Open access peer-reviewed chapter

Disease Resistance and Susceptibility Genes to Bacterial Blight of Rice

Written By

Tariq Mahmood and Frank F. White

Submitted: 12 November 2018 Reviewed: 29 March 2019 Published: 31 May 2019

DOI: 10.5772/intechopen.86126

From the Edited Volume

Protecting Rice Grains in the Post-Genomic Era

Edited by Yulin Jia

Chapter metrics overview

1,635 Chapter Downloads

View Full Metrics

Abstract

Rice (Oryza sativa L.) is a valuable resource for understanding the complex processes controlling yield and value-added traits. Bacterial blight (BB) is a vascular disease of rice, caused by strains of Xanthomonas oryzae pv. oryzae (Xoo) and provides insight, both practical and basic, into the concepts of susceptibility and resistance. Basic knowledge has been empirically and, more recently, intentionally exploited for broad and durable resistance to the disease. Bacterial blight involves representatives of most classes of resistance genes (R genes) and pathways for basal plant immunity. The study of BB also revealed novelties not observed in other models, possibly due to the long history of rice cultivation and the constant disease pressure. Conspicuous are the recessive R genes that target the notorious type III Transcription Activator-like effectors (TALes) of Xoo. Results indicate that pathogen and host are currently in a battle over a small patch of ground involving TALes function. At the same time, analyses of rice disease physiology are adding to a growing body of knowledge for plant disease processes and to how these processes are intertwined with disease susceptibility. The basic processes of BB present rich targets for the rapid advances in genome editing.

Keywords

  • Xanthomonas oryzae pv. oryzae
  • rice
  • recessive resistance
  • TAL effector
  • genome editing
  • CRISPR

1. Introduction

World population is expected to rise beyond 9 billion by 2050 [1]. Rice (Oryza sativa) is a staple food crop world-wide, providing about one fifth of the calories consumed by humans [2]. In particular, rice accounts for 35–75% of the calories consumed by more than 3 billion in Asian countries alone and planted on approximately 154 million hectares land annually [3]. Crop protection and food security go hand in hand, and breeding for resistance against crop diseases remains the essential ingredient for food security. Due to the labor-intensive nature of breeding, integrated disease control is often reduced to mere chemical control, leaving the very purpose of this environment-friendly approach in limbo. Advances in molecular tools in crop breeding, however, makes breeding an increasing sustainable effort in staying ahead of pathogen adaptation [4]. Bacterial blight (BB) of rice is a widespread vascular disease caused by Xanthomonas oryzae pv. oryzae(Xoo). Epidemics can severely reduce grain yield due to collapse of the entire crop [5]. BB was first characterized in the late nineteenth century [6]. Introduction of resistance (R) genes into rice cultivars is considered as the best option for Xoo management. A total of 42 R genes have been identified in rice against Xoo, and the number continues to grow [7, 8, 9]. Due to co-evolution and selection pressure between Xoo and rice, these R genes are selective in their efficiency against specific Xoo strains or races, which are sets of strains that share incompatibility on defined sets of R genes [10].

Advertisement

2. Post genomic era and rice grain protection

Advancements in genomics, referring here to DNA and RNA analyses, is as beneficial to crop protection as is to other discipline of biology. Rice MetaSysB, an open source which provides detailed information about BB-responsive genes, is based on the global expression analysis. The database provided 7475 unique genes and 5375 simple sequence repeats, which were responsive to Xoo in rice [11]. Such information is based on the compatible and incompatible rice-Xoo interactions. In another example, 454 and 498 differentially expressed genes were reported as exemplified by the incompatible and compatible rice-Xoo interactions, respectively, using cDNA microarray [12]. Genomics also provides functional information of genes up- and downstream of candidate resistance genes in the defense signal pathway, as is done in near-isogenic rice lines introgressed with Xa39, an as yet uncharacterized BB resistance gene [13].

Multiple rice and Xoo genomes have been sequenced, either in draft or complete form [14, 15, 16, 17, 18, 19, 20, 21, 22, 23], paving the way to identify functional connections between host and pathogen genes. The functional validation of the candidate genes is helping develop new rice varieties by introduction of the gene of interest through traditional breeding, marker assisted breeding, or genetic engineering approaches [3]. BB disease resistance is overcome by the emergence of more virulent strains of Xoo. Whole genome sequencing of 100 Xoo strains from India revealed that these strains were distinct from African and US Xoo strains [24]. Based on the reaction towards ten major resistance genes of rice, 46 out of the 100 strains were grouped into 11 pathotypes [24].

Advertisement

3. The genetic context of rice-Xoo interaction

Many BB-resistance genes in modern rice germplasm were selected long before the concepts of modern plant breeding were established, and a rich assortment of major dominant and recessive R genes has been identified by genetic and molecular studies (Table 1).

Gene Class Comments Cognate elicitor/effector Ref
Xa21 RLK1 extracellular, membrane and intracellular domains; kinase; broad resistance RaxX [25, 27]
Xa26 RLK similar to Xa21; same locus as Xa3; broad resistance Unknown [26]
Xa1, Xo1 NBS-LRR2 cytoplasm; narrow resistance Multiple TALes [31, 32, 33]
Xa4 WAK3 narrow unknown [40]
Xa27, Xa23, Xa10 TAL effector inducible membrane and cell wall; novel protein; broad resistance AvrXa27, AvrXa23, AvrXa10 [37, 38, 39]
xa5 Missense mutant of TFIIAγ5; small subunit of TFIIA transcription factor complex nuclear; broad resistance TALe interference [51, 53, 54]
xa13 promoter mutants of OsSWEET11; nodulin 3 family membrane; unresponsive to PthXo1 PthXo1 [42, 47]
xa25, OsSWEET13Kit promoter mutant of OsSWEET13, nodulin 3 family TATA box polymorphisms; unresponsive to PthXo2 PthXo2 [44, 52]

Table 1.

Cloned R genes to bacterial blight of rice.

RLK, receptor linked kinase.


NBS-LRR, nucleotide binding site, leucine-rich repeat.


WAK, wall-associated kinase.


Perhaps the best known of these genes, Xa21 represents the receptor kinase (RLK) class of R genes. Xa21 was originally introgressed into rice from the related species O. longistaminata and confers resistance to a broad range of Xoo strains [25]. Xa26, another cloned member of RLK gene family, also confers broad resistance with a somewhat different strain profile [26]. The cognate elicitor for Xa21 has been reported [27]. However, for Xa26 has not been identified.

RLKs play a central role in disease immunity pathways in plants, largely via the characterization of the bacterial flagellin receptor FLS2 and the related receptor EFR in Arabidopsis [28, 29]. A typical RLK consists of an extracellular receptor domain comprising of leucine-rich repeats (LRRs), a transmembrane domain, and an intracellular kinase domain [30]. As a class, RLKs have great potential for enhancing resistance to BB in rice and in other disease complexes of crop plants Xa21, Xa26, and other RLKs represent genetic components of the pathogen-associated molecular patterns (PAMPs)-triggered immunity (PTI) surveillance pathway in rice. Improvements in the rationale design of RLK receptor specificities, and screening for novel genes in germplasm or wild relatives could lead to general application for broad and durable resistance.

The nucleotide binding site-LRR (NBS-LRR) is another large class of R gene, represented in rice toward Xoo by Xa1 and Xo1 [31, 32, 33]. XA1 and XO1 recognize multiple TALe, and Xoo strains have adapted TALes, the so-called iTALes, that are truncated and inhibit the function of XA1 and XO1 [32, 34].

Specific TALe-dependent R genes governing dominant resistance in rice against Xoo are known as executor (E) genes. E genes are distinct from classical R genes, whose transcriptional activation by TALes of Xoo trigger immunity, leading to dominant resistance [35]. Xa27 represents the E genes class of dominant R genes and confers broad resistance to BB in rice [36]. Although not expressed in susceptible host, Xa27 is expressed only upon inoculation with Xoo strains harboring the TALe gene avrXa27 [37]. The protein is localized to apoplastic space, cell membrane and cell wall, and when expressed under a pathogen-nonspecific inducible rice OsPR1 promoter, conferred constitutive resistance to both compatible and incompatible strains alike [37]. The rice R genes Xa10 and Xa23 have similar requirements for the transcription activation domain and nuclear localization sequence (NLS) motifs of the corresponding TALes for their induction [38, 39].

Xa4 is the latest and, again, an unusual R gene of rice to be characterized. The protein is a wall-associated kinase (WAK) and provides attributes other than enhanced resistance. Rice plants with XA4 are shorter and stiffer in comparison to plants lacking the gene [40]. Xa4 is race-specific, meaning many strains of Xoo are compatible on plants with Xa4. How Xa4 functions in resistance is unknown at present.

3.1 SWEET genes and recessive resistance

A class of major TALe-dependent susceptibility (S) genes for BB in rice encodes sugar transporters, thereby named as SWEET gene family [41]. Specific TALes, referred to as major TALes, transcriptionally activate the corresponding SWEET genes in rice during infection to promote the disease in a gene-for-gene susceptibility manner [42]. Although at least five SWEET genes of the clade III members can function as an S gene in BB, only three members are known to be targeted by extant strains of Xoo [42, 43, 44, 45, 46, 47]. A member of the SWEET gene family, OsSWEET14, is induced by multiple distinct TALes, which include AvrXa7, PthXo3, Tal5 and TalC and are present in strains of different geographic origins and genetic lineages [43, 45, 46]. Similarly, PthXo2 drives OsSWEET13 expression in the susceptible rice variety IR24 [44], and OsSWEET11 is induced by the cognate PthXo1 [42]. The typical TALe possesses a central repetitive domain, a nuclear localization signal domain, and a transcription activation domain. The repetitive domain is responsible for binding of the TALe to a sequence motif called the effector binding element (EBE), which is commonly located in the promoter region of the respective S gene.

Mutated S gene alleles are proposed to be potentially more durable than dominant R genes [48, 49]. Identifying the promoter variant alleles of major S genes has been proposed in breeding for BB resistance [42, 47, 50, 51, 52, 53]. Recessive resistance is due to the cognate TALe cannot bind to the promoter variants of the S gene. The gene xa13, for example, is a recessive resistance insertion allele of 14.8 kb DNA fragment in the promoter of OsSWEET11 [42, 47]. OsSWEET11 encodes a protein related to MtN3 encoding nodulin 3 (N3) protein of Medicago truncatula. The gene was originally named Os8N3 due to its location on rice chromosome 8 and the similarity to MtN3 [42]. The critical difference between resistant (xa13/xa13) and susceptible plants is the elevated expression of OsSWEET11 during infection in otherwise susceptible plant genotypes [42]. RNAi-mediated silencing of OsSWEET11 plants was similarly resistant to Xoo strains that are solely dependent on PthXo1 for SWEET induction. Silenced plants, but not promoter variants, showed low pollen viability, corroborating the fact that Xoo hijacked otherwise developmentally important genes in rice for pathogenicity [42, 47]. Similarly, the TALe PthXo2 cannot bind to the EBE of xa25, a recessive allele of OsSWEET13, or the EBE region of OsSWEET13 in japonica rice cultivars, owing to single nucleotide polymorphisms in the respective EBEs [51, 52].

The gene xa13 is a naturally occurring allele, actually a series of alleles that protects the plant from a genetic disease vulnerability in the plant developmental pathways [42, 47]. However, xa13 is not a broad resistance provided in comparison to Xa21, Xa27 and xa5), and many strains from China, Philippines, Japan and Korea are compatible on xa13 lines [51]. Compatibility is derived by acquisition of major TALes that target alternative SWEET promoters [43]. As yet, not major TALe has been identified that replaces PthXo1 for OsSWEET11 expression.

The gene xa5 also affects TALe-dependent function but does not act at a specific SWEET gene. The recessive allele encodes a variant of the γ or small subunit of the transcription factor TFIIA [54, 55], which confers broad resistance. The gene differs from the susceptible allele by a single codon substitution of valine at position 39 to glutamic acid. TFIIA, consists of α, β, and γ subunits, and is involved in stabilizing the binding of the TATA box binding protein complex (TFIID) to the TATA box of gene promoters. The TFIIA components are highly conserved across the eukaryotes. Rice has two loci for TFIIAγ-one gene is on chromosome 5 (TFIIAγ5, xa5) and another on chromosome 1 (TFIIAγ1) [54]. The proteins are closely related but not identical. xa5 provides broad BB resistance and functions in inhibiting TALe function [51, 56]. However, xa5 is not effective against strains with the TALe PthXo1 [51].

Perhaps not all SWEET S genes are known or are not always induced in disease by Xoo. The Indian strain IX-80 was virulent but did not induce any known SWEET gene [57], suggesting an adaptation by the Xoo to relieve dependency on SWEET gene family. On the other hand, IX-80 remains TALe-dependent as the strain was not compatible on IR53 (xa13/xa13, xa5/xa5), a gene combination that blocks the xa5-compatible PthXo1 and all other major TALes at OsSWEET14 and OsSWEET14 [51].

Advertisement

4. Implication of interactions between TALes and the corresponding host genes

Due to the large reservoir of TALes in each strain of Xoo and the diverse roles of TALes in pathogenesis, the BB of rice represents an excellent plant/pathogen system for studying the biology of TALes. The apparent reason for the broad activity of Xa27 and Xa23 is the presence of the cognate TALes avrXa27 and avrXa23 in a large number of strains from southeast Asia, including Korea, China, Japan and the Philippines [37, 39]. On the other hand, the loss of avrXa27, avrXa23, or avrXa10, for that matter, does not appear to have an apparent fitness cost to the pathogen, and populations of Xoo may lose avrXa27 if Xa27 is widely deployed [37, 38, 39]. AvrXa7 is an important virulence factor for some strains of Xoo, and strains with AvrXa7 are incompatible on rice lines harboring the Xa7. In this case, loss of avrXa7, which is a major TALe for OsSWEET14, may result in strains that are weakly virulent or, essentially, nonpathogenic, if no other SWEET inducing TALes are present [43, 58]. A variety of other TALe genes are present in Xoo populations that can restore full virulence to strains missing avrXa7 [59]. Evasion of Xa7-mediated resistance is possible by loss of the gene, rearrangement of the central repeats or recombination among different TALe genes [60, 61]. However, despite rapid adaptation of bacteria by genetic changes and gene flow, field studies in the Philippines indicated that deployment of Xa7 was durable in test plots for more than 10 years [62]. Therefore, strains may have other limitations due to geographical location or rice genotype. Nevertheless, pyramiding broadly effective R genes with cognate TALes that are wide-spread in the pathogen populations should provide a degree of broad and durable resistance.

In the case of xa13, induction of the dominant allele SWEET11 is mediated by the TALe PthXo1 [42]. However, strains of Xoo that solely rely on PthXo1 cannot induce xa13 allele, and rice homozygous for xa13 is symptomless. xa13-dependent recessive resistance is phenotypically and qualitatively different from resistance provided by the dominant R gene Xa7 [42, 63]. Quantitatively, however, resistance mediated by xa13 and Xa7 are approximately equal with respect to bacterial growth and lesion length [42, 58, 64]. Xa7 resistance is the result of the presence of the appropriate AvrXa7 in the pathogen and dominant, while xa13 resistance is dependent on the absence of an effective virulence factor and recessive. The mechanism of XA7 mediated resistance is as yet unknown.

Type III effectors, in general, are hypothesized to interfere with host defense and defense signaling mechanisms. Strains of Xoo have other type III effectors, differing from TALes, and, therefore, not entirely dependent on TALes for suppression of host defenses [65]. Xoo strains lacking major TALes are still capable of causing water-soaking, if syringe inoculated, which is in contrast to type III secretion system (Hypersensitive reaction/pathogenicity or Hrp) mutants. Hrp mutant strains are incapable of secreting any type III effectors, including TALes, and are virtually symptomless [66]. The mechanism by which SWEET transporters condition susceptibility is unknown. One hypothesis is that the transporters allow cells to leak sucrose, providing the pathogen with nutrients. SWEET function may interfere with normal plant defense functions or, possibly, allow transport of other nutrients or disease promoting compounds [41]. However, little empirical evidence for the nutrition model exists at present.

Sequencing of Xoo genomes has revealed the full complement of TALes is now known [17, 18, 19, 20, 21, 22, 23]. The individual TALe genes are distinguishable on the basis of the number of repeats in the central repetitive region and by polymorphisms within each repeat sequence, particularly, at the 12th and 13th codons. Strains of the Asian lineage contain upwards of 16–19 TALe genes in each genome [18]. The large numbers of TALe genes in these species may reflect the evolutionary investment in utilizing the TALes for virulence and are essential, to the ecological niche these bacteria occupy. The maintenance of a large repertoire of TALe genes may increase the frequency of recombination between, and diversity of TALecgenes within the pathogen population [60]. Pathogen may then adapt faster to the changing host genotypes as exemplified by the appearance of pthXo5, which avoids Xa7 recognition and appears to be a hybrid between avrXa7 and pthXo6 [61].

Not all TALE genes of Xoo, however, are just substrates for new major TALEs. Two other TALE genes from PXO99 strain of Xoo, in addition to pthXo1, contribute to virulence, known to elevate the expression of two host genes distinct from SWEET11. PthXo6 elevates the expression of OsTFX1, which contributes to approximately 35% of the disease [67]. Many strains induce OsTFX1. The gene pthXo7 of PXO99 elevates the expression of OsTFIIAγ1 and would appear to be an adaption to host genotypes containing the xa5 allele of TFIIAγ5 [67]. However, introduction of pthXo7 to other strains does not restore full virulence on xa5/xa5 plants and may provide only an incremental fitness benefit [67]. All Asian strains also carry a set of truncated TALes, the inhibitory or iTALes, which function to suppress Xa1-mediated resistance [32].

4.1 Executor R genes and super promoters

Xa10, Xa23 and Xa27 are representatives of the new class of E genes, so-named because the induction of these genes executes a response of programed cell death (PCD) in the host. Xa10 induced PCD in plant species rice and N. benthamiana, and mammalian HeLa cells [38]. No cognate S genes for AvrXa10, AvrXa23, or AvrXa27 in compatible host cultivars have been reported, though the presence of AvrXa27 and AvrXa23 in many extant strains of Xoo may portend either a defeated function or an unknown cryptic function in S gene expression. Nonetheless, E genes hold great potential for broad and durable resistance in rice against extant Xoo population. A super promoter consisting of multiple EBEs, corresponding to specific TALes in extant population of Xoo, have been constructed (Figure 1). [68, 69, 70]. Addition of multiple EBEs to a pathogen strain specific rice BB resistance gene makes it effective against additional strains of Xoo. The EBEs of TALes PthXo1, PthXo6 and Tal9a when conjugated to E gene Xa27, showed resistance against PXO99 and a derivative strain lacking AvrXa27 [68]. A similar scenario was accomplished using E gene Xa10 [69]. The study suggested that broad-spectrum and potentially durable resistance is possible by stable integration of an E gene engineered in a way to respond to multiple TALes from different strains or even different pathogens. Design of a super promoter, however, needs to be done carefully. Risk that an added EBE might coincidently contain a cis regulatory element could induce the E gene expression in response to particular stimuli and cause cell death without challenge by TALes. Amended promoters should be tested thoroughly before deployment.

Figure 1.

Xoo TALe-dependent resistance and susceptibility in BB of rice. (A) Schematic of typical TALe from Xoo and (B) five types of TALe interactions affecting outcome of Xoo and rice interaction.

4.2 Targeted genome regulation and editing

Central to TALe function is the discovery of the DNA recognition cipher of TALEs [71, 72]. The central domain of a TALe, also known as binding domain, consists of variable number of tandem repeats, each consisting 33–35 amino acid residues. The 12th and 13th amino acid residues (known as repeat variable di-residues, RVDs) of each repeat preferentially binds to the respective nucleotides in the EBEs of target gene, such that HD, NG, NI and NN bind to C, T, A, and G, respectively in the effector binding elements (EBEs) of the promoter of a target gene [71, 72, 73]. The TALe recognition code allowed custom-engineer of DNA binding domains, also called designer TALes (dTALes), with novel specificity to the user-chosen DNA sequences [74, 75, 76]. dTALes provide a useful tool box to transiently activate host genes of interest for their functional analysis and assess the associated effect on host phenotype and physiology during rice-Xoo interaction. TALENs are fusions between dTALes and the nuclease domain of restriction enzyme FokI [77, 78, 79, 80]. Other C-terminal domains have also been used [81]. Target site recognition and TALEN dimerization triggers a double-strand break (DSB) and generates small random insertions or deletions at the cleavage site, resulting in an edited sequence. CRISPR-Cas editing approaches have circumvented the need to construct dTALes and achieved wide general use, including editing of rice genes [82, 83, 84].

Advertisement

5. Prospects for engineered broad and durable resistance in rice to BB

Traditional resistance breeding has identified many useful R genes and introgressed the genes into elite cultivars. Further, development of molecular markers allows the pyramiding of multiple genes into single lines. The development of designer TALENs and CRISPR-Cas genome editing brings greater flexibility and rapidity to the development of resistant germplasm. A continuous provision of novel R genes in breeding programs is possible. Of course, the adoption and utility of different approaches is dependent on the regulatory climate. Introduction of novel or alien genes may be prohibitive in the foreseeable future. Classification of genome editing techniques will also vary depending on the individual country. In the rice system, our understanding allows numerous approaches for the enhancement of resistance beyond classical breeding. TALe biology, specifically, can be exploited (Figure 2). Least intrusive is targeted genome editing of S genes. OsSWEET14 is targeted by unrelated TALEs, AvrXa7, PthXo3, Tal5 and TalC from different Xoo strains and which in some cases overlap their EBEs [43, 45, 46]. OsSWEET14 was made unresponsive to TALEs AvrXa7 and Tal5, when their respective EBEs were mutated using TALENs in otherwise susceptible rice cv. Kitake [85, 86]. Thus, recessive resistance obtained by the genome editing of OsSWEET14 is expected to be broad and contribute to durability given the apparent few major TALes in the extant population. Future efforts will be to target all EBE/S gene combinations in single elite lines. Fusion of EBEs to a variety of R and E genes has been demonstrated to provide resistance [68, 87]. The functional specificity of an E gene can be broadened by linkage to general inducible defense genes [69, 88]. Approaches are not limited to TALe-associated responses. The RLK immunity receptor EFR from Arabidopsis [89, 90], as well as XA21/EFR fusion proteins function in rice [91]. Thus, the sky is the limit for the engineering of broad and durable resistance in rice to BB.

Figure 2.

Super promoters: pyramiding of EBEs of multiple TALEs upstream of an E gene for broad and durable resistance. Subscript of each EBE corresponds to the respective TALEs. Blocks under each EBE represent the respective TALes with blunt ends as their N termini, and arrowheads as their C-termini flanking the binding repeats in center.

References

  1. 1. Béné C et al. Feeding 9 billion by 2050—Putting fish back on the menu. Food Security. 2015;7(2):261-274
  2. 2. Kennedy G, Burlingame B. Analysis of food composition data on rice from a plant genetic resources perspective. Food Chemistry. 2003;80(4):589-596
  3. 3. Khush GS. What it will take to feed 5.0 billion rice consumers in 2030. Plant Molecular Biology. 2005;59(1):1-6
  4. 4. Collard BC, Mackill DJ. Marker-assisted selection: An approach for precision plant breeding in the twenty-first century. Philosophical Transactions of the Royal Society, B: Biological Sciences. 2007;363(1491):557-572
  5. 5. Mew T. Current status and future prospects of research on bacterial blight of rice. Annual Review of Phytopathology. 1987;25(1):359-382
  6. 6. Reddy A et al. Relationship of bacterial leaf blight severity to grain yield of rice. Phytopathology. 1979;69(9):967-969
  7. 7. Ou SH. Rice Diseases. IRRI; 1985
  8. 8. Kim SM et al. Identification and fine-mapping of a new resistance gene, Xa40, conferring resistance to bacterial blight races in rice (Oryza sativa L.). Theoretical and Applied Genetics. 2015;128(10):1933-1943
  9. 9. Busungu C et al. Identification and linkage analysis of a new rice bacterial blight resistance gene from XM14, a mutant line from IR24. Breeding Science. 2016;66(4):636-645
  10. 10. Mishra D et al. Pathotype and genetic diversity amongst Indian isolates of Xanthomonas oryzae pv. oryzae. PLoS ONE. 2013;8(11):e81996
  11. 11. Sureshkumar V et al. RiceMetaSysB: A database of blast and bacterial blight responsive genes in rice and its utilization in identifying key blast-resistant WRKY genes. In: Database. 2019
  12. 12. Li Q et al. Expression profiling of rice genes in early defense responses to blast and bacterial blight pathogens using cDNA microarray. Physiological and Molecular Plant Pathology. 2006;68(1-3):51-60
  13. 13. Zhang F et al. Comparative transcriptome profiling of a rice line carrying Xa39 and its parents triggered by Xanthomonas oryzae pv. oryzae provides novel insights into the broad-spectrum hypersensitive response. BMC Genomics. 2015;16:111
  14. 14. RGSP International. The map-based sequence of the rice genome. Nature. 2005;436(7052):793
  15. 15. Sun C et al. RPAN: Rice pan-genome browser for ∼3000 rice genomes. Nucleic Acids Research. 2017;45(2):597-605
  16. 16. Ochiai H et al. Genome sequence of Xanthomonas oryzae pv. oryzae suggests contribution of large numbers of effector genes and insertion sequences to its race diversity. Japan Agricultural Research Quarterly: JARQ. 2005;39(4):275-287
  17. 17. Lee B-M et al. The genome sequence of Xanthomonas oryzae pathovar oryzae KACC10331, the bacterial blight pathogen of rice. Nucleic Acids Research. 2005;33(2):577-586
  18. 18. Salzberg SL et al. Genome sequence and rapid evolution of the rice pathogen Xanthomonas oryzae pv. oryzae PXO99A. BMC Genomics. 2008;9:204
  19. 19. Booher NJ et al. Single molecule real-time sequencing of Xanthomonas oryzae genomes reveals a dynamic structure and complex TAL (transcription activator-like) effector gene relationships. Microbial genomics. 2015;1(4):1-22. DOI: 10.1099/mgen.0.000032
  20. 20. Huguet-Tapia JC et al. Complete genome sequence of the African Strain AXO1947 of Xanthomonas oryzae pv. oryzae. Genome Announcements. 2016;4(1). DOI: 10.1128/genomeA.01730-15
  21. 21. Quibod IL et al. Effector diversification contributes to Xanthomonas oryzae pv. oryzae phenotypic adaptation in a semi-isolated environment. Scientific Reports. 2016;6:34137. DOI: 10.1038/srep34137.
  22. 22. Doucouré H et al. Functional and genome sequence-driven characterization of tal effector gene repertoires reveals novel variants with altered specificities in closely related Malian Xanthomonas oryzae pv. oryzae strains. Frontiers in Microbiology. 2018;9:1657
  23. 23. Tran TT et al. Functional analysis of African Xanthomonas oryzae pv. oryzae TALomes reveals a new susceptibility gene in bacterial leaf blight of rice. PLoS Pathogens. 2018;14:1-25
  24. 24. Midha S et al. Population genomic insights into variation and evolution of Xanthomonas oryzae pv. oryzae. Scientific Reports. 2017;7:40694
  25. 25. Song WY et al. A receptor kinase-like protein encoded by the rice disease resistance gene, Xa21. Science. 1995;270(5243):1804-1806
  26. 26. Sun X et al. Xa26, a gene conferring resistance to Xanthomonas oryzae pv. oryzae in rice, encodes an LRR receptor kinase-like protein. The Plant Journal. 2004;37(4):517-527
  27. 27. Pruitt RN et al. The rice immune receptor XA21 recognizes a tyrosine-sulfated protein from a Gram-negative bacterium. Science Advances. 2015;1(6):e1500245
  28. 28. Gómez-Gómez L, Boller T. Flagellin perception: A paradigm for innate immunity. Trends in Plant Science. 2002;7(6):251-256
  29. 29. Zipfel C et al. Perception of the bacterial PAMP EF-Tu by the receptor EFR restricts Agrobacterium-mediated transformation. Cell. 2006;125(4):749-760
  30. 30. Shiu S-H, Bleecker AB. Plant receptor-like kinase gene family: Diversity, function, and signaling. Science's STKE. 2001;2001(113):re22
  31. 31. Yoshimura S et al. Expression of Xa1, a bacterial blight-resistance gene in rice, is induced by bacterial inoculation. Proceedings of the National Academy of Sciences. 1998;95(4):1663-1668
  32. 32. Ji Z et al. Interfering TAL effectors of Xanthomonas oryzae neutralize R-gene-mediated plant disease resistance. Nature Communications. 2016;7:13435. DOI: 10.1038/ncomms13435
  33. 33. Triplett LR et al. A resistance locus in the American heirloom rice variety Carolina Gold Select is triggered by TAL effectors with diverse predicted targets and is effective against African strains of Xanthomonas oryzae pv. oryzicola. The Plant Journal. 2016;87(5):472-483
  34. 34. Read AC et al. Suppression of Xo1-mediated disease resistance in rice by a truncated, non-dna-binding TAL effector of Xanthomonas oryzae. Frontiers in Plant Science. 2016;7(1516):eCollection
  35. 35. Zhang J, Yin Z, White F. TAL effectors and the executor geneR genes. Frontiers in Plant Science. 2015;6:641
  36. 36. Gu K et al. High-resolution genetic mapping of Xa27 (t), a new bacterial blight resistance gene in rice, Oryza sativa L. Theoretical and Applied Genetics. 2004;108(5):800-807
  37. 37. Gu K et al. geneR gene expression induced by a type-III effector triggers disease resistance in rice. Nature. 2005;435(7045):1122
  38. 38. Tian D et al. The rice TAL effector-dependent resistance protein XA10 triggers cell death and calcium depletion in the endoplasmic reticulum. The Plant Cell. 2014. DOI: 10.1105/tpc. 113.119255
  39. 39. Wang C et al. XA23 is an executor R protein and confers broad-spectrum disease resistance in rice. Molecular Plant. 2015;8(2):290-302
  40. 40. Hu K et al. Improvement of multiple agronomic traits by a disease resistance gene via cell wall reinforcement. Nature Plants. 2017;3:17009
  41. 41. Chen LQ et al. Sugar transporters for intercellular exchange and nutrition of pathogens. Nature. 2010;468(7323):527-532
  42. 42. Yang B, Sugio A, White FF. Os8N3 is a host disease-susceptibility gene for bacterial blight of rice. Proceedings of the National Academy of Sciences. 2006;103(27):10503-10508
  43. 43. Antony G et al. Rice xa13 recessive resistance to bacterial blight is defeated by induction of the disease susceptibility gene Os-11N3. Plant Cell. 2010;22(11):3864-3876
  44. 44. Zhou J et al. Gene targeting by the TAL effector PthXo2 reveals cryptic resistance gene for bacterial blight of rice. The Plant Journal. 2015;82(4):632-643
  45. 45. Streubel J et al. Five phylogenetically close rice SWEET genes confer TAL effector-mediated susceptibility to Xanthomonas oryzae pv. oryzae. The New Phytologist. 2013;200(3):808-819
  46. 46. Yu Y et al. Colonization of rice leaf blades by an African strain of Xanthomonas oryzae pv. oryzae depends on a new TAL effector that induces the rice nodulin-3 Os11N3 gene. Molecular Plant-Microbe Interactions. 2011;24(9):1102-1113
  47. 47. Chu Z et al. Promoter mutations of an essential gene for pollen development result in disease resistance in rice. Genes and Development. 2000;20(10):1250-1255
  48. 48. Gust AA, Brunner F, Nurnberger T. Biotechnological concepts for improving plant innate immunity. Current Opinion in Biotechnology. 2010;21(2):204-210
  49. 49. Iyer-Pascuzzi AS, McCouch SR. Recessive resistance genes and the Oryza sativa-Xanthomonas oryzae pv. oryzae pathosystem. Molecular Plant-Microbe Interactions. 2007;20(7):731-739
  50. 50. Hutin M et al. A knowledge-based molecular screen uncovers a broad-spectrum Os SWEET 14 resistance allele to bacterial blight from wild rice. The Plant Journal. 2015;84(4):694-703
  51. 51. Huang S et al. The broadly effective recessive resistance gene xa5 of rice is a virulence effector-dependent quantitative trait for bacterial blight. The Plant Journal. 2016;86(2):186-194
  52. 52. Liu Q et al. A paralog of the MtN3/salivafamily recessively confers race-specific resistance to Xanthomonas oryzae inrice. Plant, Cell and Environment. 2011;34(11):1958-1969
  53. 53. Zaka A et al. Natural variations in the promoter of OsSWEET13 and OsSWEET14 expand the range of resistance against Xanthomonas oryzae pv. oryzae. PLoS ONE. 2018;13(9):e0203711
  54. 54. Iyer AS, McCouch SR. The rice bacterial blight resistance gene xa5 encodes anovel form of disease resistance. Molecular Plant-Microbe Interactions. 2004;17(12):1348-1354
  55. 55. Jiang G-H et al. Testifying the rice bacterial blight resistance gene xa5 by genetic complementation and further analyzing xa5 (Xa5) in comparison with its homolog TFIIAγ1. Molecular genetics and Genomics. 2006;275(4):354-366
  56. 56. Yuan M et al. A host basal transcription factor is a key component for infection of rice by TALE-carrying bacteria. eLife. 2016;5
  57. 57. Carpenter SCD et al. A strain of an emerging Indian Xanthomonas oryzae pv. oryzae pathotype defeats the rice bacterial blight resistance gene xa13 without inducing a clade III SWEET gene and is nearly identical to a recent Thai isolate. Frontiers in Microbiology. 2018;9:2703
  58. 58. Bai J et al. Xanthomonas oryzae pv. oryzae avirulence genes contribute differently and specifically to pathogen aggressiveness. Molecular Plant-Microbe Interactions. 2000;13(12):1322-1329
  59. 59. Yang B, White FF. Diverse members of the AvrBs3/PthA family of type III effectors are major virulence determinants in bacterial blight disease of rice. Molecular Plant-Microbe Interactions. 2004;17(11):1192-1200
  60. 60. Yang Y, Gabriel DW. Intragenic recombination of a single plant pathogen gene provides a mechanism for the evolution of new host specificities. Journal of Bacteriology. 1995;177(17):4963-4968
  61. 61. Yang B, Sugio A, White FF. Avoidance of host recognition by alterations in the repetitive and C-terminal regions of AvrXa7, a type III effector of Xanthomonas oryzae pv. oryzae. Molecular Plant-Microbe Interactions. 2005;18(2):142-149
  62. 62. Cruz CMV et al. Predicting durability of a disease resistance gene based on an assessment of the fitness loss and epidemiological consequences of avirulence gene mutation. Proceedings of the National Academy of Sciences. 2000;97(25):13500-13505
  63. 63. Cao J et al. Dominant and recessive major geneR genes lead to different types of host cell death during resistance to Xanthomonas oryzae in rice. Frontiers in Plant Science. 2018;9:1711
  64. 64. Yang B et al. The virulence factor AvrXa7 of Xanthomonas oryzae pv. oryzae is a type III secretion pathway-dependent nuclear-localized double-stranded DNA-binding protein. Proceedings of the National Academy of Sciences. 2000;97(17):9807-9812
  65. 65. Song C, Yang B. Mutagenesis of 18 type III effectors reveals virulence function of XopZ(PXO99) in Xanthomonas oryzae pv. oryzae. Molecular Plant-Microbe Interactions. 2010;23(7):893-902
  66. 66. Zhu W, Magbanua MM, White FF. Identification of two novel hrp-associated genes in the hrp gene cluster of Xanthomonas oryzae pv. oryzae. Journal of Bacteriology. 2000;182(7):1844-1853
  67. 67. Sugio A et al. Two type III effector genes of Xanthomonas oryzae pv. oryzae control the induction of the host genes OsTFIIAγ1 and OsTFX1 during bacterial blight of rice. Proceedings of the National Academy of Sciences. 2007;104(25):10720-10725
  68. 68. Hummel AW, Doyle EL, Bogdanove AJ. Addition of transcription activator-like effector binding sites to a pathogen strain-specific rice bacterial blight resistance gene makes it effective against additional strains and against bacterial leaf streak. The New Phytologist. 2012;195(4):883-893
  69. 69. Zeng X et al. Genetic engineering of the Xa10 promoter for broad-spectrum and durable resistance to Xanthomonas oryzae pv. oryzae. Plant Biotechnology Journal. 2015;13(7):993-1001
  70. 70. Römer P, Recht S, Lahaye T. A single plant resistance gene promoter engineered to recognize multiple TAL effectors from disparate pathogens. Proceedings of the National Academy of Sciences. 2009;106(48):20526-20531
  71. 71. Boch J et al. Breaking the code of DNA binding specificity of TAL-type III effectors. Science. 2009;326(5959):1509-1512
  72. 72. Moscou MJ, Bogdanove AJ. A simple cipher governs DNA recognition by TAL effectors. Science. 2009;326(5959):1501
  73. 73. Romer P et al. Promoter elements of rice susceptibility genes are bound and activated by specific TAL effectors from the bacterial blight pathogen, Xanthomonas oryzae pv. oryzae. The New Phytologist. 2010;187(4):1048-1057
  74. 74. Morbitzer R et al. Regulation of selected genome loci using de novo-engineered transcription activator-like effector (TALE)-type transcription factors. Proceedings of the National Academy of Sciences of the United States of America. 2010;107(50):21617-21622
  75. 75. Zhang F et al. Programmable sequence-specific transcriptional regulation of mammalian genome using designer TAL effectors. Nature Biotechnology. 2011;29(2):149
  76. 76. Li T et al. Designer TAL effectors induce disease susceptibility and resistance to Xanthomonas oryzae pv. oryzae in rice. Molecular Plant. 2013;6(3):781-789
  77. 77. Li T et al. Modularly assembled designer TAL effector nucleases for targeted gene knockout and gene replacement in eukaryotes. Nucleic Acids Research. 2011;39(14):6315-6325
  78. 78. Christian M et al. Targeting DNA double-strand breaks with TAL effector nucleases. Genetics. 2010;186(2):757-761
  79. 79. Cermak T et al. Efficient design and assembly of custom TALEN and other TAL effector-based constructs for DNA targeting. Nucleic Acids Research. 2011;39(12):e82
  80. 80. Li T et al. TAL nucleases (TALNs): Hybrid proteins composed of TAL effectors and FokI DNA-cleavage domain. Nucleic Acids Research. 2011;39(1):359-372
  81. 81. Chen K, Gao C. Targeted genome modification technologies and their applications in crop improvements. Plant Cell Reports. 2014;33(4):575-583
  82. 82. Wiedenheft B, Sternberg SH, Doudna JA. RNA-guided genetic silencing systems in bacteria and archaea. Nature. 2012;482(7385):331
  83. 83. Jinek M et al. A programmable dual-RNA-guided DNA endonuclease in adaptive bacterial immunity. Science. 2012;337(6096):816-821
  84. 84. Hsu PD, Lander ES, Zhang F. Development and applications of CRISPR-Cas9 for genome engineering. Cell. 2014;157(6):1262-1278
  85. 85. Blanvillain-Baufumé S et al. Targeted promoter editing for rice resistance to Xanthomonas oryzae pv. oryzae reveals differential activities for SWEET14-inducing TAL effectors. Plant Biotechnology Journal. 2017;15(3):306-317
  86. 86. Li T et al. High-efficiency TALEN-based gene editing produces disease-resistant rice. Nature Biotechnology. 2012;30(5):390-392
  87. 87. Hutin et al. Ectopic activation of the rice NLR heteropair RGA4/RGA5 confers resistance to bacterial blight and bacterial leaf streak diseases. The Plant Journal. 2016;88(1):43-55
  88. 88. Tian D, Yin Z. Constitutive heterologous expression of avrXa27 in rice containing the R gene Xa27 confers enhanced resistance to compatible Xanthomonas oryzae strains. Molecular Plant Pathology. 2009;10(1):29-39
  89. 89. Schwessinger B et al. Correction: Transgenic expression of the dicotyledonous pattern recognition receptor EFR in rice leads to ligand-dependent activation of defense responses. PLoS Pathogens. 2015;11(4):e1004872
  90. 90. Lu F et al. Enhancement of innate immune system in monocot rice by transferring the dicotyledonous elongation factor Tu receptor EFR. Journal of Integrative Plant Biology. 2015;57(7):641-652
  91. 91. Thomas NC et al. The rice XA21 ectodomain fused to the Arabidopsis EFR cytoplasmic domain confers resistance to Xanthomonas oryzae pv. oryzae. PeerJ. 2018;6:e4456

Written By

Tariq Mahmood and Frank F. White

Submitted: 12 November 2018 Reviewed: 29 March 2019 Published: 31 May 2019