host rate-limiting step in ABA biosynthesis for disease susceptibility. Proc. Natl. Acad. Sci. USA 2019, 116, 20938–20946. [Google Scholar] [CrossRef] [PubMed]
  156. Shah, S.M.A.; Khojasteh, M.; Wang, Q.; Haq, F.; Xu, X.; Li, Y.; Zou, L.; Osdaghi, E.; Chen, G. Comparative transcriptomic analysis of wheat cultivars in response to Xanthomonas translucens pv. cerealis and its T2SS, T3SS, and TALEs deficient strains. Phytopathology 2023, 113, 2073–2082. [Google Scholar]
  157. Lorang, J.; Kidarsa, T.; Bradford, C.S.; Gilbert, B.; Curtis, M.; Tzeng, S.C.; Maier, C.S.; Wolpert, T.J. Tricking the guard: Exploiting plant defense for disease susceptibility. Science 2012, 338, 659–662. [Google Scholar] [CrossRef]
  158. Smýkal, P.; Šafářová, D.; Navrátil, M.; Dostalová, R. Marker assisted pea breeding: eIF4E allele specific markers to pea seed-borne mosaic virus (PSbMV) resistance. Mol. Breed. 2010, 26, 425–438. [Google Scholar]
  159. Cook, A.A. A mutation for resistance to potato virus Y in pepper. Proc. Am. Soc. Hortic. Sci. 1961, 78, 550–552. [Google Scholar]
  160. Moury, B.; Verdin, E. Viruses of pepper crops in the Mediterranean basin: A remarkable stasis. Adv. Virus Res. 2012, 84, 127–162. [Google Scholar]
  161. Ayme, V.; Souche, S.; Caranta, C.; Jacquemond, M.; Chadoeuf, J.; Palloix, A.; Moury, B. Different mutations in the genome-linked protein VPg of Potato virus Y confer virulence on the pvr23 resistance in pepper. Mol. Plant Microbe Interact. 2006, 19, 557–563. [Google Scholar] [CrossRef]
  162. Zlobin, N.; Taranov, V. Plant eIF4E isoforms as factors of susceptibility and resistance to potyviruses. Front. Plant Sci. 2023, 14, 1041868. [Google Scholar] [CrossRef] [PubMed]
  163. Masuta, C.; Nishimura, M.; Morishita, H.; Hataya, T. A single amino acid change in viral genome associated protein of potato virus Y correlates with resistance breaking in ‘Virgin A Mutant’ tobacco. Phytopathology 1999, 89, 118–123. [Google Scholar] [CrossRef] [PubMed]
  164. Truniger, V.; Aranda, M.A. Recessive resistance to plant viruses. Adv. Virus Res. 2009, 75, 119–231. [Google Scholar]
  165. Wittmann, S.; Chatel, H.; Fortin, M.G.; Laliberté, J.F. Interaction of the viral protein genome linked of turnip mosaic potyvirus with the translational eukaryotic initiation factor (iso) 4E of Arabidopsis thaliana using the yeast two-hybrid system. Virology 1997, 234, 84–92. [Google Scholar] [CrossRef]
  166. Ruffel, S.; Dussault, M.H.; Palloix, A.; Moury, B.; Bendahmane, A.; Robaglia, C.; Caranta, C. A natural recessive resistance gene against potato virus Y in pepper corresponds to the eukaryotic initiation factor 4E (eIF4E). Plant J. 2002, 32, 1067–1075. [Google Scholar] [CrossRef] [PubMed]
  167. Nicaise, V.; German-Retana, S.; Sanjuán, R.; Dubrana, M.-P.; Mazier, M.; Maisonneuve, B.; Candresse, T.; Caranta, C.; LeGall, O. The eukaryotic translation initiation factor 4E controls lettuce susceptibility to the potyvirus lettuce mosaic virus. Plant Physiol. 2003, 132, 1272–1282. [Google Scholar] [CrossRef]
  168. Gao, Z.; Johansen, E.; Eyers, S.; Thomas, C.L.; Ellis, T.H.N.; Maule, A.J. The potyvirus recessive resistance gene, sbm1, identifies a novel role for eIF4E in cell-to-cell trafficking. Plant J. 2004, 40, 376–385. [Google Scholar] [CrossRef] [PubMed]
  169. Stein, N.; Perovic, D.; Kumlehn, J.; Pellio, B.; Stracke, S.; Streng, S.; Ordon, F.; Graner, A. The eukaryotic translation initiation factor 4E confers multiallelic recessive Bymovirus resistance in Hordeum vulgare (L.). Plant J. 2005, 42, 912–922. [Google Scholar] [CrossRef] [PubMed]
  170. Abdul-Razzak, A.N.A.S.; Guiraud, T.; Peypelut, M.; Walter, J.; Houvenaghel, M.C.; Candresse, T.; Le Gall, O.; German-Retana, S. Involvement of the cylindrical inclusion (CI) protein in the overcoming of an eIF4E-mediated resistance against Lettuce mosaic potyvirus. Mol. Plant Pathol. 2009, 10, 109–113. [Google Scholar] [CrossRef]
  171. Nakahara, K.S.; Shimada, R.; Choi, S.H.; Yamamoto, H.; Shao, J.; Uyeda, I. Involvement of the P1 cistron in overcoming eIF4E-mediated recessive resistance against Clover yellow vein virus in pea. Mol. Plant Microbe Interact. 2010, 23, 1460–1469. [Google Scholar] [CrossRef]
  172. Chandrasekaran, J.; Brumin, M.; Wolf, D.; Leibman, D.; Klap, C.; Pearlsman, M.; Sherman, A.; Arazi, T.; Gal-On, A. Development of broad virus resistance in non-transgenic cucumber using CRISPR/Cas9 technology. Mol. Plant Pathol. 2016, 17, 1140–1153. [Google Scholar] [CrossRef]
  173. Pyott, D.E.; Sheehan, E.; Molnar, A. Engineering of CRISPR/Cas9-mediated potyvirus resistance in transgene-free Arabidopsis plants. Mol. Plant Pathol. 2016, 17, 1276–1288. [Google Scholar] [CrossRef]
  174. Macovei, A.; Sevilla, N.R.; Cantos, C.; Jonson, G.B.; Slamet-Loedin, I.; Čermák, T.; Voytas, D.F.; Choi, I.R.; Chadha-Mohanty, P. Novel alleles of rice eIF4G generated by CRISPR/Cas9-targeted mutagenesis confer resistance to rice tungro spherical virus. Plant Biotechnol. J. 2018, 16, 1918–1927. [Google Scholar] [CrossRef]
  175. Coutinho de Oliveira, L.; Volpon, L.; Rahardjo, A.K.; Osborne, M.J.; Culjkovic-Kraljacic, B.; Trahan, C.; Oeffinger, M.; Kwok, B.H.; Borden, K.L.B. Structural studies of the eIF4E–VPg complex reveal a direct competition for capped RNA: Implications for translation. Proc. Natl. Acad. Sci. USA 2019, 116, 24056–24065. [Google Scholar] [CrossRef] [PubMed]
  176. Bastet, A.; Zafirov, D.; Giovinazzo, N.; Guyon-Debast, A.; Nogué, F.; Robaglia, C.; Gallois, J.L. Mimicking natural polymorphism in eIF4E by CRISPR-Cas9 base editing is associated with resistance to potyviruses. Plant Biotechnol. J. 2019, 17, 1736–1750. [Google Scholar] [CrossRef]
  177. Atarashi, H.; Jayasinghe, W.H.; Kwon, J.; Kim, H.; Taninaka, Y.; Igarashi, M.; Ito, K.; Yamada, T.; Masuta, C.; Nakahara, K.S. Artificially edited alleles of the eukaryotic translation initiation factor 4E1 gene differentially reduce susceptibility to cucumber mosaic virus and potato virus Y in tomato. Front. Microbiol. 2020, 11, 564310. [Google Scholar] [CrossRef]
  178. Yoon, Y.J.; Venkatesh, J.; Lee, J.H.; Kim, J.; Lee, H.E. Genome editing of eIF4E1 in tomato confers resistance to pepper mottle virus. Front. Plant Sci. 2020, 11, 1098. [Google Scholar] [CrossRef] [PubMed]
  179. Kuroiwa, K.; Thenault, C.; Nogué, F.; Perrot, L.; Mazier, M.; Gallois, J.L. CRISPR-based knock-out of eIF4E2 in a cherry tomato background successfully recapitulates resistance to pepper veinal mottle virus. Plant Sci. 2022, 316, 111160. [Google Scholar] [CrossRef] [PubMed]
  180. Gomez, M.A.; Lin, Z.D.; Moll, T.; Chauhan, R.D.; Hayden, L.; Renninger, K. Simultaneous CRISPR/Cas9-mediated editing of cassava eIF4E isoforms nCBP-1 and nCBP-2 reduces cassava brown streak disease symptom severity and incidence. Plant Biotechnol. J. 2019, 17, 421–434. [Google Scholar] [CrossRef]
  181. Noureen, A.; Khan, M.Z.; Amin, I.; Zainab, T.; Mansoor, S. CRISPR/Cas9-mediated targeting of susceptibility factor eIF4E-enhanced resistance against potato virus Y. Front. Genet. 2022, 13, 922019. [Google Scholar] [CrossRef]
  182. Shirazi Parsa, H.; Sabet, M.S.; Moieni, A.; Shojaeiyan, A.; Dogimont, C.; Boualem, A.; Bendahmane, A. CRISPR/Cas9-mediated cytosine base editing using an improved transformation procedure in melon (Cucumis melo L.). Int. J. Mol. Sci. 2023, 24, 11189. [Google Scholar] [CrossRef] [PubMed]
  183. Pechar, G.S.; Donaire, L.; Gosalvez, B.; García-Almodovar, C.; Sánchez-Pina, M.A.; Truniger, V.; Aranda, M.A. Editing melon EIF4E associates with virus resistance and male sterility. Plant Biotechnol. J. 2022, 20, 2006–2022. [Google Scholar] [CrossRef]
  184. Fidan, H.; Calis, O.; Ari, E.; Atasayar, A.; Sarikaya, P.; Tek, M.I.; Izmirli, A.; Oz, Y.; Firat, G. Knockout of eIF4E using CRISPR/Cas9 for large-scale production of resistant cucumber cultivar against WMV, ZYMV, and PRSV. Front. Plant Sci. 2023, 14, 1143813. [Google Scholar] [CrossRef]
  185. Li, M.; Qiu, Y.; Zhu, D.; Xu, X.; Tian, S.; Wang, J.; Yu, Y. Editing eIF4E in the watermelon genome using CRISPR/Cas9 technology confers resistance to ZYMV. Int. J. Mol. Sci. 2024, 25, 11468. [Google Scholar] [CrossRef]
  186. Jørgensen, I.H. Discovery, characterization and exploitation of Mlo powdery mildew resistance in barley. Euphytica 1992, 63, 141–152. [Google Scholar] [CrossRef]
  187. Tek, M.I.; Calis, O.; Fidan, H.; Shah, M.D.; Celik, S.; Wani, S.H. CRISPR/Cas9-based Mlo-mediated resistance against Podosphaera xanthii in cucumber (Cucumis sativus L.). Front. Plant Sci. 2022, 13, 1081506. [Google Scholar] [CrossRef] [PubMed]
  188. Li, S.; Lin, D.; Zhang, Y.; Deng, M.; Chen, Y.; Lv, B.; Li, B.; Lei, Y.; Wang, Y.; Zhao, L. Genome-edited powdery mildew resistance in wheat without growth penalties. Nature 2022, 602, 455–460. [Google Scholar] [CrossRef] [PubMed]
  189. Wan, D.Y.; Guo, Y.; Cheng, Y.; Hu, Y.; Xiao, S.; Wang, Y. CRISPR/Cas9-mediated mutagenesis of VvMLO3 results in enhanced resistance to powdery mildew in grapevine (Vitis vinifera). Hortic. Res. 2020, 7, 116. [Google Scholar] [CrossRef]
  190. Shnaider, Y.; Elad, Y.; Rav-David, D.; Pashkovsky, E.; Leibman, D.; Kravchik, M.; Shtarkman-Cohen, M.; Gal-On, A.; Spiegelman, Z. Development of powdery mildew resistance in cucumber using CRISPR/Cas9-mediated mutagenesis of CsaMLO8. Phytopathology 2023, 113, 786–790. [Google Scholar] [CrossRef]
  191. Nekrasov, V.; Wang, C.; Win, J.; Lanz, C.; Weigel, D.; Kamoun, S. Rapid generation of a transgene-free powdery mildew resistant tomato by genome deletion. Sci. Rep. 2017, 7, 482. [Google Scholar] [CrossRef]
  192. Pramanik, D.; Shelake, R.M.; Park, J.; Kim, M.J.; Hwang, I.; Park, Y.; Kim, J.Y. CRISPR/Cas9-mediated generation of pathogen-resistant tomato against tomato yellow leaf curl virus and powdery mildew. Int. J. Mol. Sci. 2021, 22, 1878. [Google Scholar] [CrossRef]
  193. Bui, T.P.; Le, H.; Ta, D.T. Enhancing powdery mildew resistance in soybean by targeted mutation of MLO genes using the CRISPR/Cas9 system. BMC Plant Biol. 2023, 23, 533. [Google Scholar] [CrossRef]
  194. Antony, G.; Zhou, J.; Huang, S.; Li, T.; Liu, B.; White, F.; Yang, B. Rice xa13 recessive resistance to bacterial blight is defeated by induction of the disease susceptibility gene Os-11N3. Plant Cell 2010, 22, 3864–3876. [Google Scholar] [CrossRef] [PubMed]
  195. Oliva, R.; Ji, C.; Atienza-Grande, G.; Huguet-Tapia, J.C.; Perez-Quintero, A.; Li, T.; Eom, J.S.; Li, C.; Nguyen, H.; Liu, B.; et al. Broad-spectrum resistance to bacterial blight in rice using genome editing. Nat. Biotechnol. 2019, 37, 1344–1350. [Google Scholar] [CrossRef]
  196. Xu, Z.; Xu, X.; Gong, Q.; Li, Z.; Li, Y.; Wang, S. Engineering broad-spectrum bacterial blight resistance by simultaneously disrupting variable TALE-binding elements of multiple susceptibility genes in rice. Mol. Plant 2019, 12, 1434–1446. [Google Scholar] [CrossRef]
  197. Zafar, K.; Khan, M.Z.; Amin, I.; Mukhtar, Z.; Yasmin, S.; Arif, M.; Ejaz, K.; Mansoor, S. Precise CRISPR-Cas9 mediated genome editing in Super Basmati rice for resistance against bacterial blight by targeting the major susceptibility gene. Front. Plant Sci. 2020, 11, 575. [Google Scholar] [CrossRef]
  198. Zeng, X.; Luo, Y.; Vu, N.T.Q. CRISPR/Cas9-mediated mutation of OsSWEET14 in rice cv. Zhonghua11 confers resistance to Xanthomonas oryzae pv. oryzae without yield penalty. BMC Plant Biol. 2020, 20, 313. [Google Scholar]
  199. Aji, C.M.; Parjanto; Ahmad, Y. Evaluation of Agronomic Performance of Mutant Rice Lines of Mentik Wangi Variety (Oryza sativa L.) Resulting from OSSWEET11 Gene Editing. Int. J. Des. Nat. Ecodyn. 2025, 20, 289–298. [Google Scholar] [CrossRef]
  200. Li, C.; Liu, B.; Dong, H.; Yang, B. Enhancing resistance to bacterial blight in rice using CRISPR-based base editing technology. Crop J. 2025, 13, 115–124. [Google Scholar]
  201. Thomazella, P.D.; Brail, Q.; Dahlbeck, D.; Staskawicz, B. CRISPR-Cas9 mediated mutagenesis of a DMR6 ortholog in tomato confers broad-spectrum disease resistance. bioRxiv 2016. [Google Scholar] [CrossRef]
  202. Frye, C.A.; Tang, D.; Innes, R.W. Negative regulation of defense responses in plants by a conserved MAPKK kinase. Proc. Natl. Acad. Sci. USA 2001, 98, 373–378. [Google Scholar] [CrossRef]
  203. Zhang, Y.; Bai, Y.; Wu, G.; Zou, S.; Chen, Y.; Gao, C.; Tang, D. Simultaneous modification of three homoeologs of TaEDR1 by genome editing enhances powdery mildew resistance in wheat. Plant J. 2017, 91, 714–724. [Google Scholar] [CrossRef]
  204. Pompili, V.; Dalla Costa, L.; Piazza, S.; Pindo, M.; Malnoy, M. Reduced fire blight susceptibility in apple cultivars using a high-efficiency CRISPR/Cas9-FLP/FRT-based gene editing system. Plant Biotechnol. J. 2019, 18, 845–858. [Google Scholar] [CrossRef]
  205. Santillán Martínez, M.I.; Bracuto, V.; Koseoglou, E.; Appiano, M.; Jacobsen, E.; Visser, R.G.F.; Wolters, A.M.A.; Bai, Y. CRISPR/Cas9-targeted mutagenesis of the tomato susceptibility gene PMR4 for resistance against powdery mildew. BMC Plant Biol. 2020, 20, 497. [Google Scholar] [CrossRef]
  206. Kieu, N.P.; Lenman, M.; Wang, E.S.; Petersen, B.L.; Andreasson, E. Mutations introduced in susceptibility genes through CRISPR/Cas9 genome editing confer increased late blight resistance in potatoes. Sci. Rep. 2021, 11, 4487. [Google Scholar] [CrossRef]
  207. Yang, X.; Zhang, L.; Yang, Y.; Schmid, M.; Wang, Y. miRNA mediated regulation and interaction between plants and pathogens. Int. J. Mol. Sci. 2021, 22, 2913. [Google Scholar] [CrossRef] [PubMed]
  208. Zhang, C.; Anarjan, M.B.; Win, K.T.; Begum, S.; Lee, S. QTL-seq analysis of powdery mildew resistance in a Korean cucumber inbred line. Theor. Appl. Genet. 2021, 134, 435–451. [Google Scholar] [CrossRef] [PubMed]
  209. Cha, J.Y.; Uddin, S.; Macoy, D.M.; Shin, G.I.; Jeong, S.Y.; Ali, I.; Hwang, J.W.; Ji, M.G.; Lee, S.C.; Park, J.H. Nucleoredoxin gene SINRX1 negatively regulates tomato immunity by activating SA signaling pathway. Plant Physiol. Biochem. 2023, 200, 107804. [Google Scholar] [CrossRef]
  210. Azad, M.F.; Dawar, P.; Esim, N.; Rock, C.D. Role of miRNAs in sucrose stress response, reactive oxygen species, and anthocyanin biosynthesis in Arabidopsis thaliana. Front. Plant Sci. 2023, 14, 1278320. [Google Scholar] [CrossRef] [PubMed]
  211. Zhang, Y.; Waseem, M.; Zeng, Z.; Xu, J.; Chen, C.; Liu, Y.; Zhai, J.; Xia, R. MicroRNA482/2118, a miRNA superfamily essential for both disease resistance and plant development. New Phytol. 2022, 233, 2047–2057. [Google Scholar] [CrossRef]
  212. Bigini, V.; Camerlengo, F.; Botticella, E.; Sestili, F.; Savatin, D.V. Biotechnological resources to increase disease-resistance by improving plant immunity: A sustainable approach to save cereal crop production. Plants 2021, 10, 1146. [Google Scholar] [CrossRef]
  213. Bi, W.; Liu, J.; Li, Y.; He, Z.; Chen, Y.; Zhao, T.; Liang, X.; Wang, X.; Meng, X.; Dou, D.; et al. CRISPR/Cas9-guided editing of a novel susceptibility gene in potato improves Phytophthora resistance without growth penalty. Plant Biotechnol. J. 2024, 22, 4–6. [Google Scholar] [CrossRef]
  214. Huang, Q.; Lin, B.; Cao, Y.; Zhang, Y.; Song, H.; Huang, C.; Sun, T.; Long, C.; Liao, J.; Zhuo, K. CRISPR/Cas9-mediated mutagenesis of the susceptibility gene OsHPP04 in rice confers enhanced resistance to rice root-knot nematode. Front. Plant Sci. 2023, 14, 1134653. [Google Scholar] [CrossRef] [PubMed]
  215. Tripathi, J.N.; Ntui, V.O.; Shah, T.; Tripathi, L. CRISPR/Cas9-mediated editing of DMR6 orthologue in banana (Musa spp.) confers enhanced resistance to bacterial disease. Plant Biotechnol. J. 2021, 19, 1291–1293. [Google Scholar] [CrossRef]
  216. Ma, M.; Yang, L.; Hu, Z.; Mo, C.; Geng, S.; Zhao, X.; He, Q.; Xiao, L.; Lu, L.; Wang, D. Multiplex gene editing reveals cucumber Mildew Resistance Locus O family roles in powdery mildew resistance. Plant Physiol. 2024, 195, 1069–1088. [Google Scholar] [CrossRef]
  217. Giacomelli, L.; Zeilmaker, T.; Giovannini, O.; Salvagnin, U.; Masuero, D.; Franceschi, P.; Vrhovsek, U.; Scintilla, S.; Rouppe van der Voort, J.; Moser, C. Simultaneous editing of two DMR6 genes in grapevine results in reduced susceptibility to downy mildew. Front. Plant Sci. 2023, 14, 1242240. [Google Scholar] [CrossRef]
  218. Djennane, S.; Gersch, S.; Le-Bohec, F.; Piron, M.-C.; Baltenweck, R.; Lemaire, O.; Merdinoglu, D.; Hugueney, P.; Nogué, F.; Mestre, P. CRISPR/Cas9 editing of Downy mildew resistant 6 (DMR6-1) in grapevine leads to reduced susceptibility to Plasmopara viticola. J. Exp. Bot. 2023, 75, 2100–2112. [Google Scholar] [CrossRef]
  219. Nizan, S.; Amitzur, A.; Dahan-Meir, T.; Benichou, J.I.; Bar-Ziv, A.; Perl-Treves, R. Mutagenesis of the melon Prv gene by CRISPR/Cas9 breaks papaya ringspot virus resistance and generates an autoimmune allele with constitutive defense responses. J. Exp. Bot. 2023, 74, 4579–4596. [Google Scholar] [CrossRef] [PubMed]
  220. Norouzi, M.; Nazarain-Firouzabadi, F.; Ismaili, A.; Ahmadvand, R.; Poormazaheri, H. CRISPR/Cas StNRL1 gene knockout increases resistance to late blight and susceptibility to early blight in potato. Front. Plant Sci. 2024, 14, 1278127. [Google Scholar] [CrossRef]
  221. Ye, W.; Hossain, R.; Pröbsting, M.; Ali, A.A.M.; Han, L. Knock-out of BnHva22c reduces the susceptibility of Brassica napus to infection with the fungal pathogen Verticillium longisporum. Crop J. 2024, 12, 503–514. [Google Scholar] [CrossRef]
  222. Zhao, C.; Zhang, Y.; Gao, L.; Xie, M.; Zhang, X.; Zeng, L.; Liu, J.; Liu, Y.; Zhang, Y.; Tong, C.; et al. Genome editing of RECEPTOR-LIKE KINASE 902 confers resistance to necrotrophic fungal pathogens in Brassica napus without growth penalties. Plant Biotechnol. J. 2024, 22, 538–540. [Google Scholar] [CrossRef]
  223. Hasley, J.A.R.; Navet, N.; Tian, M. CRISPR/Cas9-mediated mutagenesis of sweet basil candidate susceptibility gene ObDMR6 enhances downy mildew resistance. PLoS ONE 2021, 16, e0253245. [Google Scholar] [CrossRef] [PubMed]
  224. Ortigosa, A.; Gimenez-Ibanez, S.; Leonhardt, N.; Solano, R. Design of a bacterial speck resistant tomato by CRISPR/Cas9-mediated editing of SlJAZ2. Plant Biotechnol. J. 2019, 17, 665–673. [Google Scholar] [CrossRef] [PubMed]
  225. Hong, Y.; Meng, J.; He, X.; Zhang, Y.; Liu, Y.; Zhang, C.; Qi, H.; Luan, Y. Editing mir482b and mir482c simultaneously by CRISPR/Cas9 enhanced tomato resistance to Phytophthora infestans. Phytopathology 2021, 111, 1008–1016. [Google Scholar] [CrossRef]
  226. Thomazella, D.P.; Seong, K.; Mackelprang, R.; Dahlbeck, D.; Geng, Y.; Gill, U.S.; Qi, T.; Pham, J.; Giuseppe, P.; Lee, C.Y. Loss of function of a DMR6 ortholog in tomato confers broad-spectrum disease resistance. Proc. Natl. Acad. Sci. USA 2021, 118, e2026152118. [Google Scholar] [CrossRef]
  227. Aalders, T.R.; de Sain, M.; Gawehns, F.; Oudejans, N.; Jak, Y.D.; Dekker, H.L.; Takken, F.L. Specific members of the TOPLESS family are susceptibility genes for Fusarium wilt in tomato and Arabidopsis. Plant Biotechnol. J. 2024, 22, 248–261. [Google Scholar] [CrossRef] [PubMed]
  228. Blanvillain-Baufum, S.M.R.; Sol, M.; Auguy, F.; Doucoure, H.; Szurek, B.; Meynard, D.; Portefaix, M.; Cunnac, S.; Guiderdoni, E.; Boch, J.; et al. Targeted promoter editing for rice resistance to Xanthomonas oryzae pv. oryzae reveals differential activities for SWEET14-inducing TAL effectors. Plant Biotechnol. J. 2017, 15, 306–317. [Google Scholar] [CrossRef]
  229. Zhou, J.; Peng, Z.; Long, J.; Sosso, D.; Liu, B.; Eom, J.S.; Huang, S.; Liu, S.; Vera Cruz, C.; Frommer, W.B.; et al. Gene targeting by the TAL effector PthXo2 reveals cryptic resistance gene for bacterial blight of rice. Plant J. 2015, 82, 632–643. [Google Scholar] [CrossRef]
  230. Xu, Z.; Xu, X.; Li, Y.; Liu, L.; Wang, Q.; Wang, Y. Tal6b/AvrXa27A, a hidden TALE targeting the susceptibility gene OsSWEET11a and the resistance gene Xa27 in rice. Plant Commun. 2024, 5, 100721. [Google Scholar] [CrossRef]
  231. Sha, G.; Sun, P.; Kong, X.; Han, X.; Sun, Q.; Fouillen, L. Genome editing of a rice CDP-DAG synthase confers multipathogen resistance. Nature 2023, 618, 1017–1023. [Google Scholar] [CrossRef]
  232. Song, H.; Lin, B.; Huang, Q.; Sun, L.; Chen, J.; Hu, L.; Zhuo, K.; Liao, J. The Meloidogyne graminicola effector MgMO289 targets a novel copper metallochaperone to suppress immunity in rice. J. Exp. Bot. 2021, 72, 5638–5655. [Google Scholar] [CrossRef] [PubMed]
  233. Tao, H.; Shi, X.; He, F.; Wang, D.; Xiao, N.; Fang, H.; Wang, R.; Zhang, F.; Wang, M.; Li, A. Engineering broad-spectrum disease-resistant rice by editing multiple susceptibility genes. J. Integr. Plant Biol. 2021, 63, 1639–1648. [Google Scholar] [CrossRef]
  234. Wang, F.; Wang, C.; Liu, P.; Lei, C.; Hao, W.; Gao, Y.; Zhao, K. Enhanced rice blast resistance by CRISPR/Cas9-targeted mutagenesis of the ERF transcription factor gene OsERF922. PLoS ONE 2016, 11, e0154027. [Google Scholar] [CrossRef] [PubMed]
  235. Peng, A.; Chen, S.; Lei, T.; Xu, L.; He, Y.; Wu, L.; Yao, L.; Zou, X. Engineering canker-resistant plants through CRISPR/Cas9-targeted editing of the susceptibility gene CsLOB1 promoter in citrus. Plant Biotechnol. J. 2017, 15, 1509–1519. [Google Scholar] [CrossRef] [PubMed]
  236. Zhang, M.; Liu, Q.; Yang, X.; Xu, J.; Liu, G.; Yao, X.; Lou, L. CRISPR/Cas9-mediated mutagenesis of Clpsk1 in watermelon to confer resistance to Fusarium oxysporum f. sp. niveum. Plant Cell Rep. 2020, 39, 589–595. [Google Scholar] [CrossRef]
  237. Wang, N.; Fan, X.; He, M.; Hu, Z.; Tang, C.; Zhang, S.; Wang, X. Transcriptional repression of TaNOX10 by TaWRKY19 compromises ROS generation and enhances wheat susceptibility to stripe rust. Plant Cell 2022, 34, 1784–1803. [Google Scholar] [CrossRef] [PubMed]
  238. Liu, S.; Liu, H.; Guo, M.; Pan, Y.; Hao, C.; Hou, J.; Li, T. Knockout of GRAIN WIDTH2 has a dual effect on enhancing leaf rust resistance and increasing grain weight in wheat. Plant Biotechnol. J. 2024, 22, 2007. [Google Scholar] [CrossRef]
  239. El-Mounadi, K.; Morales-Floriano, M.L.; Garcia-Ruiz, H. Principles, applications, and biosafety of plant genome editing using CRISPR-Cas9. Front. Plant Sci. 2020, 11, 56. [Google Scholar] [CrossRef]
  240. Plan, D.; Van den Eede, G. The EU Legislation on GMOs, An Overview; Publications Office of the European Union: Luxembourg, 2010. [Google Scholar]
  241. Callaway, E. EU law deals blow to CRISPR crops. Nature 2018, 560, 16. [Google Scholar] [CrossRef]
  242. Singh, D.J.K.; Mat Jalaluddin, N.S.; Sanan-Mishra, N.; Harikrishna, J.A. Genetic modification in Malaysia and India: Current regulatory framework and the special case of non-transformative RNAi in agriculture. Plant Cell Rep. 2019, 38, 1449–1463. [Google Scholar] [CrossRef]
  243. Waltz, E. Gene-edited CRISPR mushroom escapes US regulation. Nature 2016, 532, 293. [Google Scholar] [CrossRef]
  244. Fister, A.S.; Landherr, L.; Maximova, S.N.; Guiltinan, M.J. Transient expression of CRISPR/Cas9 machinery targeting TcNPR3 enhances defense response in Theobroma cacao. Front. Plant Sci. 2018, 9, 268. [Google Scholar] [CrossRef]
  245. Lee, S.-W. Current status on the modification of the scope for GMO regulation on the gene edited plants with no remnants of inserted foreign DNA fragments. J. Plant Biotechnol. 2019, 46, 137–142. [Google Scholar] [CrossRef]
  246. Buchholzer, M.; Frommer, W.B. An increasing number of countries regulate genome editing in crops. New Phytol. 2023, 237, 12–15. [Google Scholar] [CrossRef]
  247. Bhatta, B.P.; Malla, S. Improving horticultural crops via CRISPR/Cas9: Current successes and prospects. Plants 2020, 9, 1360. [Google Scholar] [CrossRef] [PubMed]
  248. Rukavtsova, E.B.; Zakharchenko, N.S.; Lebedev, V.G.; Shestibratov, K.A. CRISPR-Cas genome editing for horticultural crops improvement: Advantages and prospects. Horticulturae 2023, 9, 38. [Google Scholar] [CrossRef]
  249. Li, J.; Zhang, C.; He, Y.; Li, S.; Yan, L.; Li, Y.; Zhu, Z.; Xia, L. Plant base editing and prime editing: The current status and future perspectives. J. Integr. Plant Biol. 2023, 65, 444–467. [Google Scholar] [CrossRef] [PubMed]
  250. Cui, Y.; Cao, Q.; Li, Y.; He, M.; Liu, X. Advances in cis-element- and natural variation-mediated transcriptional regulation and applications in gene editing of major crops. J. Exp. Bot. 2023, 74, 5441–5457. [Google Scholar] [CrossRef]
Figure 1. Plant defense mechanisms against biotic stress involve multiple layers of immunity. PAMPs/DAMPs are detected by PRRs on the cell surface, such as FLS2, which trigger PTI through the activation of MAPK cascades or calcium-dependent protein kinases (CDPKs). Effectors produced by bacteria, virus, fungi, and oomycetes that entre the plant cell are recognized by NOD-like receptors (NLRs), initiating effector-triggered immune ETI and leading to HR responses. The letter P indicates phosphorylation, SA/JA—salicylic acid (SA) and jasmonic acid (JA); ROS—Reactive oxygen species.
Figure 1. Plant defense mechanisms against biotic stress involve multiple layers of immunity. PAMPs/DAMPs are detected by PRRs on the cell surface, such as FLS2, which trigger PTI through the activation of MAPK cascades or calcium-dependent protein kinases (CDPKs). Effectors produced by bacteria, virus, fungi, and oomycetes that entre the plant cell are recognized by NOD-like receptors (NLRs), initiating effector-triggered immune ETI and leading to HR responses. The letter P indicates phosphorylation, SA/JA—salicylic acid (SA) and jasmonic acid (JA); ROS—Reactive oxygen species.
Plants 14 03080 g001
Figure 2. Comparative overview of R-genes and S-genes, highlighting mechanisms, editing applications, advantages, and limitations.
Figure 2. Comparative overview of R-genes and S-genes, highlighting mechanisms, editing applications, advantages, and limitations.
Plants 14 03080 g002
Figure 3. Methods for identifying S-genes include QTL mapping, GWAS, transcriptomic analysis, and homology-based searches to discover candidate genes associated with pathogen interactions.
Figure 3. Methods for identifying S-genes include QTL mapping, GWAS, transcriptomic analysis, and homology-based searches to discover candidate genes associated with pathogen interactions.
Plants 14 03080 g003
Figure 4. Mode of action of different classes of S-genes during pathogen infection.
Figure 4. Mode of action of different classes of S-genes during pathogen infection.
Plants 14 03080 g004
Figure 5. Global regulatory landscape for genome-edited crops.
Figure 5. Global regulatory landscape for genome-edited crops.
Plants 14 03080 g005
Table 1. Databases and computational tools for susceptibility (S) gene identification and functional prediction (all links accessed on 27 September 2025).
Table 1. Databases and computational tools for susceptibility (S) gene identification and functional prediction (all links accessed on 27 September 2025).
Database
/Software
Domain NameUsesFramework & Model TypeReferences
PRGdb 4.0 http://prgdb.org/prgdb4/To predict resistance genes and investigate gene expression under specific plant–pathogen conditions-García et al., 2022 [84]
webTWAS http://www.webtwas.net/#/A database of potential disease S-genes identified through transcriptome-wide association studies.-Ca et al., 2022 [85]
HMMER www.hmmer.orgGeneral profile-HMM database search and alignment tool-Finn et al., 2011 [86]
PSIBLASThttps://www.ebi.ac.uk/jdispatcher/sss/psiblastProgressive BLAST algorithm employing PSSMs-Altschul et al., 1997 [87]
Hhblitshttp://www.github.com/soedinglab/hh-suiteIdentification of distant sequence homologs-Remmert et al., 2012 [88]
SignalPhttp://www.cbs.dtu.dk/services/SignalPPrediction of secreted proteins by detecting N-terminal signal peptides, a common initial step for effector prediction-Teufel et al., 2022 [89]
TargetPhttp://www.cbs.dtu.dk/services/TargetP General prediction of protein subcellular or extracellular localization -Emanuelsson et al., 2007 [90]
TMHMMhttp://www.cbs.dtu.dk/services/TMHMM Prediction of transmembrane domains in proteins -Chen 2003 [91]
LOCALIZERhttps://localizer.csiro.au Prediction of localization within host plant cells -Sperschneider et al., 2017 [92]
ApoplastPhttps://apoplastp.csiro.au Prediction of proteins localized in the plant apoplast -Sperschneider et al., 2018 [93]
dbSWEEThttps://ngdc.cncb.ac.cn/databasecommons/database/id/6133SWEET transporter predictionPython/HTMLGupta et al., 2018 [94]
EffectorPhttps://effectorp.csiro.au/Predict fungal and oomycete effectors and classify localizationNaïve Bayes + C4.5 Decision Trees (Ensemble)Sperschneider et al., 2021 [95]
DeepGOhttps://deepgo.cbrc.kaust.edu.sa/deepgo/Predicts protein function to prioritize S-gene candidatesConvolutional Neural Networks (CNNs) and Deep Neural Networks (DNN)Maxat et al., 2018 [96]
GeneMANIAhttp://genemania.orgPredict gene function using network topology, co-expression, and orthologous featuresLabel propagation and adaptive network weightingWarde-Farley et al., 2010 [97]
BLAST https://blast.ncbi.nlm.nih.gov/Blast.cgiS-gene (homologous) identificationAlignment-basedAltschul et al., 1990 [98]
InterProScan https://www.ebi.ac.uk/interpro/search/sequence/ Identification of S-genes based on protein domains or families Rule-based domain annotationBlum et al., 2025 [99]
Effhunter https://github.com/GisCarreon/EffHunter_v.1.0Fungal effector predictionRule-driven pipelineCarreón-Anguiano et al., 2020 [100]
Pathoplanthttps://ngdc.cncb.ac.cn/databasecommons/database/id/1500Reference for Plant-pathogen interaction Relational DatabaseBülow et al., 2004 [101]
InterSPPI-AraPathogen2.0 http://zzdlab.com/intersppi/arapathogen/Predict Arabidopsis-pathogen interactionXGBoost (Extreme Gradient Boosting)Lei et al., 2023 [102]
HPIDB3.0 https://cales.arizona.edu/hpidb/Host–pathogen protein InteractionRelational DatabaseAmmari et al., 2016 [103]
PHIbasehttp://www.phi-base.org/Host–pathogen interaction databaseCurated databaseUrban et al. 2022 [104]
Phytozomehttps://phytozome-next.jgi.doe.govPredict gene families and evolutionary relationshipsDistance- and rule-based comparative genomics databaseGoodstein et al., 2012 [105]
Plant Ensemblhttps://plants.ensembl.org/Gene sequences and orthologsRest & Perl APIBolser et al., 2017 [106]
TAIR (The Arabidopsis Information Resource)https://www.arabidopsis.orgReference for model species to identify orthologsCurated resourcesReiser et al., 2024 [107]
Plant Resistance Gene Database (PRGdb)http://www.prgdb.org/prgdb4/Predicts primarily R-genes, but useful for cross-referencing S-gene candidatesDomain scoring and rule-based thresholdsGarcía et al., 2021 [84]
OrthoMCLhttps://orthomcl.orgGene family clustering and ortholog identificationMarkov Cluster Algorithm (MCL)Li et al., 2003 [108]
Gene Expression Omnibus (GEO)https://www.ncbi.nlm.nih.gov/geo/Identify differentially expressed genes as S-gene candidatesExpression repositoryBarrett et al., 2013 [109]
Table 2. List of S-gene editing events in various crops.
Table 2. List of S-gene editing events in various crops.
CropS-GeneDiseaseAssociated PathogenFunction of S-GeneResult/OutcomeReference
Arabidopsis thalianaelF(iso)4EViral diseaseYellow mosaic virus (YMV)Recessive resistance alleles against potyvirusesInduced mutation in elF(iso)4E imparts complete resistancePyott et al., 2016 [173]
Banana (Musa acuminate)MusaDMR6Xanthomonas wiltXanthomonas campestris pv. musacearumSalicylic acid degradation (SA-5-hydroxylase), suppresses host immunityImproved resistance with no detrimental impact on plantTripathi et al., 2021 [215]
Cassava (Manihot esculenta)nCBP1 and nCBP2Viral resistanceCassava brown streak virus (CBSV)Susceptibility factorResilience to viral diseaseGomez et al., 2019 [180]
Cucumber (Cucumis sativus L.)CsaMLO1, CsaMLO8, CsaMLO11Powdery mildew (PM)Podosphaera xanthiiNegative regulator of pre- (CsaMLO8) and post-invasive (CsaMLO1, CsaMLO11) defenseCsaMLO8 loss-of-function conferred highest penetration resistance; with CsaMLO1 and CsaMLO11 double mutations seemed good candidates for HR-based resistance against PM pathogenTek et al., 2022 [187], Ma et al., 2024 [216]
elF4EViral diseasesPotyvirusesEncodes a translation initiation factor that interacts with potyviral VPg proteins for viral infection.Loss-of-function mutations disrupt viral replication, conferring resistance. Homozygous elF4E_1DEL and elF4E_1-3DEL mutants showed complete resistance to watermelon mosaic virus (WMV), papaya ringspot virus (PRSV), and zucchini yellow mosaic virus (ZYMV)Chandrasekaran et al., 2016 [172]
Fidan et al., 2023 [184]
Grapevine (Vitis vinifera)VvDMR6-1 and VvDMR6-2Downy mildew (DM)Plasmopara viticolaNegative regulator of plant immunityReduced severity, simultaneous editing of both genes is required for reduced susceptibilityGiacomelli et al., 2023 [217]
Djennane et al., 2023 [218]
Melon (Cucumis melo)elF4EViral diseaseRingspot mosaic virus-W (RMV-W)Encodes a cap-binding protein essential for translation initiation. It acts as a proviral factor by facilitating viral RNA translation and in sexual developmentLoss-of-function mutations confer virus resistance but also cause male sterilityShirazi et al., 2023 [182]
Pechar et al., 2022 [183]
PrvViral diseasePRSV-WNLR resistance gene required for PRSV immunityKnockout mutants lost PRSV resistance; one allele (prvΔ154) exhibited an autoimmune dwarf phenotype suppressed at high temperaturesNizan et al., 2023 [219]
Potato (Solanum tuberosum)StNRL1Late blightPhytophthora infestansForms a protein complex with the P. infestans effector, leading to degradation of SWAP70, a positive regulator of cell deathKnockdown enhanced resistance to P. infestans but increased susceptibility to Alternaria alternata, suggesting a dual role in defenseNorouzi et al., 2024 [220]
eIF4EVirus resistancePotato virus Y (PVY)Translation initiation factor required by PVY via VPg interactionKnockout of eIF4E conferred broad-spectrum resistance to PVY with significantly delayed/reduced virus titer, and no adverse growth or developmental abnormalitiesNoureen et al., 2022 [181]
StPM1Late blightPhytophthora infestansEncodes a plasma membrane protein that interacts with StRbohC, promoting its degradation and negatively regulating reactive oxygen species (ROS) productionKnockout mutants exhibited enhanced resistance to P. infestans without growth penalties; increased expression of defense-related genes.Bi et al., 2024 [213]
Rapeseed (Brassica napus)BnHva22cStem stripingVerticillium longisporumSusceptibility factorImproved resistanceYe et al., 2024 [221]
BnaA05.RLK902Stem rot disease (SRD), grey mold disease (GMD)Sclerotinia sclerotiorum, Botrytis cineraPlasma membrane RLK negatively regulating necrotrophic immunityResistance to both diseases; no growth trade-offZhao et al., 2024 [222]
BnWRKY70Sclerotinia stem rotSclerotinia sclerotiorumTranscription factor; negatively regulates defense response against S. sclerotiorumKnockouts showed enhanced resistance; overexpression increased susceptibilitySun et al., 2018
[33]
Sweet basil (Ocimum basilicum)ObDMR6DMHyaloperonospora arabidopsidisSA-5-hydroxylase homolog that degrades salicylic acid, lowering host immunity and promoting pathogen growthKnockout of ObDMR6 led to enhanced resistance with reduced sporangia and pathogen biomassHasley et al., 2021 [223]
Tomato (Solanum lycopersicum)SlMlo1PMOidium neolycopersiciSusceptibility factor for fungal PMKnockout of SlMlo1 conferred complete resistance to PMPramanik et al., 2021 [192]
SlPMR4PMOidium neolycopersici (On)Encodes callose synthase at fungal penetration site, which is exploited by pathogenDisrupting SlPMR4 reduces susceptibility by increasing HRSantillán Martínez et al., 2020 [205]
SlJAZ2Bacterial speckPseudomonas syringae pv. Tomato (Pto) DC300Encodes coronatine co-receptor in stomatal guard cell that facilitates pathogen colonizationKnockout resulted in increased resistanceOrtigosa et al., 2019 [224]
SlNRX1Bacterial speckAlternaria brassicicola and Pseudomonas syringae pv. maculicolaMutation boosting the salicylic pathway for immunityEnhances plant immunity by negatively modulating the expression of the geneCha et al., 2023
[209]
miR482b, miR482cLate blightPhytophthora infestansRepress NBS-LRR defense gene transcriptsCRISPR knockout of miR482b + miR482c conferred resistance to P. infestans. Double mutants showed stronger resistance than miR482b aloneHong et al., 2021 [225]
eIF4E1Virus resistancePepper mottle virus (PepMoV)Translation initiation factor (eIF4E1) is hijacked by potyviruses via VPg interaction to facilitate viral translationKnockout conferred strong resistance to PepMoV, with no resistance to tobacco etch virus (TEV) and no adverse growth defectsAtarashi et al., 2020 [177]
Yoon et al., 2020 [178]
SlDMR6-1Resistance to bacterial, oomycete, and fungal pathogensPseudomonas syringae, Xanthomonas gardneri, Xanthomonas perforans, Phytophthora capsici, Pseudoidium neolycopersiciEncodes a 2-oxoglutarate Fe (II)-dependent dioxygenase that acts as a salicylic acid (SA) 5-hydroxylase, converting SA to 2,5-dihydroxybenzoic acidKnockout of the tomato susceptibility gene SlDMR6-1 significantly enhances resistance against a broad range of pathogens, including bacteria, oomycetes, and fungiThomazella et al., 2021 [226]
eIF4E2Virus resistancePepper veinal mottle virus (PVMV)Loss-of-function prevents PVMV from hijacking the cap-binding complexKnockout of eIF4E2 conferred resistance to PVMVKuroiwa et al., 2022 [179]
TPL1, TPL2Fusarium wiltFusarium oxysporumEncode transcriptional co-repressors that interact with the fungal SIX8 effector, promoting disease susceptibilityTPL1 knockout reduced susceptibility; TPL1/TPL2 double knockout provided higher resistanceAadlers et al., 2023 [227]
Rice (Oryza sativa)OsSWEET14, Os SWEET11, OsSWEET13Bacterial leaf blightXanthomonas oryzae pv. oryzae (Xoo)Encodes a sugar transporter that is used by Xoo TALE (transcription-ativator-like effectors) (e.g., AvrXa7, PthXo3/2, TalC, Tal5) for nutrient acquisition, promoting pathogen virulenceMutations in TALE-binding elements (EBEs) prevent pathogen-induced expression and confer resistanceBlanvillain-Baufume et al., 2016 [228]
Zhou et al., 2015 [229]
Olivia et al., 2019 [195]
Zafer et al., 2020 [197]
Aji et al., 2025 [199]; Li et al., 2025 [200]
EBETal6b of OsSWEET11aBacterial blightXanthomonas oryzae pv. oryzaeSugar transporterRapid resistance response that blocked disease developmentXu et al., 2024
[230]
RBL1Δ12Rice blastMagnaporthe oryzaeAssociated with effector secretion and fungal infectionConfers broad-spectrum disease resistanceSha et al., 2023
[231]
OsHPP04Root-knot nematodeMeloidogyne graminicolaAct as a negative regulator of host immunity against rice root-knot nematode (Meloidogyne graminicola)Enhance resistance with no adverse effect on main agronomic traitsHuang et al., 2023 [214]
Song et al., 2021 [232]
Pi21 and Bsr-d1Rice blastMagnaporthe oryzaeSuppresses basal defense mechanismsThe triple mutant had much higher resistance to both M. oryzae and Xoo than the single mutantsTao et al., 2021
[233]
Xa5Bacterial blightXanthomonas oryzae pv. oryzaeSusceptibility factor for diseaseTriple mutant with higher resistanceTao et al., 2021
[233]
eIF4GRTD (Rice tungro disease)Rice tungro spherical virus (RTSV)Translation initiation factor exploited by viral RNA for protein synthesis and infectionResistance to RTSV without growth penaltyMacovei et al., 2018 [174]
Cao et al., 2020 [74]
OsERF922Rice blastMagnaporthe oryzaeEncodes an ERF transcription factor that negatively regulates plant defenseReduced blast lesion formation without affecting agronomic traits; heritable resistance in T1 and T2 generationsWang et al., 2016 [234]
Wanjincheng orange (Citrus sinensis Osbeck)CsLOB1Citrus cankerZucchini yellow mosaic virus (ZYMV)Encodes a transcription factor activated by Xcc effector PthA4, promoting disease developmentPromoter editing of CsLOB1 enhanced resistance; no symptoms in some mutantsPeng et al., 2017 [235]
Watermelon (Citrullus lanatus)Clpsk1Fusarium wiltFusarium oxysporum f. sp. Niveum (FON)Encodes the precursor of phytosulfokine (PSK), a peptide hormone that negatively regulates plant immunityLoss-of-function mutations enhance resistance to FONZhang et al., 2020 [236]
CleIF4E1Viral diseaseZYMV, Cucumber green mottled mosaic virus (CGMMV)Major recessive factors for many viruses (especially potyviruses)Mutant line exhibited resistance to ZYMV but not CGMMV, with developmental defects and reduced yieldLi et al., 2024
[185]
Wheat (Triticum aestivum)TaMLO-A1, TaMLO-B1, TaMLO-D1PMBlumeria graminis f. sp. tritici (Bgt)MLO proteins negatively regulate defenseTriple-mutant plants exhibited heritable, broad-spectrum resistance to PMLi et al., 2022
[188]
SlMlo1PMBlumeria graminis f. sp. tritici (Bgt)MLO proteins negatively regulate defenseComplete resistance to PMNekrasov et al. 2017 [191]
Pramanik et al., 2021 [192]
TaEDR1 (homoeo alleles in A, B, and D subgenomes)PMBlumeria graminis f. sp. tritici (Bgt)Negative regulator of PM resistanceTriple-mutant Taedr1 wheat plants showed resistance to powdery mildew with no off-target mutations or pleiotropic effectsZhang et al., 2017 [203]
TaWRKY19Stripe rustPuccinia striiformis f. sp. tritici (Pst)WRKY transcription factor; negative regulator of plant immune responseKnockout resulted in strong resistance to stripe rustWang et al., 2022 [237]
TaGW2Leaf rustPuccinia triticina Eriksson (Pt)E3 ubiquitin ligase; negative regulator of wheat grain width and weightKnockout led to resistance to leaf rust with increased grain width and weightLiu et al., 2024 [238]
TaPslPK1Stripe rustPuccinia striiformis f. sp. tritici (Pst)Encodes a receptor-like cytoplasmic kinase targeted by the fungal effector PsSpg1, promoting virulence by phosphorylating TaCBF1d and modulating gene expressionKnockout of TaPsIPK1 conferred broad-spectrum resistance against Pst without affecting agronomic traits in field testsWang et al., 2022 [237]
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

Thakur, S.; Kaur, S.; Adhikari, S.; Sabharwal, P.; Fu, Y.; Meru, G. Turning Susceptibility into Strength: A New Era of Durable Resistance in Plants Through Genome Editing. Plants 2025, 14, 3080. https://doi.org/10.3390/plants14193080

AMA Style

Thakur S, Kaur S, Adhikari S, Sabharwal P, Fu Y, Meru G. Turning Susceptibility into Strength: A New Era of Durable Resistance in Plants Through Genome Editing. Plants. 2025; 14(19):3080. https://doi.org/10.3390/plants14193080

Chicago/Turabian Style

Thakur, Shallu, Simranjot Kaur, Sudeep Adhikari, Prerna Sabharwal, Yuqing Fu, and Geoffrey Meru. 2025. "Turning Susceptibility into Strength: A New Era of Durable Resistance in Plants Through Genome Editing" Plants 14, no. 19: 3080. https://doi.org/10.3390/plants14193080

APA Style

Thakur, S., Kaur, S., Adhikari, S., Sabharwal, P., Fu, Y., & Meru, G. (2025). Turning Susceptibility into Strength: A New Era of Durable Resistance in Plants Through Genome Editing. Plants, 14(19), 3080. https://doi.org/10.3390/plants14193080

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