fungal and oomycete effectors – strategies to subdue a host · can. j. plant pathol. (2011),...

22
Can. J. Plant Pathol. (2011), 33(4): 425–446 Minireview/Minisynthèse Fungal and oomycete effectors – strategies to subdue a host SHAWKAT ALI 1,2 AND GUUS BAKKEREN 1 1 Agriculture and Agri-Food Canada, Pacific Agri-Food Research Center, Summerland, BC V0H 1Z0, Canada 2 Department of Botany, University of British Columbia, Vancouver, BC V6T 1Z4, Canada; current address: Horticulture R&D Center, Agriculture and Agri-Food Canada, St-Jean-sur-Richelieu Québec J3B 3E6, Canada (Accepted 4 september 2011) Abstract: Molecular studies focusing on the interface between microbes and plant hosts have provided major insights into the basis underlying pathogenesis, symbiosis and plant defence and resistance mechanisms. A more recent focus on microbes, facilitated by the generation of complete genome sequences, has uncovered the sheer number of protein effectors microbes deliver in this interface as well as inside host cells to manipulate the plant immune system. Although studies on the characterization and roles of bacterial effectors are further advanced, in this review we focus on the current knowledge of fungal and oomycete effectors and their roles. Examples are given of effectors disarming plant defence enzymes, such as the apoplastic effector AVR2 from Cladosporium fulvum which inhibits the tomato defence cysteine protease. Other effectors interfere with the perception by the host of microbes exposing molecular determinants such as Phytophthora infestans INF1 protein. Many effectors alter gene expression induced by the host during defence, exemplified in fungi by Ustilago maydis Pit2 suppressing maize defence genes. Effectors recognized by resistance gene products, either directly or indirectly, and eliciting defence, represent the classical avirulence genes and almost 50 have now been cloned from fungi and oomycetes. Evolutionary adaptations and arms races have produced diversification in both pathogen and host, and in pathogens, are the cause of breaking crop resistance in agricultural settings. Molecular insight provides valuable information for applications. For example, some effectors are crucial for pathogenesis, thereby revealing targets for disease control and others interact with host resistance gene products and could be used to screen germplasm for novel sources of disease resistance. Variation among effectors will likely yield diagnostic tools for pathogen race identification. The study of model systems is providing insight into avenues by which other, major plant diseases can potentially be controlled. Keywords: avirulence genes, disease resistance, effector proteins, microbe–plant interactions Résumé: Les études moléculaires ciblant l’interface entre les microorganismes et les plantes hôtes ont ouvert de nouvelles perspectives sur les bases sous-jacentes de la pathogenèse, de la symbiose ainsi que des mécanismes de défenses et de résistance. Une nouvelle approche des microorganismes, facilitée par la génération de séquences entières de génomes, a permis de découvrir l’importance du nombre de protéines effectrices que les microorganismes sécrètent dans cette interface ainsi que dans les cellules hôtes afin de modifier le système immunitaire de la plante. Bien que les études sur la caractérisation et les rôles des effecteurs microbiens soient avancées, dans cet exposé, nous mettons l’accent sur les connaissances actuelles relatives aux effecteurs fongiques et à ceux des oomycètes, ainsi que sur leurs rôles. On y présente des exemples d’effecteurs neutralisant les enzymes de défense de plantes, comme l’effecteur apoplastique AVR2 de Cladosporium fulvum qui inhibe chez la tomate les mécanismes de défense dépendant de la protéase à cystéine. D’autres effecteurs modifient la perception que l’hôte a de microorganismes exposant des déterminants moléculaires comme la protéine INF1 de Phytophthora infestans. Un grand nombre d’effecteurs altèrent l’expression du gène induite durant la phase de défense, phénomène illustré chez les champignons par la protéine Pit-2 d’Ustilago maydis qui inactive les gènes de défense du maïs. Les effecteurs reconnus, directement ou indirectement, par les produits géniques de résistance suscitant la défense sont les gènes classiques d’avirulence et presque 50 ont à ce jour été clonés à partir de champignons et d’oomycètes. Les adaptations évolutives et les « courses aux armements » ont induit la diversification tant chez l’hôte que chez les agents pathogènes et, chez ces derniers, elles sont la cause de l’effondrement de la résistance des cultures commerciales. Les perspectives moléculaires fournissent des données précieuses en vue d’applications utiles. Par exemple, certains effecteurs sont essentiels à la Correspondence to: G. Bakkeren. E-mail: [email protected] ISSN: 0706-0661 print/ISSN 1715-2992 online © 2011 The Canadian Phytopathological Society http://dx.doi.org/10.1080/07060661.2011.625448 Downloaded by [Canadian Agriculture Library, Agriculture and Agri-Food Canada] at 13:23 08 March 2012

Upload: others

Post on 05-Oct-2020

4 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Fungal and oomycete effectors – strategies to subdue a host · Can. J. Plant Pathol. (2011), 33(4): 425–446 Minireview/Minisynthèse Fungal and oomycete effectors – strategies

Can. J. Plant Pathol. (2011), 33(4): 425–446

Minireview/Minisynthèse

Fungal and oomycete effectors – strategies to subdue a host

SHAWKAT ALI1,2 AND GUUS BAKKEREN1

1Agriculture and Agri-Food Canada, Pacific Agri-Food Research Center, Summerland, BC V0H 1Z0, Canada2Department of Botany, University of British Columbia, Vancouver, BC V6T 1Z4, Canada; current address: Horticulture R&D Center,Agriculture and Agri-Food Canada, St-Jean-sur-Richelieu Québec J3B 3E6, Canada

(Accepted 4 september 2011)

Abstract: Molecular studies focusing on the interface between microbes and plant hosts have provided major insights into the basisunderlying pathogenesis, symbiosis and plant defence and resistance mechanisms. A more recent focus on microbes, facilitated by thegeneration of complete genome sequences, has uncovered the sheer number of protein effectors microbes deliver in this interface as well asinside host cells to manipulate the plant immune system. Although studies on the characterization and roles of bacterial effectors are furtheradvanced, in this review we focus on the current knowledge of fungal and oomycete effectors and their roles. Examples are given of effectorsdisarming plant defence enzymes, such as the apoplastic effector AVR2 from Cladosporium fulvum which inhibits the tomato defence cysteineprotease. Other effectors interfere with the perception by the host of microbes exposing molecular determinants such as Phytophthorainfestans INF1 protein. Many effectors alter gene expression induced by the host during defence, exemplified in fungi by Ustilago maydisPit2 suppressing maize defence genes. Effectors recognized by resistance gene products, either directly or indirectly, and eliciting defence,represent the classical avirulence genes and almost 50 have now been cloned from fungi and oomycetes. Evolutionary adaptations and armsraces have produced diversification in both pathogen and host, and in pathogens, are the cause of breaking crop resistance in agriculturalsettings. Molecular insight provides valuable information for applications. For example, some effectors are crucial for pathogenesis, therebyrevealing targets for disease control and others interact with host resistance gene products and could be used to screen germplasm for novelsources of disease resistance. Variation among effectors will likely yield diagnostic tools for pathogen race identification. The study of modelsystems is providing insight into avenues by which other, major plant diseases can potentially be controlled.

Keywords: avirulence genes, disease resistance, effector proteins, microbe–plant interactions

Résumé: Les études moléculaires ciblant l’interface entre les microorganismes et les plantes hôtes ont ouvert de nouvelles perspectives sur lesbases sous-jacentes de la pathogenèse, de la symbiose ainsi que des mécanismes de défenses et de résistance. Une nouvelle approche desmicroorganismes, facilitée par la génération de séquences entières de génomes, a permis de découvrir l’importance du nombre de protéineseffectrices que les microorganismes sécrètent dans cette interface ainsi que dans les cellules hôtes afin de modifier le système immunitaire dela plante. Bien que les études sur la caractérisation et les rôles des effecteurs microbiens soient avancées, dans cet exposé, nous mettonsl’accent sur les connaissances actuelles relatives aux effecteurs fongiques et à ceux des oomycètes, ainsi que sur leurs rôles. On y présente desexemples d’effecteurs neutralisant les enzymes de défense de plantes, comme l’effecteur apoplastique AVR2 de Cladosporium fulvum quiinhibe chez la tomate les mécanismes de défense dépendant de la protéase à cystéine. D’autres effecteurs modifient la perception que l’hôte ade microorganismes exposant des déterminants moléculaires comme la protéine INF1 de Phytophthora infestans. Un grand nombred’effecteurs altèrent l’expression du gène induite durant la phase de défense, phénomène illustré chez les champignons par la protéinePit-2 d’Ustilago maydis qui inactive les gènes de défense du maïs. Les effecteurs reconnus, directement ou indirectement, par les produitsgéniques de résistance suscitant la défense sont les gènes classiques d’avirulence et presque 50 ont à ce jour été clonés à partir dechampignons et d’oomycètes. Les adaptations évolutives et les « courses aux armements » ont induit la diversification tant chez l’hôte quechez les agents pathogènes et, chez ces derniers, elles sont la cause de l’effondrement de la résistance des cultures commerciales. Lesperspectives moléculaires fournissent des données précieuses en vue d’applications utiles. Par exemple, certains effecteurs sont essentiels à la

Correspondence to: G. Bakkeren. E-mail: [email protected]

ISSN: 0706-0661 print/ISSN 1715-2992 online © 2011 The Canadian Phytopathological Societyhttp://dx.doi.org/10.1080/07060661.2011.625448

Dow

nloa

ded

by [

Can

adia

n A

gric

ultu

re L

ibra

ry, A

gric

ultu

re a

nd A

gri-

Food

Can

ada]

at 1

3:23

08

Mar

ch 2

012

Page 2: Fungal and oomycete effectors – strategies to subdue a host · Can. J. Plant Pathol. (2011), 33(4): 425–446 Minireview/Minisynthèse Fungal and oomycete effectors – strategies

S. Ali and G. Bakkeren 426

pathogenèse, déterminant de ce fait des cibles de lutte contre les maladies des plantes; d’autres interagissent avec les produits géniques de larésistance de l’hôte et pourraient être utilisés pour analyser les germoplasmes afin d’y déceler de nouvelles sources de résistance aux maladies.La variation observée chez les effecteurs contribuera de toute évidence au développement d’outils de diagnostic permettant l’identification deraces pathogènes. L’étude de modèles ouvre de nouvelles voies grâce auxquelles d’autres maladies importantes pourront être traitées.

Mots clés: gènes d’avirulence, interactions microorganismes plantes, protéines effectrices, résistance aux maladies

Introduction

Among the myriad of microbes larger organismsencounter, there are a few that have adapted to over-come the many barriers and defence mechanisms of theseorganisms to become pathogens. The more tools suchpathogens have at their disposal, the more virulent theyare on a given host. Over the last 10 years, tremendousprogress has been made in the field of plant–microbeinteractions and the discovery of many pathogen and hostgenes involved in these interactions has provided someinsight into the molecular mechanisms that decide the out-come of such encounters i.e. plant disease or host defenceand resistance. Pathogens flood their hosts with numer-ous proteins and compounds to counteract the defenceand ensure proliferation. The host, on its part, resists andthe uncovering of the increasingly astonishing ways eachinteracting organism tries to decide the outcome of thisdelicate balance in their favour, promises novel strategiesto help plants boost their defence responses. Although thefocus of this paper is on fungal and oomycete effectors,many recent discoveries into their roles have emergedfrom work on bacterial systems, some of which will behighlighted because they might direct research on fungaland oomycete effectors.

Strategies used by pathogens to infect host plants

Pathogens use different strategies to enter their hostplants. Fungi and oomycetes enter either through nat-ural openings or directly through the plant epidermalcells by mechanical and chemical means, or expand theirhyphae on the top of, within or between the plant cells(Jones & Dangl, 2006). Pathogens need to adhere to theplant before penetrating the plant cuticle. Fungal hyphaeand spores use mucilaginous and adhesive substances attheir tips, and the intermolecular forces between plantand pathogen are responsible for the close contact. Mostrust fungi enter plants through stomata by developingappressoria over the stomata to penetrate into the cavitybelow, while ascomycetes such as the rice blast fungusMagnaporthe grisea and the powdery mildews, such asBlumeria graminis f. sp. hordei (Bgh), penetrate the cuti-cle of plants directly by means of an appressorium. For

direct penetration of the cuticle, M. grisea uses turgorpressure in its melanized appressorium while Bgh pene-trates cell walls by the combined activity of cellulases andturgor pressure (Pryce-Jones et al., 1999; Talbot, 2003).Other pathogens secrete cell wall-degrading enzymes,such as cutinases, cellulases, pectinases and wax degrad-ing enzymes for penetration of their host. After gainingentrance to the host plants, pathogens require additional‘tools’ to neutralize the defence reaction of the host and togain access to nutrients.

Resistance in plants to pathogens

Resistance in plants to pathogens is often categorized inthree types; nonhost, race-nonspecific and race-specificresistance. Nonhost resistance is defined by the resistanceof an entire plant species against a specific non-adaptedpathogenic microbe and is the most common and durableform of disease resistance exhibited by plants. It is theresult of both preformed and inducible defence mecha-nisms and seems to be under complex genetic controlwhich can involve multiple defence factors that individ-ually may segregate within host species without com-promising overall resistance (Heath, 2002; Mysore &Ryu, 2004). Race-nonspecific resistance is known as gen-eral, quantitative or partial resistance of a plant against aspecies of an adapted pathogen. It is generally durable andis mostly controlled by genes with incremental/additiveeffects, such as combinations of wheat leaf rust resis-tance gene Lr34 and several ‘slow-rusting’ genes (Singh& Rajaram, 1992). Race-specific resistance is qualitative,usually controlled by dominant avirulence (Avr) genesin the pathogen and corresponding dominant resistance(R) genes in a specific plant genotype or cultivar of anotherwise susceptible host species. This resistance is ofthe ‘gene-for-gene interaction’ type and is often easilyovercome by the pathogen.

Preformed physical barriers can include waxy cuticu-lar surface layers of the leaves and thick and stable cellwalls that most microbes are not equipped to penetrate.Even after successful penetration, the pathogen needs toovercome the biochemical barriers that include low pHof the apoplast, broad-spectrum antimicrobial compoundsand ‘defence’ enzymes that degrade microbial cell walls

Dow

nloa

ded

by [

Can

adia

n A

gric

ultu

re L

ibra

ry, A

gric

ultu

re a

nd A

gri-

Food

Can

ada]

at 1

3:23

08

Mar

ch 2

012

Page 3: Fungal and oomycete effectors – strategies to subdue a host · Can. J. Plant Pathol. (2011), 33(4): 425–446 Minireview/Minisynthèse Fungal and oomycete effectors – strategies

Fungal and oomycete effectors 427

(Felle, 1998; Dangl & Jones, 2001; Huckelhoven, 2007).Cell wall components such as actin microfilaments playan important role in defence against fungal penetrationand whose disruption results in loss of nonhost resistanceagainst several nonhost fungi (Kobayashi et al., 1997;Heath, 2002; Mysore & Ryu, 2004). Biochemical barriersconsist of many peptides, proteins and non-proteinaceoussecondary antimicrobial metabolites which may deter-mine the host range of some pathogens (Broekaert et al.,1995; Morrissey & Osbourn, 1999), and fungitoxic sub-stances on the surface of leaves in sufficient concen-tration to inhibit the germination of spores (Agrios,2005). Phytoanticipins, preformed antimicrobial com-pounds such as tannins and fatty acid-like dienes, oftenpresent in high concentration in cells of young fruits,leaves or seeds, or saponins, are responsible for resis-tance to pathogens that lack the corresponding detoxifyingenzymes (Osbourn et al., 1996; Pedras & Ahiahonu,2005). Other classes of antimicrobial compounds areproteases, such as cysteine proteases secreted into theapoplast by tomato and potato (Lucas, 1998; Kruger et al.,2002) and protease inhibitors, such as a cysteine proteaseinhibitor produced by pearl millet (Joshi et al., 1999).

Some of the above-mentioned defence systems arepreformed or could be kicked into higher gear, orrepresent inducible mechanisms only activated in thehost when attacked. Examples are induced physicaldefences, such as the formation of cell wall deposi-tions (papilla) directly under the penetration sites ofpathogens; this can stop up to 90% of the penetra-tion attempts in the Bgh/Arabidopsis nonhost interaction(Collins et al., 2003). The production of phytoalexinsis often induced upon attack (Hammerschmidt, 1999).Pathogenesis-related (PR) defence proteins such as chiti-nases and β-1,3 glucanase, (per-)oxidases, proteinaseinhibitors, thionins and lipid-transfer proteins, are inducedin response to pathogen attack and the accumulation ofthese proteins is usually associated with the acquisitionof systemic resistance in plants against a wide rangeof other pathogens. These are specific families of pro-teins, currently in 17 classes (van Loon et al., 2006)although a larger group of ‘‘inducible defence-related pro-teins’’, including phenylalanine ammonia lyase, one ofthe key enzymes involved in the synthesis of aromaticcompounds, like phytoalexins, is often included in the PRproteins.

Pathogen-associated molecular patterns

In addition to preformed and inducible physical and bio-chemical barriers, plants also have evolved a surveillancesystem to recognize various pathogen surface-exposed

and cytoplasmic molecules known as pathogen (microbe)associated molecular patterns: PAMPs or MAMPs (Shiu& Bleecker, 2003). These are highly conserved moleculesof microbes that are perceived by host receptors (PAMPrecognition receptors or PRRs) at an early stage of infec-tion. Recognition results in induction of PAMP-triggeredimmunity (PTI). Examples of surface-exposed PAMPsthat have been shown to be capable of triggering PTIare bacterial flagellin (Felix et al., 1999), lipopolysaccha-rides (LPS; Meyer et al., 2001; Erbs & Newman, 2003),lipooligosaccharides from Gram-negative bacteria, chitinfrom cell walls of higher fungi (Bartnicki-Garcia, 1968;Ren & West, 1992), invertase from Saccharomyces cere-visiae (Basse et al., 1992), and 1,3-1,6-hepta-β-glucosidefrom the cell walls of Phytophthora (Sharp et al., 1984a,1984b). Examples of cytoplasmic PAMPs that inducehost defence are cold shock protein (CSP) and elonga-tion factor Tu (Ef-Tu; Felix & Boller, 2003; Kunze et al.,2004).

Pathogen effectors and their delivery duringplant–microbe interactions

Pathogens secrete a wide range of so-called ‘effectors’,small molecules and proteins that can modify host cellstructures or functions. Effectors from several groups ofpathogens such as bacteria, oomycetes and fungi can enterplant cells (Huang et al., 2003; Chisholm et al., 2006;Kamoun, 2007). Bacteria use the Type II, Type IV andType III Secretion System (T3SS) to deliver 20 to hun-dreds of protein effectors into the host plant (Lindeberget al., 2006; Cunnac et al., 2009). Oomycetes and fungilikely secrete even more protein effectors; computationalanalysis of their genomes has revealed hundreds (Kamperet al., 2006; Tyler et al., 2006; Jiang et al., 2008; Muelleret al., 2008; Haas et al., 2009; Schirawski et al., 2010),even more than 1700 of potential effectors for rust fungi(Duplessis et al., 2011).

Predicted effectors from oomycetes, in addition to aN-terminal signal peptide (SP), carry a host targeting sig-nal (HTS) next to the SP that contains conserved RXLRand a dEER motifs that can target them into host cells inthe absence of the pathogen (Rehmany et al., 2005; Tyleret al., 2006; Whisson et al., 2007; Jiang et al., 2008; Douet al., 2008b; Haas et al., 2009; Kale et al., 2010), whileeffectors from fungi have an N-terminal SP and in somecases an RXLR-like motif (Dean et al., 2005; Kamperet al., 2006; Kale et al., 2010; Schirawski et al., 2010).Many examples exist where predicted effectors with aSP are shown to be delivered into the host cytoplasm(Kemen et al., 2005; Catanzariti et al., 2006; Mosqueraet al., 2009; Doehlemann et al., 2009; Khang et al.,

Dow

nloa

ded

by [

Can

adia

n A

gric

ultu

re L

ibra

ry, A

gric

ultu

re a

nd A

gri-

Food

Can

ada]

at 1

3:23

08

Mar

ch 2

012

Page 4: Fungal and oomycete effectors – strategies to subdue a host · Can. J. Plant Pathol. (2011), 33(4): 425–446 Minireview/Minisynthèse Fungal and oomycete effectors – strategies

S. Ali and G. Bakkeren 428

2010) and some can do so through N-terminal sequenceswithout the help of the pathogen (Rafiqi et al., 2010; Kaleet al., 2010). Recently, Kale et al. (2010) showed that theconserved RXLR motif from oomycetes and the RXLR-like motif from other fungi bind specifically to phos-pholipids, in particular phosphatidylinositol-3-phosphate(PI3P) on the surface of the plasma membrane and likelyenter the cell through lipid raft-mediated endocytosis;results on lipid-binding assays have been controversial(Gan et al., 2010). In a search for possible equivalentmotifs in other fungi, Godfrey et al. (2010) showed thatsmall secreted proteins from haustoria-forming fungalpathogens share a conserved Y/F/WxC motif in additionto an N-terminal secretion signal although no function hasyet been ascribed to these motifs.

Roles of effectors in infection

Breaking the physical barrier

After landing on the surface of plants, pathogens eitherenter their host through natural openings (stomata andhydathodes) or penetrate the surface directly. Bacteriause natural openings or wounds to enter the apoplastfor colonization. Pathogenic bacteria sense compoundsreleased during photosynthesis from stomata and movetowards them. The PRR of the guard cells can sensePAMPs such as bacterial flagellin or lipopolysaccharidesor lipooligosaccharides, which induces the closure ofstomata (Gohre & Robatzek, 2008). Pathogenic bacte-ria, such as Pseudomonas syringae, produce coronatine,a phytotoxin that mimics jasmonic acid (JA) to interferewith salicylic acid (SA) and abscisic acid (ABA) sig-nalling for reopening stomata to get access to the hostapoplast (Melotto et al., 2006). It is not yet known whetherMAMP-associated defence pathways also close stomatato eukaryotic pathogens and whether these pathogens,such as cereal rusts, use effectors in a similar way to over-come this hurdle. Powdery mildew fungi, such as Bgh,penetrate the cuticle of plant cells directly by forming anappressorium. It has been shown that effectors Avra10 andAvrk1 increase the penetration efficiency of Bgh on sus-ceptible barley cultivars but the exact mechanism is notyet known (Ridout et al., 2006). After getting into theplant apoplast, the next physical barrier is the plant cellwall. To promote nutrient leakage from the cytosol into theapoplast, lytic enzymes that degrade the cell wall locallyare secreted. At the same time, pathogens secrete effectormolecules to suppress the host defence that is activated bydanger-associated molecular patterns (DAMPs) generatedfrom degrading cell wall molecules and signalling dam-age to the plant integrity and hence potential danger ofinfection (Jha et al., 2007).

Disarming plant defence enzymes

Plants produce antimicrobial enzymes such as proteases,hydrolases, glucanases and chitinases that can degrade thecell wall of invading pathogenic fungi in the apoplast,without detrimental effect to the plant itself (Lucas, 1998).This has a dual role in defence; the degradation of cellwalls can attenuate fungal growth while on the other handthe molecules released from degraded cell walls serveas elicitors for the induction of host defence. Pathogensuse effector molecules either to stop the delivery ofthese antimicrobial enzymes and compounds by prevent-ing their secretion, or by inhibiting their activity after theyare secreted (Bent & Mackey, 2007), and/or by inhibitingdownstream signalling. Bacteria secrete plant cell wall-degrading enzymes locally in order to construct the T3SSand use effectors such as HopP1 to suppress defenceinduced by DAMPs in the N. benthamiana apoplast (Gustet al., 2007; Oh et al., 2007). HopP1 either sequestersor processes the fragments that are produced during cellwall degradation so that they cannot function as DAMPs(Gohre & Robatzek, 2008).

Examples from fungi are represented by the apoplasticfungus Cladosporium fulvum which secretes effectorAVR2, a cysteine protease inhibitor, during infection thatbinds directly to RCR3, a tomato cysteine protease, to pro-tect the fungus from the deleterious effect of the enzyme.AVR2 also promotes virulence for other fungal pathogensthat cause diseases in tomato, such as Verticillium dahliaeand Botrytis cinerea, when expressed heterologously inArabidopsis (van Esse et al., 2008). Similarly, C. fulvumeffector AVR4 binds to chitin of fungal cell walls to pro-tect it from tomato host chitinases (van den Burg et al.,2006). AVR4 can also protect chitin against plant chiti-nases in the cell wall of other fungi, such as Trichodermaviride and Fusarium solani f. sp. phaseoli (van den Burget al., 2006). In this way, AVR4 not only protects thefungi from hydrolysis by plant chitinases but also keepschitin fragments from eliciting PTI (Libault et al., 2007).Effector ECP6 from C. fulvum has a Lys-M domain thatbinds to carbohydrates including chitin and protects thepathogen from plant chitinases or may be involved inscavenging of chitin fragments that are released duringcell wall degradation by plant chitinases, thus preventingthem from inducing PTI (Bolton et al., 2008).

The oomycete pathogen, P. infestans, is known tosecrete a suite of Cysteine and Kazal family proteaseinhibitors. The tomato papain-like protease, PIP1, whichis induced by SA, is blocked by the EPIC2B inhibitorof P. infestans (Tian & Kamoun, 2005; Tian et al.,2007; van Esse et al., 2008). PIP1 is related to RCR3,and AVR2 from C. fulvum apparently can also inhibit

Dow

nloa

ded

by [

Can

adia

n A

gric

ultu

re L

ibra

ry, A

gric

ultu

re a

nd A

gri-

Food

Can

ada]

at 1

3:23

08

Mar

ch 2

012

Page 5: Fungal and oomycete effectors – strategies to subdue a host · Can. J. Plant Pathol. (2011), 33(4): 425–446 Minireview/Minisynthèse Fungal and oomycete effectors – strategies

Fungal and oomycete effectors 429

PIP1 and two other plant cysteine proteases, aleurainand TDI65 (Rooney et al., 2005; Shabab et al., 2008;van Esse et al., 2008). P. infestans EPIC1 and EPIC2Bcan also bind and inhibit RCR3, similar to AVR2, butunlike AVR2, these EPICs do not elicit an HR on Cf-2/Rcr3pimp tomato plants, suggesting that P. infestansevolved stealthy effectors that can inhibit tomato pro-teases without activating defence responses (Song et al.,2009). These findings show that effectors from differ-ent pathogens can target the same apoplastic enzymesto increase pathogen fitness in the host (Shabab et al.,2008). Recently, another apoplastic tomato and potatopapain-like cysteine protease, C14, was shown to be tar-geted by EPIC1 and EPIC2B (Kaschani et al., 2010).Other P. infestans effectors, EPI1 and EPI10, target thesubtilisin-like serine protease of tomato P69B, a PR pro-tein (Tian et al., 2004; Tian & Kamoun, 2005; Tianet al., 2007). AVRP123, a secreted protein from the flaxrust fungus M. lini, also shows similarity to Kazal serineprotease inhibitors (Catanzariti et al., 2006). The soybeanpathogen, P. sojae, secretes glucanase inhibitor proteins,GIP1 and GIP2, that target endo-β-1,3-glucanaseA of thehost plant to protect the pathogen during infection andalso to prevent PTI induced by oligoglucosides (Roseet al., 2002) as discussed next.

Suppression of receptor activation

The recognition of PAMPs by plant PRRs leads to the acti-vation of defence responses against the pathogen. It wasproposed in the beginning that resistance induced byPRRs is only basic and not as strong as that induced byresistance genes, but it is clear now, at least in the caseof FLS2 (the flagellin receptor), that the contribution ofPRRs towards overall resistance is huge (He et al., 2006;de Torres et al., 2006; Hann & Rathjen, 2007). It is there-fore not surprising that pathogens have evolved to targetwith their effectors these signalling pathways in order toreduce resistance responses. Insight into the molecularmechanisms involved has emerged over the last few years.For example, several effectors target the PRRs directlyto jam all the downstream resistance responses (Jamiret al., 2004; Jones & Dangl, 2006; Blocker et al., 2008)such as P. syringae AVRPto and AVRPtoB which canalso suppress nonhost HR in N. benthamiana induced byFLG22 or P. infestans INF1 (Hann & Rathjen, 2007).

Several oomycete RXLR effectors can suppress hostcell immunity in a similar manner. AVR3A fromP. infestans can block an HR induced by INF1 (Boset al., 2009) as can several random P. infestans effectors(Oh et al., 2009). Similarly, AVR1B from P. sojaealso suppresses programmed cell death triggered by

the mouse protein, BAX, in plants and yeast (Douet al., 2008a) and M. oryzae AvrPiz-t can also suppressmouse BAX protein-mediated programmed cell death intobacco leaves (Li et al., 2009). ATR1 and ATR13 fromHyaloperonospora arabidopsidis increased the virulenceof P. syringae DC3000 on susceptible Arabidopsis whenthese effectors were delivered by the P. syringae T3SS,and ATR13 seemed to target PTI by suppressing calloseaccumulation and ROS production (Sohn et al., 2007).In the genus Phytophthora, effectors such as P. infes-tans IpiO1 bind to a membrane-spanning lectin receptorkinase and seem to prevent downstream defence sig-nalling (Bouwmeester et al., 2011).

Suppression of R gene-triggered resistance

Pathogenic fungi such as Fusarium oxysporum f. sp.lycopersici (Fol) can avoid host defences by evolvingeffectors that can suppress R gene-triggered resistance(Houterman et al., 2008). AVR3 and AVR2 are requiredfor full virulence on tomato plants but they are also rec-ognized by tomato lines that have resistance genes I-3or I-2, respectively, triggering an HR and cause arrestof the pathogen (Huang & Lindhout, 1997; Rep et al.,2004, 2005). In contrast, AVR1 is a small cysteine-richsecreted protein that is recognized by resistance gene Ior I-1 but is not required for virulence. It seems gener-ally true that effectors that contribute to full virulence aremaintained in the species and tellingly, AVR3 is presentin all Fol strains analysed, while AVR1 is present onlyin Fol strains that are virulent on I-3 lines. Houtermanet al. (2008) showed that AVR1 actually suppresses theresistance triggered by I-2 and I-3, as the transformationof Avr1 to Fol strains that were avirulent on I-2 or I-3became virulent on these lines. It was proposed that Folstrains acquired Avr1 as a mechanism of partial functionalredundancy so that they can avoid the consequences oflosing Avr3 and probably Avr2 that are required for fullvirulence (Stergiopoulos & de Wit, 2009). In the flax rustfungus, M. lini, the interaction of AvrL567 in strain CH5-89 with the flax cultivar ‘Barnes’ that contains the L7gene is inhibited by an inhibitor gene and thus results in alower virulence reaction (Lawrence et al., 2010). P. infes-tans secreted effector SNE1 can suppress programmedcell death resulting from the interaction of several Avr-Rprotein interactions (Kelley et al., 2010).

Alteration of the plant defence transcriptome

Bacterial effectors have been identified many years beforefungal and oomycete effectors and it is therefore not sur-prising that a better understanding of the impact of such

Dow

nloa

ded

by [

Can

adia

n A

gric

ultu

re L

ibra

ry, A

gric

ultu

re a

nd A

gri-

Food

Can

ada]

at 1

3:23

08

Mar

ch 2

012

Page 6: Fungal and oomycete effectors – strategies to subdue a host · Can. J. Plant Pathol. (2011), 33(4): 425–446 Minireview/Minisynthèse Fungal and oomycete effectors – strategies

S. Ali and G. Bakkeren 430

effectors on the host transcriptome during infection hascome from the study of bacteria–plant interactions. Forexample, the perception of PAMPs such as bacterial flag-ellin changes the expression of at least 1000 genes inArabidopsis (Zipfel et al., 2004). Microbes have evolvedto become pathogens in part because their effectors inhibitthe activation of defence genes. Effectors can have an indi-rect effect on transcription by dephosphorylating MAPKsignalling components in order to suppress the defenceresponse triggered by the recognition of PAMPs by PRRs(reviewed in Gohre & Robatzek, 2008). Another mech-anism by which effectors have been shown to affectthe plant transcriptome is by causing changes in RNAstability. For example, P. syringae HopU1, a mono-ADP-ribosyltransferase, acts on glycine-rich RNA-binding pro-teins such as Arabidopsis AtGRP7 and AtGRP8 which areRNA chaperones and thus, HopUI changes the plant tran-scriptome by reducing transcript levels and hence affectsthe production of defence response proteins (Fu et al.,2007). Some pathogen effectors act as transcription fac-tors and can induce the expression of host genes for thebenefit of the pathogen. For example, the AVRBs3 fam-ily of effectors from X. campestris pv. vesicatoria hasplant nuclear localization signals and binds to a ‘upa-box’(upregulated by AVRBs3) that is found in the promoter ofUpa20, a master regulator of cell size inducing hypertro-phy, and several other host genes, ensuring proper nutrientsupply for pathogen multiplication (Szurek et al., 2002;Gurlebeck et al., 2006; Kay et al., 2007); however, inresistant plant cultivars, the promoter of Bs3 also carriesa ‘upa-box’ and, thus, binding of the AVRBs3 effectorinduces transcription of Bs3 and cell death (Kay et al.,2007). This shows that under selection pressure, plantscan evolve to recognize effectors and use them for theirown defence.

Several studies in fungal and oomycete pathosys-tems have reported on changes in host transcriptomes:in compatible interactions, initial defence reactions sup-posedly triggered by recognition of PAMPs, are theresult of the upregulation of genes in defence pathways;at later infection stages, these are suppressed and thiseffect is proposed to be caused by suites of secretedpathogen effectors. Such a scenario has been demon-strated in the smut fungus Ustilago maydis–maize inter-action (Doehlemann et al., 2008). In this latter system,effectors likely even reprogram specific developmentalpathways in the host to create the best niche for theirown development, depending on the age and part ofthe plant infected (Skibbe et al., 2010). One particulareffector, Pit2, was deduced to suppress defence genes(Doehlemann et al., 2011). In the M. oryzae–rice inter-action, changes in the host transcriptome were noted

including upregulation of susceptibility genes and down-regulation of defence genes and these effects on genetranscription were attributed to the actions of secretedeffectors, although no particular effector could be iden-tified to cause specific effects (Mosquera et al., 2009).

Killing of host cells

Necrotrophic pathogens produce several phytotoxins inaddition to cell wall hydrolysing enzymes in order tokill the host tissue for colonization during infection.Some necrotrophic fungi produce proteinaceous effectors,also called host selective toxins (HST), that are requiredfor infection on susceptible host plants that have thecorresponding dominant receptor gene (Wolpert et al.,2002). This represents a situation opposite to the classi-cal gene-for-gene interaction in which a dominant gene isrequired for disease resistance rather than susceptibility.Two wheat snecrotrophs, Stagonospora nodurum (Sn) andPyrenophora tritici-repentis (Ptr), produce several host-specific peptide effectors, such as PtrTOXA, SnTOX1,SnTOX2 and SnTOX4, that, when recognized by theircorresponding dominant susceptibility genes in wheat(TsN1, Snn1, Snn2 and Snn4), cause disease (Liu et al.,2004, 2006; Friesen et al., 2007; Abeysekara et al., 2009;de Wit et al., 2009; Manning et al., 2009). PtrTOXA, thebest-studied effector, has an N-terminal secretion signal,followed by an RGD domain (Arg-Gly-Asp-containingloop) for host targeting and a C-terminal domain witheffector function (Sarma et al., 2005; Manning et al.,2007). After entering the host cell, PtrTOXA was reportedto enter the chloroplast and bind to ToxABP1, therebyinterfering with functions of the chloroplast and affectingphotosystem I and II function in a light-dependent man-ner (Manning et al., 2007). PtrTOXA is an ortholog toSnTOX1 and both effectors are recognized by the samewheat susceptibility gene, Tsn1 (Liu et al., 2006).

Some pathogenic bacteria, oomycetes and fungi pro-duce NEP1-like (necrosis-inducing protein) proteins(NLPs), effectors that are toxic to only dicotyledonousplants, possibly because of the different molecularcompositions of dicot and monocot cell membranes(Pemberton & Salmond, 2004; Gijzen & Nurnberger,2006; Ottmann et al., 2009). The common hepta-peptidemotif, GHRHDWE, and two conserved cysteine residuesmake NLPs structurally similar to actinoporins, cytolytictoxins from marine organisms. Hemibiotrophs, such as P.infestans and P. sojae, produce NLPs such as NPP1 andPiNPP1, in the late necrotrophic phase which couldcontribute to disease development with their cytolyticactivities (Qutob et al., 2002; Kanneganti et al., 2006).

Dow

nloa

ded

by [

Can

adia

n A

gric

ultu

re L

ibra

ry, A

gric

ultu

re a

nd A

gri-

Food

Can

ada]

at 1

3:23

08

Mar

ch 2

012

Page 7: Fungal and oomycete effectors – strategies to subdue a host · Can. J. Plant Pathol. (2011), 33(4): 425–446 Minireview/Minisynthèse Fungal and oomycete effectors – strategies

Fungal and oomycete effectors 431

Suppression of host defence by symbionts

Symbiotic microorganisms also secrete effectors into hostplants to suppress host defence responses, a conditionessential for their lifestyle. Many rhizobial strains usethe T3SS to deliver effectors into host cells to suppresshost defence responses in a similar way that pathogenicbacteria do (Deakin & Broughton, 2009). This couldindicate that this ancient delivery system is used for dif-ferent purposes or that symbiotic organisms have evolvedtheir effectors such as to overcome most host defenceresponses. In the fungal kingdom, the genome of Laccariabicolor, an ectomycorrhizal fungus, revealed more than3000 predicted secreted proteins of which 10% were smallsecreted proteins (SSPs) of the effector-type (Martin et al.,2008; Martin & Selosse, 2008). Some of these SSPs arehomologous to rust fungus ‘haustoria-expressed secretedproteins’ (HESPs) and are differentially expressed dur-ing infection. One effector, MiSSP7, was recently shownto localize to host cell nuclei upon cell entry where italtered host transcription and was shown to be necessaryfor promoting symbiosis and evading the host defence(Plett et al., 2011).

Gene-for-gene or R-Avr gene interaction

Adapted pathogens secrete effector molecules into thehost plant to surmount physical barriers, neutralizepreformed antimicrobial compounds, and overcome PTI.As a result, during co-evolution of plant host and adaptedpathogen, plants have developed sophisticated recognitionsystems, usually encoded by R genes to recognize theseeffectors or their action and trigger defence responses;this induced resistance has been called effector-triggeredimmunity (ETI) and leads to a rapid and enhanced defenceresponse in the host plant often including an HR (Jones& Dangl, 2006). These effectors represent avirulence(AVR) factors since they activate the plant defence sys-tem and make the pathogen unable to cause disease whenthe plant has the ‘recognizing’ R gene. This geneticallysuperimposed R and Avr interaction has been known formany years as ‘gene-for-gene’ resistance, or host/cultivar-level resistance, as particular cultivars of the host witha certain R gene product recognize an Avr gene productfrom a particular race of the pathogen. This concept wasformulated after genetic studies on two fungal pathosys-tems: the Melampsora lini–flax (Flor, 1942) and theUstilago tritici–wheat interactions (Oort, 1944). As men-tioned, some Avr genes encode effectors that suppress hostdefence or carry out other essential functions and, thus,act as virulence factors when the plant does not have the‘recognizing’ resistance gene. ETI represents the quali-tative, secondary layer of resistance and has led to an

evolutionary arms race between the pathogen and the plantin which the pathogen either mutates or discards effectorsto avoid recognition by the host or develops new effectorsto suppress ETI, while the plant develops new R genes torecognize the mutated or new effectors (Jones & Dangl,2006; de Wit, 2007; Bent & Mackey, 2007)

Avr (-triggering effector) genes

Because, until recently, the term avirulence gene/factorfeatured prominently in the literature, we will discussthese below but would prefer the term ‘Avr-triggeringeffector genes’. The first Avr gene was cloned from abacterium in 1984 (Staskawicz et al., 1984), which wasfollowed by the cloning and characterization of more than40 bacterial Avr genes in the following decades (van ’tSlot & Knogge, 2002; Mudgett, 2005). The cloning andcharacterization of Avr genes from fungi and oomyceteslagged behind because of these organisms’ larger genomesizes and sometimes inefficient transformation systems.The first Avr gene from a fungus was isolated from C. ful-vum in 1991 (van Kan et al., 1991) and the first oomyceteAvr gene was cloned from P. sojae and P. infestans in2004 (Shan et al., 2004). In the following years, mostfungal Avr genes were cloned from ascomycetes, althoughmore recently, a few have been reported from a basid-iomycete, the flax rust fungus, M. lini (Catanzariti et al.,2006; Table 1).

Avr genes isolated and characterized to date differamong one another both in sequence and function, andthose from different plant pathogens do not seem toshare many common features, other than the ones alreadymentioned. Exceptions are found among some familymembers, e.g. in Pseudomonas and Xanthomonas species(Lahaye & Bonas, 2001; Deslandes et al., 2003). And it istrue that among genomes from related species, sometimeseven among genera, homologous effectors are found, butthese, or not all of these, have proven avirulence functions.Most fungal and oomycete Avr genes characterized to dateencode small proteins (28–311 amino acids), except theACE1 of M. grisea (see below) and most have a secre-tion signal/protein transport motif at the N-terminus (Elliset al., 2006), although there are exceptions (see further).

Fungal Avr (-triggering effector) genes

Cladosporium fulvum

This fungus is an apoplastic pathogen of the ascomycetesubgroup that reproduces asexually and causes leaf moldof tomato (de Wit et al., 1997; Joosten & de Wit, 1999;Thomma et al., 2005). Four avirulence genes have beencloned: Avr2, Avr4, Avr4E and Avr9, which all encodesmall secreted cysteine-rich effector proteins that are

Dow

nloa

ded

by [

Can

adia

n A

gric

ultu

re L

ibra

ry, A

gric

ultu

re a

nd A

gri-

Food

Can

ada]

at 1

3:23

08

Mar

ch 2

012

Page 8: Fungal and oomycete effectors – strategies to subdue a host · Can. J. Plant Pathol. (2011), 33(4): 425–446 Minireview/Minisynthèse Fungal and oomycete effectors – strategies

S. Ali and G. Bakkeren 432

Tabl

e1.

Eff

ecto

rpr

otei

nsof

fung

alan

doo

myc

ete

plan

tpat

hoge

ns.

Prot

ein

Org

anis

m

Len

gth

aare

sidu

es(m

atur

e)N

umbe

rof

cyst

eine

sSP

leng

th(a

a)B

iolo

gica

lact

ivity

/ho

mol

ogy

Prot

ein

loca

lizat

ion

Rol

ein

viru

lenc

e/pa

thog

enic

ityC

orre

spon

-din

gR

gene

Ref

eren

ces

AV

R2

C.f

ulvu

m78

(58)

820

Indu

ces

HR

inth

epr

esen

ceof

Tom

ato

Rcr

3,Pr

otea

sein

hibi

tor

Apo

plas

tIn

hibi

tsR

cr3

and

othe

rpr

otea

ses

Cf-

2St

ergi

opou

los

etal

.,20

10A

VR

4C

.ful

vum

135

(86)

818

Indu

ces

HR

,Chi

tin-b

indi

ng,

orth

olog

sin

som

eot

her

fung

iA

popl

ast;

fung

alce

llw

allc

hitin

Prot

ects

agai

nst

chiti

nase

sC

f-4

Joos

ten

etal

.,19

94;

Ster

giop

oulo

set

al.,

2010

;van

den

Bur

get

al.,

2006

AV

R4E

C.f

ulvu

m12

1(1

01)

610

Indu

ces

HR

Apo

plas

tU

nkno

wn

Hcr

9-4E

Wes

teri

nket

al.,

2004

AV

R9

C.f

ulvu

m63

(28)

623

Indu

ces

HR

,Car

boxy

pept

idas

ein

hibi

tor

Apo

plas

tU

nkno

wn

Cf-

9va

nde

nA

cker

veke

net

al.,

1993

EC

P1C

.ful

vum

96(6

5)8

23In

duce

sH

R,T

umor

-nec

rosi

sfa

ctor

rece

ptor

Apo

plas

tD

isru

ptio

nle

ads

tore

duce

dvi

rule

nce

Cf-

Ecp

1va

nde

nA

cker

veke

net

al.,

1993

EC

P2C

.ful

vum

165

(143

)4

22in

duce

sH

R,

Apo

plas

tD

isru

ptio

nle

ads

tore

duce

dvi

rule

nce

Cf-

Ecp

2va

nde

nA

cker

veke

net

al.,

1993

EC

P4C

.ful

vum

119

(101

)6

18In

duce

sH

RA

popl

ast

Unk

now

nC

f-E

cp4

Bol

ton

etal

.,20

08E

CP5

C.f

ulvu

m11

5(9

8)6

17In

duce

necr

osis

,A

popl

ast

Unk

now

nC

f-E

cp5

Bol

ton

etal

.,20

08E

CP6

C.f

ulvu

m22

2(1

99)

823

LysM

-dom

ains

;chi

tin-b

indi

ng,

orth

olog

foun

din

diff

eren

tpa

thog

enan

dno

npat

hoge

nic

sp.

Apo

plas

tK

nock

-dow

nle

ads

tore

duce

dvi

rule

nce

Unk

now

nde

Jong

e&

Tho

mm

a,20

09

EC

P7C

.ful

vum

–(1

00)

6–

Unk

now

nA

popl

ast

Unk

now

nU

nkno

wn

Bol

ton

etal

.,20

08N

IP1

R.s

ecal

is82

(60)

1022

non-

spec

ific

toxi

n/in

duce

sne

cros

isan

dpl

asm

a-m

embr

ane

H+

AT

Pase

Prob

ably

inap

opla

stN

otre

quir

edfo

rvi

rule

nce

Rrs

-1R

ohe

etal

.,19

95

NIP

2R

.sec

alis

109

(?)

7(6

)16

Non

-spe

cific

toxi

n/in

duce

sne

cros

isin

seve

ralp

lant

spp.

Prob

ably

inap

opla

stN

otre

quir

edfo

rfu

llvi

rule

nce

Unk

now

nR

ohe

etal

.,19

95;

Ster

giop

oulo

s&

De

Wit,

2009

NIP

3R

.sec

alis

115

(?)

9(8

)17

Non

-spe

cific

toxi

n/in

duce

sne

cros

isin

seve

ralp

lant

ssp

ecie

sPr

obab

lyin

apop

last

Not

requ

ired

for

full

viru

lenc

eU

nkno

wn

Roh

eet

al.,

1995

;St

ergi

opou

los

&de

Wit,

2009

AV

Ra1

0B

.gra

min

is28

64

–>

30pa

ralo

gsin

Bgh

and

othe

rf.

sp.

No

N-t

erm

inal

SP,i

nduc

esH

RPr

obab

lyin

cyto

plas

mU

nkno

wn

Mla

10R

idou

teta

l.,20

06

AV

Rk1

B.g

ram

inis

177

3–

>30

para

logs

inB

ghan

dot

her

f.sp

.N

oN

-ter

min

alSP

,ind

uces

HR

Prob

ably

incy

topl

asm

Unk

now

nM

lk1

Rid

oute

tal.,

2006

AV

RL

567

(A,B

and

C)

M.l

ini

150

(127

)1

23U

nkno

wn,

indu

ces

HR

,fun

ctio

nal

RX

LR

like

mot

ifC

ytop

lasm

Unk

now

nL

5,L

6an

dL

7D

odds

etal

.,20

04;

Kal

eet

al.,

2010

;R

afiqi

etal

.,20

10

Dow

nloa

ded

by [

Can

adia

n A

gric

ultu

re L

ibra

ry, A

gric

ultu

re a

nd A

gri-

Food

Can

ada]

at 1

3:23

08

Mar

ch 2

012

Page 9: Fungal and oomycete effectors – strategies to subdue a host · Can. J. Plant Pathol. (2011), 33(4): 425–446 Minireview/Minisynthèse Fungal and oomycete effectors – strategies

Fungal and oomycete effectors 433

AV

RM

M.l

ini

314

(286

)1

28U

nkno

wn,

indu

ces

HR

,RX

LR

like

mot

ifC

ytop

lasm

Unk

now

nM

Cat

anza

riti

etal

.,20

06;K

ale

etal

.,20

10;R

afiqi

etal

.,20

10A

VR

P123

M.l

ini

117

(94)

1123

Indu

ces

HR

,Kaz

alSe

rine

prot

ease

inhi

bito

rPr

obab

lyin

cyto

plas

mU

nkno

wn

P,P

1,P

2an

d/or

P3

Cat

anza

riti

etal

.,20

06A

VR

P4M

.lin

i95

(67)

728

Indu

ces

HR

,cys

tein

e-kn

otte

dpe

ptid

ePr

obab

lyin

cyto

plas

mU

nkno

wn

P4

Cat

anza

riti

etal

.,20

06A

vr-P

ita1

M.o

ryza

e22

3(1

76)

816

Hom

olog

yto

met

allo

-pro

teas

es,R

XL

Rlik

em

otif

Cyt

opla

smN

otre

quir

edfo

rvi

rule

nce

onri

ceP

i-ta

Orb

ach

etal

.,20

00;

Kal

eet

al.,

2010

;K

hang

etal

.,20

10A

VR

-Pita

2M

.ory

zae

224

(?)

816

Hom

olog

yto

met

allo

-pro

teas

esPr

obab

lyin

apop

last

Prob

ably

not

requ

ired

for

viru

lenc

eon

rice

Pi-

taK

hang

etal

.,20

08

AV

R-P

ita3

M.o

ryza

e22

6(?

)8

16H

omol

ogy

tom

etal

lo-p

rote

ases

Prob

ably

incy

topl

asm

Prob

ably

not

requ

ired

for

viru

lenc

eon

rice

Unk

now

nK

hang

etal

.,20

08

PWL

1M

.ory

zae

147

(124

)2

23G

lyci

ne-r

ich

hydr

ophi

licpr

otei

nB

iotr

ophi

cin

terf

acia

lco

mpl

ex

Unk

now

nU

nkno

wn

Kan

get

al.,

1995

;K

hang

etal

.,20

10PW

L2

M.o

ryza

e14

5(1

24)

221

Gly

cine

-ric

hhy

drop

hilic

prot

ein

Cyt

opla

smU

nkno

wn

Unk

now

nSw

eiga

rdet

al.,

1995

;Kha

nget

al.,

2010

PWL

3M

.ory

zae

137

(116

)0

21G

lyci

ne-r

ich

hydr

ophi

licpr

otei

nPr

obab

lyin

apop

last

Non

-fun

ctio

nal

Unk

now

nK

ang

etal

.,19

95

PWL

4M

.ory

zae

138

(117

)0

21G

lyci

ne-r

ich

hydr

ophi

licpr

otei

nPr

obab

lyin

apop

last

Non

-fun

ctio

nal

Unk

now

nK

ang

etal

.,19

95

AC

E1

M.o

ryza

e40

3543

–H

ybri

dpo

lyke

tide

synt

hase

/no

nrib

osom

alpe

ptid

esy

nthe

tase

Not

secr

eted

Unk

now

nP

i33

Boh

nert

etal

.,20

04

AV

R1-

CO

39M

.ory

zae

notc

lone

dye

t–

–U

nkno

wn

Unk

now

nU

nkno

wn

Pi-

CO

39(t

)Fa

rman

etal

.,20

02

AV

R-P

iaM

.ory

zae

85(6

6)2

19In

duce

sH

RPr

obab

lyin

cyto

plas

mU

nkno

wn

Pia

Yos

hida

etal

.,20

09

AV

R-P

iiM

.ory

zae

70(5

1)3

19In

duce

sH

RPr

obab

lyin

cyto

plas

mU

nkno

wn

Pii

Yos

hida

etal

.,20

09

AV

R-P

iiM

.ory

zae

70(5

1)3

19In

duce

sH

RPr

obab

lyin

cyto

plas

mU

nkno

wn

Pii

Yos

hida

etal

.,20

09

AV

R-

Pik/

km/kp

M.o

ryza

e11

3(9

2)3

21In

duce

sH

RPr

obab

lyin

cyto

plas

mU

nkno

wn

Pik

Yos

hida

etal

.,20

09

AV

RL

m1

L.m

acul

ans

205

(183

)1

22In

duce

sH

RPr

obab

lyin

cyto

plas

mR

equi

red

for

full

viru

lenc

eR

lm1

Gou

teta

l.,20

06

AV

RL

m6

L.m

acul

ans

144

(124

)6

20U

nkno

wn,

func

tiona

lRX

LR

like

mot

ifPr

obab

lyin

apop

last

Unk

now

nR

lm6

Fuda

leta

l.,20

07;

Kal

eet

al.,

2010

(Con

tinu

ed)

Dow

nloa

ded

by [

Can

adia

n A

gric

ultu

re L

ibra

ry, A

gric

ultu

re a

nd A

gri-

Food

Can

ada]

at 1

3:23

08

Mar

ch 2

012

Page 10: Fungal and oomycete effectors – strategies to subdue a host · Can. J. Plant Pathol. (2011), 33(4): 425–446 Minireview/Minisynthèse Fungal and oomycete effectors – strategies

S. Ali and G. Bakkeren 434Ta

ble

1.(C

ontin

ued.

)

Prot

ein

Org

anis

m

Len

gth

aare

sidu

es(m

atur

e)N

umbe

rof

cyst

eine

sSP

leng

th(a

a)B

iolo

gica

lact

ivity

/ho

mol

ogy

Prot

ein

loca

lizat

ion

Rol

ein

viru

lenc

e/pa

thog

enic

ityC

orre

spon

-din

gR

gene

Ref

eren

ces

AV

RL

m4-

7L

.mac

ulan

s14

3(1

22)

821

Unk

now

nPr

obab

lyin

apop

last

Req

uire

dfo

rfu

llvi

rule

nce

Rlm

4an

d/or

Rlm

7Pa

rlan

geet

al.,

2009

AV

R3

(SIX

1)F.

oxys

po-

rum

f.sp

.ly

cope

r-si

ci

284

(189

)6

or8

21U

nkno

wn

Xyl

emR

equi

red

for

full

viru

lenc

eI-

3R

epet

al.,

2004

;St

ergi

opou

los

&de

Wit,

2009

AV

R4

(SIX

2)F.

oxys

po-

rum

f.sp

.ly

cope

r-si

ci

232

(172

)8

20U

nkno

wn

Xyl

emPr

obab

lyno

tre

quir

edfo

rvi

rule

nce

Unk

now

nH

oute

rman

etal

.,20

07;

Ster

giop

oulo

s&

deW

it,20

09A

VR

2(S

IX3)

F.ox

yspo

-ru

mf.

sp.

lyco

per-

sici

163

(144

)3

(2)

19In

duce

sH

R,u

nkno

wn,

func

tiona

lR

XL

Rlik

em

otif

Xyl

emR

equi

red

for

full

viru

lenc

eI-

2H

oute

rman

etal

.,20

07;

Ster

giop

oulo

s&

deW

it,20

09;K

ale

etal

.,20

10A

VR

1(S

IX4)

F.ox

yspo

-ru

mf.

sp.

lyco

per-

sici

242

(184

)6

17U

nkno

wn

Xyl

emSu

ppre

ssio

nof

I-2

and

I-3

resi

stan

ce

Ior

I-1

Hou

term

anet

al.,

2007

;St

ergi

opou

los

&de

Wit,

2009

AT

R1N

dWsB

H. ar

abid

op-

sidi

s

311

(296

)–

15in

duce

sH

R,R

XL

Rdo

mai

nC

ytop

lasm

Supp

ress

host

defe

nse

RP

P1W

sBan

dR

PP

1Nd

Reh

man

yet

al.2

005;

Sohn

etal

.,20

07

AT

R13

H. ar

abid

op-

sidi

s

187

(168

)–

19In

duce

sH

R,R

XL

Rdo

mai

nC

ytop

lasm

Supp

ress

host

defe

nse

RP

P13

Alle

net

al.,

2004

;So

hnet

al.,

2007

AV

R1-

b1P.

soja

e13

8(1

17)

121

Indu

ces

HR

,sup

pres

ses

BA

X-i

nduc

edce

llde

ath,

RX

LR

dom

ain

Prob

ably

cyto

plas

mU

nkno

wn

Rps

1ban

dR

psk1

Shan

etal

.,20

04;

Dou

etal

.,20

08a,

b

AV

R1a

P.so

jae

121

(98)

–23

RX

LR

dom

ain

Unk

now

nR

ps1a

Qut

obet

al.,

2009

AV

R3a

P.so

jae

111

(91)

–20

RX

LR

dom

ain

Unk

now

nR

ps3a

Qut

obet

al.,

2009

AV

R3c

P.so

jae

220

(200

)1

20In

duce

sH

R,R

XL

Rdo

mai

nU

nkno

wn

Rps

3cD

ong

etal

.,20

09A

VR

3aP.

infe

stan

s14

7(1

26)

–21

Indu

ces

HR

,sup

pres

ses

INF1

-ind

uced

HR

,RX

LR

dom

ain,

inte

ract

with

CM

PG1

ubiq

uitin

E3

ligas

e

Cyt

opla

smR

equi

red

for

full

viru

lenc

eR

3aA

rmst

rong

etal

.,20

05;B

oset

al.,

2006

,200

9,20

10

MfA

VR

4M

.fiji

ensi

s12

1(1

00)

1021

Chi

tin-b

indi

ngpe

ritr

ophi

n-A

,in

duce

sH

Ror

necr

osis

inC

f4to

mat

o

Prob

ably

inap

opla

stPr

otec

tfun

giag

ains

tchi

tinas

esC

f4an

dH

cr9

Ster

giop

oulo

set

al.,

2010

MfE

CP2

M.fi

jien

sis

161

(142

)4

19In

duce

sH

Ror

necr

osis

inC

fEcp

2to

mat

oPr

obab

lyin

apop

last

Prom

ote

viru

lenc

eby

inte

ract

ing

with

host

cell

targ

etca

usin

gne

cros

is

Ecp

2St

ergi

opou

los

etal

.,20

10

Dow

nloa

ded

by [

Can

adia

n A

gric

ultu

re L

ibra

ry, A

gric

ultu

re a

nd A

gri-

Food

Can

ada]

at 1

3:23

08

Mar

ch 2

012

Page 11: Fungal and oomycete effectors – strategies to subdue a host · Can. J. Plant Pathol. (2011), 33(4): 425–446 Minireview/Minisynthèse Fungal and oomycete effectors – strategies

Fungal and oomycete effectors 435

recognized in tomato by Cf2, Cf4, Cf4E and Cf9 resis-tance proteins, respectively (Table 1; de Wit et al., 1997;Joosten & de Wit, 1999; Thomma et al., 2005). Thevirulence function of AVR2 and AVR4 has been dis-cussed earlier. AVR4E is a 101 amino acid (aa) proteinbut does not have a known virulence function (Westerinket al., 2004). Several field isolates that overcome Cf4E-mediated resistance reveal point mutations in AVR4E ora complete loss of the Avr4E gene, indicating that lossof this effector does not affect fitness of the pathogensignificantly (Stergiopoulos et al., 2007). Avr9 encodes a28 aa mature protein with six cysteine residues after it isprocessed by plant and fungal proteases at its C- and N-termini (van Kan et al., 1991; van den Ackerveken et al.,1993). Structurally, AVR9 is similar to carboxy-peptidaseinhibitor but no definitive function has been identified sofar (van Kan et al., 1991; van den Ackerveken et al., 1993;van den Hooven et al., 2001). All natural strains of C.fulvum that overcome Cf9 resistance lack Avr9, suggest-ing that it is not required for full virulence (Stergiopouloset al., 2007). C. fulvum deleted for Avr9 is fully viru-lent on tomato plants, but the heterologous expression ofAvr9 in tomato plants makes it more susceptible to C.fulvum strains lacking Avr9, indicating some (redundant)virulence function (Marmeisse et al., 1993; de Wit et al.,2009).

Besides Avr effectors, four extracellular cysteine-richproteins (ECP), such as ECP1, ECP2, ECP4 and ECP5,have been cloned and characterized from C. fulvum thatinduce an HR in tomato lines containing the correspond-ing Cf-Ecp resistance genes (Lauge et al., 2000; deKock et al., 2005). Ecp6 and Ecp7 have been clonedbut the corresponding tomato lines that recognize thesegenes have not yet been identified (Bolton et al., 2008).ECPs are present in all strains of C. fulvum and aresecreted during infection. They contain an even num-ber of cysteine residues that are most likely involved indisulphide bridge formation and secondary structure for-mation to protect them from apoplastic proteases (Ludereret al., 2002). Three of the ECPs, ECP1, ECP2 andECP6, have virulence functions on host plants, basedon data showing that deleting or suppressing expressionof these genes reduced virulence (Lauge et al., 1997;Bolton et al., 2008). Orthologs for AVR4 and ECP6 havebeen identified in several fungal species because of thepresence of CMB14 and lysin motif (LysM, carbohydrate-binding) domains in these proteins (Bolton et al., 2008).Orthologs of AVR4 and ECP2 have been identified inMycosphaerella fijiensis that causes black Sigatoka dis-ease of banana (Stergiopoulos et al., 2010). The M.fijiensis ortholog of AVR4 induces an HR in tomatolines containing the corresponding Cf4 gene and binds

to chitin of fungal cell walls to protect against cell walldegradation, similar to C. fulvum AVR4 (Stergiopouloset al., 2010). Similarly, M. fijiensis ECP2 is a functionalortholog of C. fulvum ECP2 and is recognized by Cf2 oftomato to induce an HR, while in the absence of Cf2, itpromotes virulence on tomato plants (Stergiopoulos et al.,2010).

Rynchosporium secalis

The imperfect fungus R. secalis causes leaf scald diseaseon barley by secreting low molecular-weight toxic pro-teins. The genes for three of these effectors, designatedas necrosis-inducing proteins NIP1, NIP2 and NIP3, havebeen cloned (Table 1). They encode small secreted pro-teins with toxicity in a genotype non-specific manner onbarley and related cereal plant species (Hahn et al., 1993;Rohe et al., 1995; Steiner-Lange et al., 2003). MatureNIP1 is a 60 aa protein with 10 cysteine residues thatare involved in intramolecular disulphide bond formation.NIP1 triggers specific defence responses without an HRon barley cultivars that have the corresponding resistancegene, Rrs1 (Lehnackers & Knogge, 1990). The injectionof NIP1 into leaves of barley and other cereal plant speciescauses scald-like lesion formation (Wevelsiep et al., 1991;van’t Slot et al., 2007). A nip1 disruption mutant of R.secalis is slightly less virulent than the wild type on sus-ceptible plants, demonstrating a (minor) role in virulence(Knogge & Marie 1997). Virulent strains of R. secalisovercome Rrs1 resistance of barley either by a point muta-tion in the ORF that results in a single aa substitution orby jettison of the Nip1 gene (Rohe et al., 1995). It hasbeen shown that NIP1 interacts with a single plasma mem-brane receptor (different from Rrs1) that is involved bothin necrosis and defence induction (van’t Slot et al., 2007).A field population study of this pathogen showed a posi-tive diversifying selection on the Nip1 locus, as three outof the 14 isoforms gained virulence on Rrs1 barley linesand a high deletion frequency was observed in the Nip1locus compared with Nip2 and Nip3 (de Wit et al., 2009).The deletion frequency of Nip1 was higher than the occur-rence of the point mutation that gains virulence indicatinga reduced fitness penalty for the loss of the Nip1 gene.Nip2 encodes a 109 aa protein with a predicted secre-tion signal of 16 aa and a mature protein with 6 cysteineresidues, while Nip3 encodes a 115 aa protein with a pre-dicted secretion signal of 17 aa and a mature protein with8 cysteine residues (de Wit et al., 2009).

Magnaporthe oryzae

M. oryzae (formerly known as M. grisea) is a filamentousascomycete fungus that causes rice blast disease,

Dow

nloa

ded

by [

Can

adia

n A

gric

ultu

re L

ibra

ry, A

gric

ultu

re a

nd A

gri-

Food

Can

ada]

at 1

3:23

08

Mar

ch 2

012

Page 12: Fungal and oomycete effectors – strategies to subdue a host · Can. J. Plant Pathol. (2011), 33(4): 425–446 Minireview/Minisynthèse Fungal and oomycete effectors – strategies

S. Ali and G. Bakkeren 436

destructive to rice production worldwide, but can alsocause disease in many other members of graminaceousplants (Kato et al., 2000; Couch & Kohn, 2002). Morethan 40 resistance genes have been identified in rice con-trolling the blast fungus and several of them have beenextensively used in resistant rice lines in the past fewdecades (Bryan et al., 2000; Chen et al., 2006). Theseresistant rice lines are overcome quickly in the field by theemergence of new races of the pathogen through variousmechanisms in the pathogen, such as deletion of Avr genesfrom the genome (Yoshida et al., 2009), changes in theirgene expression (Kang et al., 2001; Fudal et al., 2005) orpoint mutations in their genes (Orbach et al., 2000) result-ing in escaping recognition by R genes (Kolmer, 1989;Leach et al., 2001; McDonald & Linde, 2002). Eightcultivar- and species-specific Avr genes have been clonedand characterized from M. oryzae: Avr-Pita, Avr1-CO39,Ace1, Pwl1, Pwl2, AvrPiz-t, Avr-Pia, Avr-Pii and Avr-pik/km/kp (Table 1; Valent et al., 1991; Farman & Leong,1998; Orbach et al., 2000; Bohnert et al., 2004; Collemareet al., 2008; Li et al., 2009; Miki et al., 2009; Yoshidaet al., 2009).

The AVR-Pita effector shows similarity to fungal metal-loproteases of the deuterolysin family and is not requiredfor full virulence on rice plants (Jia et al., 2000; Orbachet al., 2000). Avr-Pita encodes a 223 aa protein thatis predicted to be secreted and needs to be processedinto a 176 aa active form (AVR-Pita 176) to interactdirectly with the LRR (leucine-rich repeat) domain ofthe PITA resistance protein and trigger PITA-mediateddefence responses, as assayed by yeast 2-hybrid assays, invitro binding analyses and direct expression in rice cells(Jia et al., 2000). In certain strains of M. oryzae, Avr-Pita undergoes spontaneous mutations in the laboratoryand also under field conditions, such as deletion, pointmutation, and the insertion of transposons, all resultingin overcoming Pita resistance in rice cultivars (Orbachet al., 2000; Kang et al., 2001; Zhou et al., 2007; Khanget al., 2008). Avr-Pita is located close to the telomereof chromosome 3 in the genome of M. oryzae and thismay be responsible for the genetic instability of thisgene. Avr-Pita was renamed Avr-Pita1 after identificationof Avr-Pita2 and Avr-Pita3 (Khang et al., 2008). Avr-Pita2 is recognized by the rice Pita gene and elicits thedefence response while Avr-Pita3 is not recognized byPita (Khang et al., 2008).

Avr1-Co39 was isolated from M. oryzae isolate 4091-5-8 pathogenic on weeping lovegrass and specifies avir-ulence on rice cultivar CO39 that contains the resistantgene, Pi-CO39(t) in a gene-for-gene manner (Valent et al.,1991; Chauhan et al., 2002). Isolates virulent on ricecultivar ‘CO-39’, lack Avr1-CO39 in most of the cases

(Farman et al., 2002). It has been shown that Avr1-CO39is a species-specific rather than a cultivar-specific typeof Avr gene (Zheng et al., 2011). The Pwl genes stopthis pathogen from causing disease on weeping lovegrassand finger millet in a species-specific manner, but theystill can infect rice (Kang et al., 1995; Sweigard et al.,1995). PWL effectors are small glycine-rich secreted pro-teins that are evolving fast and belong to a gene familydesignated as PWL1-PWL4. Pwl2 strains virulent onweeping lovegrass appear due to spontaneous mutations,predominantly by genetic rearrangement and large dele-tions (Kang et al., 1995; Sweigard et al., 1995). In thethree homologs of Pwl2, identified by homology searches,only Pwl1 is the functional homolog while Pwl3 and Pwl4are not functional; however, Pwl4 is functional only whenexpressed under the control of the Pwl1 or Pwl2 pro-moter, while Pwl3 is not functional in that case (Kanget al., 1995). Three new Avr genes, Avr-Pia, Avr-Pii andAvr-Pik/km/kp, have been isolated from M. oryzae byassociation genetics, i.e. by looking for polymorphismsamong 1032 predicted secreted proteins in field isolates(Yoshida et al., 2009).

An Avr gene from a different class, Ace1, encodes a4035 aa polyketide synthase (PKS) fused to a nonriboso-mal peptide synthetase (NRPS); these are two differentclasses of enzymes that are probably involved in theproduction of a secondary metabolite that triggers Pi33-mediated resistance in rice cultivars (Bohnert et al., 2004).M. grisea genome analysis revealed that Ace1 is present ina cluster of 15 genes of which 14 encode enzymes such asa second PKS-NRPS (Syn2), two enoyl reductases (Rap1and Rap2) and a putative Zn(II)(2)Cys(6) transcriptionfactor (BC2) which probably all play a role in secondarymetabolism (Collemare et al. 2008). Ace1 and all othergenes in the cluster are specifically expressed during pen-etration into the host plant, defining an infection-specificgene cluster, which suggests that Ace1 might have a rolein virulence; however, an Ace1 disruption mutant did notshow any reduction in virulence (Bohnert et al., 2004;Fudal et al., 2005).

Leptosphaeria maculans

This ascomycete fungus causes blackleg (phoma stemcanker) disease on oilseed rape (Brassica napus). Geneticanalysis of the interaction has revealed at least nineavirulence genes designated as AvrLm1–AvrLm9 that arerecognized by corresponding resistance genes Rlm1–Rlm9of the host (Balesdent et al., 2002; Rouxel & Balesdent,2005; Yu et al., 2005; Fitt et al., 2006). Seven ofthese nine genes are present in two unlinked clusters(AvrLm1-2-6 and AvrLm 3-4-7-9) while the remaining two

Dow

nloa

ded

by [

Can

adia

n A

gric

ultu

re L

ibra

ry, A

gric

ultu

re a

nd A

gri-

Food

Can

ada]

at 1

3:23

08

Mar

ch 2

012

Page 13: Fungal and oomycete effectors – strategies to subdue a host · Can. J. Plant Pathol. (2011), 33(4): 425–446 Minireview/Minisynthèse Fungal and oomycete effectors – strategies

Fungal and oomycete effectors 437

are individual genes (Balesdent et al., 2002). AvrLm1,AvrLm6 and AvrLm4-7 have been cloned by a map-basedstrategy and all encode small putative secreted proteinsthat have no similarity to sequences in public databases(Table 1; Gout et al., 2006; Fudal et al., 2007; Parlangeet al., 2009). Both AvrLm1 and AvrLm6 are located ina gene-poor, AT-rich, non-coding heterochromatin-likeregion as solo genes in stretches of 269 and 131 kb,respectively, which contain a number of degeneratednested copies of long terminal repeat (LTR) retrotrans-poson (Gout et al., 2006; Fudal et al., 2007). Also, bothAvrLm1 and AvrLm6 are single copy genes that encodesmall proteins of 205 and 144 aa, respectively, with anN-terminal secretion signal but no other conserved motifor any similarity to each other. The expression of bothgenes is strongly induced during early leaf infection butthey are also expressed in culture media at relatively lowlevels when not in contact with the host (Gout et al.,2006; Fudal et al., 2007). AvrLm1 has only one cysteineresidue and is likely taken up by the host cell (Gout et al.,2006), while AvrLm6 contains six cysteine residues thatmake disulphide bridges that could provide stability inthe apoplastic environment (Fudal et al., 2007). Strainsvirulent on Rlm1 oilseed rape cultivars lack the AvrLm1gene. Repeat-induced point (RIP) mutation, a commonmechanism to inactivate genes in ascomycetes, and dele-tion were also responsible for gain of virulence on Rlm1cultivars (Fudal et al., 2009). Similar to the AvrLm1 andAvrLm6 loci, the isolated AvrLm4-7 gene was located toa 238 kb genetic locus having multiple LTR retrotrans-posons; this gene induces a resistance response in plantshaving either Rlm7 or Rlm4. Within the locus, this singlegene is embedded in a gene-poor, AT-rich, recombination-deficient 60 kb isochor (Parlange et al., 2009). It encodesa small cysteine-rich secreted protein of 143 aa with a pre-dicted N-terminal secretion signal of 21 aa. Like othereffectors from this fungus, the expression of AvrLm4-7is induced in the plant but is also expressed at low lev-els in culture media (Parlange et al., 2009). The partial orcomplete loss of AvrLm4-7 in field isolates is responsiblefor gain of virulence on both Rlm4 and Rlm7 cultivars,while a point mutation that changes a glycine to an argi-nine residue, can overcome recognition by Rlm4 (Parlangeet al., 2009).

Fusarium oxysporum

F. oxysporum f. sp. lycopersici (Fol) infects tomato rootsvia wounds or by direct penetration after which it colo-nizes xylem vessels. Several small cysteine-rich proteinswere identified from xylem sap during Fol infection, andthree of them induced resistance in an R gene-specific

manner. They are coded for by avirulence gene SIX(secreted in xylem) 1 to 4 (Table 1; Rep et al., 2004;Houterman et al., 2007, 2008, 2009). Fusarium genomecomparisons revealed that all the SIX genes are locatedon the same chromosome that is absent from non-pathogenic isolates. The transfer of this chromosome tonon-pathogenic strains converted them to pathogens (Maet al., 2010). The function of three of these genes invirulence has been discussed in the previous section.

Blumeria graminis

Powdery mildews are biotrophic ascomycete fungi thatcause diseases on various mono- and dicotyledonous plantspecies, including food and feed crops and ornamentalplants (Bushnell, 2002). They are obligate biotrophs thatneed a living host for growth and reproduction and pro-duce intracellular feeding structures, the haustoria, in theepidermis of their host plants (Yarwood, 1957; Glawe,2008). Bgh causes powdery mildew on barley and isthe most thoroughly studied powdery mildew fungus.It interacts with its host in a ‘gene-for-gene’ manner(Both et al., 2005; Zhang et al., 2005). Genetic anal-ysis revealed that there are more than 85 dominant orsemi-dominant mildew (Ml) resistance genes that rec-ognize different Bgh races including 28 highly similargenes at the Mla locus on barley chromosome 5 (Jensenet al., 1980). Seven Mla genes at this locus have beencloned and they all encode closely related intracellularcoiled-coil, nucleotide-binding site, leucine-rich repeat(CC-NBS-LRR) type R proteins that recognize differ-ent Bgh avirulent effector proteins (Halterman et al.,2001; Halterman & Wise, 2004). Two of these effectors,AVRa10 and AVRk1, that are recognized by the bar-ley R proteins, Mla10 and Mlk1, respectively, inducingan HR, have been cloned; however, they are also vir-ulence factors in that they promote infection on sus-ceptible barley cultivars (Table 1; Ridout et al., 2006).Both Avr genes belong to multi-gene families that havemore than 30 paralogs and they also have orthologsin other forma speciales that are pathogenic on othergrasses (Ridout et al., 2006). The predicted AVRa10 andAVRk1 effectors lack an N-terminal secretion signal orhost targeting sequence, but it has been shown by flu-orescence microscopy that the Mla10 protein is presentintracellularly, both in the cytoplasm and the nucleus ofinvaded barley cells. Assuming a necessary interactionwith effectors, this means that AVRa10 is secreted fromthe fungus by non-endomembranous pathways and takenup by the cell by an as yet unknown mechanism (Bieriet al., 2004; Shen et al., 2007). Using a novel approachto gene silencing, Nowara et al. (2010) showed that by

Dow

nloa

ded

by [

Can

adia

n A

gric

ultu

re L

ibra

ry, A

gric

ultu

re a

nd A

gri-

Food

Can

ada]

at 1

3:23

08

Mar

ch 2

012

Page 14: Fungal and oomycete effectors – strategies to subdue a host · Can. J. Plant Pathol. (2011), 33(4): 425–446 Minireview/Minisynthèse Fungal and oomycete effectors – strategies

S. Ali and G. Bakkeren 438

expressing double-stranded or antisense RNA sequencesin barley but targeted to the Bgh-specific Avra10 andAvrk1 effector genes, they could reduce fungal develop-ment in hosts lacking the corresponding Mla10 resistancegene, confirming their role in virulence. Interestingly, noclear release from R-gene recognition could be seen bysilencing the Avra10 gene using this system which theytermed ‘Host-Induced Gene Silencing’ or HIGS. BesidesAVRa10 and AVRk1, two other Avr genes, Avra22 andAvra12, have been mapped recently (Skamnioti et al.,2008).

Melampsora lini

Another obligate biotrophic fungus, but one that belongsto the basidiomycetes, is M. lini that causes flax rustdisease not only in flax but also in other species ofthe genus Linum. A number of flax R proteins havebeen analysed; these are highly polymorphic cytoplasmic(Toll/Interleukin1 Receptor) TIR-NBS-LRR proteins thatrecognize effector proteins that are delivered into flax cellsduring colonization. This interaction triggers an HR in a‘gene-for-gene’ manner, arresting growth of the fungus(Lawrence et al., 2007). The genetic analysis of M. liniwith its host plant flax (the basis of the original ‘gene-for-gene’ theory by Flor in 1942) revealed at least 30Avr genes and corresponding R genes (Ellis et al., 1997).Several Avr genes have been cloned from M. lini, mainlyfrom four different loci: AvrL567, AvrM, AvrP123 andAvrP4 (Table 1). These genes encode haustoria-expressedsecreted proteins (HESPs), suggesting that they have vir-ulence functions, but elicit defence responses in hosts thathave the corresponding R genes (Catanzariti et al., 2006).The AvrL567 locus has a cluster of three polymorphicAvr genes: AvrL567A, AvrL567B and AvrL567C. All threeencode 127 aa mature proteins after cleavage of a 23 aaSP and are recognized directly by the L5, L6 and L7 pro-teins inside the cell (Dodds et al., 2004). The matureAVR proteins are highly polymorphic with at least oneor more aa substitutions in the exposed surface of theproteins, suggesting functional interactions (Ellis et al.,2007; Wang et al., 2007). Some of the isolates harbouringthese AVR proteins became virulent by defeating match-ing plant resistance genes, indicating that genes in theAvrL567 locus are under positive diversifying selection(Dodds et al., 2006). The analysis of six flax rust iso-lates revealed 12 members of the AvrL567 effector genefamily, including the three previously isolated AvrL567A-C genes. Seven of these caused avirulence while fivebelonged to virulent strains and could no longer be rec-ognized by L5, L6 or L7 (Dodds et al., 2006). TheAvrM gene family is a small family that consists of five

avirulence paralogs, AvrMA to AvrME, and one virulentone, avrM, which is not recognized by any known flaxR protein (Catanzariti et al., 2006). These AvrM proteinsdo not have any known homologs and are highly variableboth in sequence and size due to deletions or insertions ofDNA, or to premature termination of the protein becauseof the location of stop codons (Catanzariti et al., 2006).

AvrP123 is a small cysteine-rich protein that containsthe characteristic CX7CX6YX3CX2-3C signature of theKazal serine protease inhibitor family, suggesting its roleas an inhibitor of host proteases. This is similar to thefunction of C. fulvum AVR2 that inhibits the Rcr3 cysteineprotease in the tomato apoplast (Catanzariti et al., 2006).AvrP4 also encodes a small cysteine-rich protein of 67 aaafter cleavage of a 28 aa SP. The 28 aa C-terminal part ofAvrP4 has 6 cysteine residues with a spacing consensus ofa typical ‘cysteine-knotted’ peptide, similar to the C. ful-vum AVR9 protein (van den Hooven et al., 2001). AvrP4is expressed only in planta while AvrM is expressed bothin planta and in vitro (Stergiopoulos & de Wit, 2009).Agroinfiltration of AvrP4 and AvrM genes in flax plantswith matching resistance genes results in a HR indicat-ing that the produced effectors are functional in host cellsand are, therefore, likely translocated into the host dur-ing infections. This is in agreement with the predictedcytoplasmic location of P and M resistance proteins inflax (Anderson et al., 1997). Using a yeast 2-hybrid sys-tem, the AvrM-A and -D effectors were shown to interactdirectly with the M resistance protein resulting in fastnecrosis, and that a globular C-terminal domain, variableamong alleles, is important for this interaction (Catanzaritiet al., 2010).

Oomycete Avr (-triggering effector) genes

Oomycetes belong to the kingdom Stramenopila and areevolutionarily related to algae and include some well-known plant pathogens. Many effector genes have beenisolated, some of which have proven avirulence functions(Table 1).

Phytophthora infestans

P. infestans causes late blight disease in potato andtomato and was responsible for the ‘Irish Famine’ in1840. Many effectors have been identified and isolatedfrom P. infestans such as INF1 ‘elicitin’, a secreted pro-tein that elicits hypersensitive cell death and is used inmany current functional assays (Kamoun et al., 1997;Kamoun, 2006; Haas et al., 2009). However, only onerace-specific avirulence gene, Avr3a has been cloned.Avr3a encodes a cytoplasmic RXLR effector and when

Dow

nloa

ded

by [

Can

adia

n A

gric

ultu

re L

ibra

ry, A

gric

ultu

re a

nd A

gri-

Food

Can

ada]

at 1

3:23

08

Mar

ch 2

012

Page 15: Fungal and oomycete effectors – strategies to subdue a host · Can. J. Plant Pathol. (2011), 33(4): 425–446 Minireview/Minisynthèse Fungal and oomycete effectors – strategies

Fungal and oomycete effectors 439

transiently expressed lacking its SP, in potato cells, R3a-mediated cell death ensues illustrating its recognition inthe cytoplasm and avirulence function (Armstrong et al.,2005). The mature secreted protein is 147 aa in lengthand allelic polymorphic residues distinguishing function-ality were identified in various pathogen races: one allelicvariant Avr3AK80/I103 (abbreviated as Avr3A KI) causesavirulence on potato plants expressing R3a with the lysineresidue at position 80 being critical for recognition, whilethe virulent allele has two aa substitutions, glutamate atposition 80 and methionine at position 103 (designatedas Avr3AE80/M103 or Avr3AEM). In the absence of R3a,Avr3A KI strongly suppresses P. infestans INF1 elicitin-induced cell death in plants, thereby illustrating a majorvirulence function. The virulent allele Avr3a EM can-not induce an R3a-mediated HR but can still (weakly)suppress INF1-induced cell death suggesting that thiseffector has various parts with distinct functions thatlikely act through amino acid interactions with host pro-teins (Bos et al., 2006). These two distinct activitieshave been separated through saturated high-throughputmutant screens revealing important aa residues (Bos et al.,2009). Deletions or mutations in the C-terminal residueat tyrosine 147 maintained the R3A-mediated HR activ-ity while it caused the loss of the ability to suppressINF1-induced cell death. Molecular analysis of its celldeath-suppressing function showed that Avr3AKI interactsand stabilizes the host ubiquitin E3-ligase CMPG1 whichis required for INF1-induced cell death (Bos et al., 2010).The effector Avr3A likely targets CMPG1, interferingwith the ubiquitin proteasome system to block signaltransduction upon pathogen perception (Gilroy et al.,2011). P. infestans Avr3a is located in a region syn-tenic to a part in the genome of another oomycete,Hyaloperonospora arabidopsidis which harbours aviru-lence effector ATR1NdWsB (see below), suggesting thatthis locus is ancient in these oomycetes.

The P. infestans genome sequence revealed many pre-dicted secreted effectors and targeted searches for func-tion revealed novel Avr activities for some effectorsinteracting with specific resistance genes found in cer-tain potato species to induce hypersensitive cell death(Oh et al., 2009). Another secreted effector, SNE1, likelyis delivered into the plant host nucleus where it orches-trates the suppression of the effects of cell death-inducingeffectors secreted during the biotrophic phase (Kelleyet al., 2010).

Phytophthora sojae

P. sojae causes root and stem rot of soybean, resultingin huge damage to soybean production in North America

(Shan et al., 2004). Four avirulence genes, designatedAvr1b-1, Avr1a, Avr3a and Avr3c, have been cloned fromthis pathogen; they are recognized by soybean resis-tance genes Rps1b, Rps1a, Rps3a and Rps3c, respectively(Table 1; Shan et al., 2004; Dong et al., 2009; Qutob et al.,2009). Avr1b-1 was cloned by map-based cloning and ispredicted to encode a secreted protein of 138 aa witha RXLR motif. Interestingly, this gene requires anothergene, Avr1-b2, at the locus for accumulation of Avr1b-1 mRNA. Virulent P. sojae isolates such as P6497 andP9073, harbour a complete Avr1b-1 gene but cannot accu-mulate Avr1b-1 mRNA like avirulent strains (Shan et al.,2004). In addition to recognition by Rps1B, Avr1B-1 isalso recognized in plants having the Rpsk1 resistancegene resulting in limited cell death and showing that sucheffectors might have multiple targets (Kamoun, 2006).As R proteins from the Rps1 gene clusters are cytoplas-mic, it is assumed that Avr1-B is recognized inside thehost cytoplasm (Bhattacharyya et al., 1997; Shan et al.,2004).

P. sojae Avr1a, Avr3a and Avr3c were cloned througha combination of genetic mapping, transcript profiling,and functional analysis (Qutob et al., 2009). All P. sojaestrains, whether virulent or avirulent on Rps1 plants,contain the Avr1a gene which is present in four nearlyidentical copies of 5.2 kb. Virulence was attributed to dif-ferences in transcription of Avr1a in some strains whereasin other virulent strains, two of the multiple copies ofthe fragments containing Avr1a were additionally deleted(Qutob et al., 2009). Avr3a and four other predicted ORFswere also found in four duplicated copies, of a fragmentof 10.8 kb. Transcriptional silencing of these four Avr3acopies was found to be responsible for avoiding recogni-tion by Rps3 plants in some P. sojae virulent strains, whilein other virulent strains, three copies were deleted and thefourth one was transcriptionally silenced (Qutob et al.,2009). The Avr3c gene was located to a 33.7 kb fragmenttogether with eight other predicted ORFs and three near-identical copies of this fragment are present in a tandemarray in the P. sojae genome (Dong et al., 2009). Avr3cvirulent strains avoid recognition by Rps3A through spe-cific mutations in the effector and subsequent sequenceexchange between two copies of Avr3c in the arrays. Thisexample illustrates ways by which pathogens can evolvetheir effector repertoire as to avoid recognition by host Rgenes (Dong et al., 2009).

As for P. infestans, the release of the P. sojae genomesequence has allowed the large-scale analysis of candidateeffectors predicted to be secreted through their RXLR-dEER motif. Among 169 recently tested effectors, mostcould suppress cell death triggered by BAX, by othereffectors and/or the PAMP INF, while several caused

Dow

nloa

ded

by [

Can

adia

n A

gric

ultu

re L

ibra

ry, A

gric

ultu

re a

nd A

gri-

Food

Can

ada]

at 1

3:23

08

Mar

ch 2

012

Page 16: Fungal and oomycete effectors – strategies to subdue a host · Can. J. Plant Pathol. (2011), 33(4): 425–446 Minireview/Minisynthèse Fungal and oomycete effectors – strategies

S. Ali and G. Bakkeren 440

cell death themselves and are hence possible avirulencefactors (Wang et al., 2011).

Hyaloperonospora arabidopsidis

H. arabidopsidis, formerly known as (Hyalo)peronosporaparasitica, is an obligate oomycete pathogen that causesdowny mildew disease on the model plant Arabidopsis.From this pathogen, two avirulence genes have beencloned and characterized: (Arabidopsis thaliana recog-nized 1), Atr1 and Atr13 (Table 1). The product ofthe Atr1 gene is recognized by Arabidopsis R pro-teins from two ecotypes, Niederzenz (RPP1Nd) andWassilewskija (RPP1WsB) and hence the effector alleleis called Atr1NdWsB. Transient expression of Atr1NdWsB inArabidopsis leaves by particle bombardment triggered celldeath. Extensive searches for allelic variants found thatRPP1Nd recognized the product of a single allele of Atr1,while RPP1WsB recognized the products of four differentdiverged alleles and provided resistance to a wide rangeof isolates (Rehmany et al., 2005). Atr1NdWsB encodesa 311 aa protein with a predicted secretion signal anda conserved RXLR motif. Atr1NdWsB is a highly poly-morphic protein and six different alleles that differ inabout one-third of all residues were identified in eightisolates; intense diversifying selection and recombinationplayed an important role in the evolution of this locus(Rehmany et al., 2005; Kamoun, 2006). Recent molec-ular studies have shown that binding of RPP1 occurs atthe LRR domain and is a prerequisite for HR induction,while the actual induction is facilitated by the TIR domain(Krasileva et al., 2010).

ATR13 is recognized by resistance protein RPP13 andtriggers a HR when transiently expressed in Arabidopsiscells using particle bombardment. The ATR13 geneencodes a 187 aa effector protein for which no relatedsequences have been found in public databases (Allenet al., 2004). ATR13 revealed apparent domain structures:in addition to the N-terminal SP and an RXLR motif,it has a heptad leucine-isoleucine repeat motif that isrequired for RPP13 recognition, followed by an imper-fect direct repeat of 4 × 11 aa which lies between aaresidues 93 and 136. All pathogen Atr13 effector alle-les as well as identified alleles of the interacting hostRPP13 resistance gene reveal a high level of aa polymor-phisms among their protein products and apparently havebeen under intense diversifying selection suggesting a co-evolutionary arms race at these loci (Allen et al., 2004).Both ATR1 and ATR13 suppress basal defence responsesof host plants when delivered by the P. syringae T3SS,revealing a virulence function (Sohn et al., 2007).

Concluding remarks

Over the last two decades, the study of different fungal,oomycete and bacterial pathogen effectors and their func-tion during the interaction with host plants has led toremarkable insights into the molecular basis underpin-ning plant diseases. These effectors facilitate a pathogen’sentry into the host by suppressing defence responses atmultiple levels. Examples include interference with recog-nition of PAMPs by PRRs and/or downstream signallingthereby evading basal immunity that otherwise wouldbe sufficient to stop infection. Effectors also have rolesin establishing feeding interactions and/or nutrient leak-age from the host to the benefit of the pathogens. Theseeffectors function at the interface of pathogen and hostplant or inside plant cells. Inadvertently, when recognizedby components of the host surveillance system, they cantrigger defence responses, revealing avirulence or elici-tor functions. The sequencing of complete genomes ofmany bacterial, fungal, and oomycete plant pathogens andsubsequent computational analyses have revealed a myr-iad of effectors, many of which are potential avirulencegenes that can trigger ETI. These then have the poten-tial of revealing novel resistance potential, in particularresistance genes in varied germplasm if suitable assayscan be designed. This has become an important goal ofmany research laboratories and has already led to the dis-covery of novel resistance genes (van der Hoorn et al.,2000; Torto et al., 2003; Stergiopoulos et al., 2010). Theidentification of complete sets of pathogen effectors, theirfunctions, and their host targets will advance the knowl-edge of molecular plant–pathogen interactions and sparkthe design of novel disease control strategies.

References

ABEYSEKARA, N.S., FRIESEN, T.L., KELLER, B., & FARIS, J.D. (2009).Identification and characterization of a novel host-toxin interaction inthe wheat–Stagonospora nodorum pathosystem. Theor. Appl. Genet., 120,117–126.

AGRIOS, G.N. (2005). Plant pathology (5th ed.). San Diego, CA: AcademicPress.

ALLEN, R.L., BITTNER-EDDY, P.D., GRENVILLE-BRIGGS, L.J., MEITZ,J.C., REHMANY, A.P., ROSE, L.E. et al. (2004). Host–parasite coevolu-tionary conflict between Arabidopsis and downy mildew. Science, 306,1957–1960.

ANDERSON, P.A., LAWRENCE, G.J., MORRISH, B.C., AYLIFFE, M.A.,FINNEGAN, E.J., & ELLIS, J.G. (1997). Inactivation of the flax rustresistance gene M associated with loss of a repeated unit within theleucine-rich repeat coding region. Plant Cell, 9, 641–651.

ARMSTRONG, M.R., WHISSON, S.C., PRITCHARD, L., BOS, J.I., VENTER,E., AVROVA, A.O. et al. (2005). An ancestral oomycete locus containslate blight avirulence gene Avr3a, encoding a protein that is recognized inthe host cytoplasm. Proc. Natl. Acad. Sci. USA, 102, 7766–7771.

Dow

nloa

ded

by [

Can

adia

n A

gric

ultu

re L

ibra

ry, A

gric

ultu

re a

nd A

gri-

Food

Can

ada]

at 1

3:23

08

Mar

ch 2

012

Page 17: Fungal and oomycete effectors – strategies to subdue a host · Can. J. Plant Pathol. (2011), 33(4): 425–446 Minireview/Minisynthèse Fungal and oomycete effectors – strategies

Fungal and oomycete effectors 441

BALESDENT, M.H., ATTARD, A., KUHN, M.L., & ROUXEL, T. (2002). Newavirulence genes in the phytopathogenic fungus Leptosphaeria maculans.Phytopathology, 92, 1122–1133.

BARTNICKI-GARCIA, S. (1968). Cell wall chemistry, morphogenesis, andtaxonomy of fungi. Annu. Rev. Microbiol., 22, 87–108.

BASSE, C.W., BOCK, K., & BOLLER, T. (1992). Elicitors and suppressorsof the defense response in tomato cells. Purification and characterizationof glycopeptide elicitors and glycan suppressors generated by enzymaticcleavage of yeast invertase. J. Biol. Chem., 267, 10258–10265.

BENT, A.F., & MACKEY, D. (2007). Elicitors, effectors, and R genes:The new paradigm and a lifetime supply of questions. Annu. Rev.Phytopathol., 45, 399–436.

BHATTACHARYYA, M.K., GONZALES, R.A., KRAFT, M., & BUZZELL, R.I.(1997). A copia-like retrotransposon tgmr closely linked to the Rps1-kallele that confers race-specific resistance of soybean to Phytophthorasojae. Plant Mol. Biol., 34, 255–264.

BIERI, S., MAUCH, S., SHEN, Q.H., PEART, J., DEVOTO, A.,CASAIS, C. et al. (2004). RAR1 positively controls steady state levelsof barley MLA resistance proteins and enables sufficient MLA6 accumu-lation for effective resistance. Plant Cell, 16, 3480–3495.

BLOCKER, A.J., DEANE, J.E., VEENENDAAL, A.K.J., ROVERSI, P.,HODGKINSON, J.L., JOHNSON, S. et al. (2008). What’s the point ofthe type III secretion system needle? Proc. Natl. Acad. Sci. USA, 105,6507–6513.

BOHNERT, H.U., FUDAL, I., DIOH, W., THARREAU, D., NOTTEGHEM, J.L.,& LEBRUN, M.H. (2004). A putative polyketide synthase/peptide syn-thetase from Magnaporthe grisea signals pathogen attack to resistant rice.Plant Cell, 16, 2499–2513.

BOLTON, M.D., VAN ESSE, H.P., VOSSEN, J.H., DE JONGE, R.,STERGIOPOULOS, I., STULEMEIJER, I.J.E. et al. (2008). The novelCladosporium fulvum lysin motif effector ECP6 is a virulence factor withorthologues in other fungal species. Mol. Microbiol., 69, 119–136.

BOS, J.I.B., ARMSTRONG, M.R., GILROY, E.M., BOEVINK, P.C., HEIN,I., TAYLOR, R.M. et al. (2010). Phytophthora infestans effector Avr3a isessential for virulence and manipulates plant immunity by stabilizing hostE3 ligase CMPG1. Proc. Natl. Acad. Sci. USA, 107, 9909–9914.

BOS, J.I., CHAPARRO-GARCIA, A., QUESADA-OCAMPO, L.M.,MCSPADDEN GARDENER, B.B., & KAMOUN, S. (2009). Distinctamino acids of the Phytophthora infestans effector AVR3a conditionactivation of R3a hypersensitivity and suppression of cell death. Mol.Plant–Microbe Interact., 22, 269–281.

BOS, J.I.B., KANNEGANTI, T.-D., YOUNG, C., CAKIR, C., HUITEMA, E.,WIN, J. et al. (2006). The C-terminal half of Phytophthora infestansRXLR effector AVR3a is sufficient to trigger R3a-mediated hypersensi-tivity and suppress INF1-induced cell death in Nicotiana benthamiana.Plant J., 48, 165–176.

BOTH, M., CSUKAI, M., STUMPF, M.P., & SPANU, P.D. (2005). Geneexpression profiles of Blumeria graminis indicate dynamic changes to pri-mary metabolism during development of an obligate biotrophic pathogen.Plant Cell, 17, 2107–2122.

BOUWMEESTER, K., DE SAIN, M., WEIDE, R., GOUGET, A., KLAMER,S., CANUT, H. et al. (2011). The lectin receptor kinase lecrk-i.9 is anovel Phytophthora resistance component and a potential host target fora RXLR effector. PLoS Pathog 7, e1001327.

BROEKAERT, W.F., TERRAS, F.R., CAMMUE, B.P., & OSBORN, R.W.(1995). Plant defensins: Novel antimicrobial peptides as components ofthe host defense system. Plant Physiol., 108, 1353–1358.

BRYAN, G.T., WU, K.S., FARRALL, L., JIA, Y., HERSHEY, H.P.,MCADAMS, S.A. et al. (2000). tA single amino acid difference distin-guishes resistant and susceptible alleles of the rice blast resistance genePi-Ta. Plant Cell, 12, 2033–2046.

BUSHNELL, W.R. (2002). The role of powdery mildew research in under-standing host–parasite interaction: Past, present, and future. In R.R.Belanger, W.R. Bushnell, A.J. Dik, & T.L.W. Carver (Eds.), The powdery

mildews: a comprehensive treatise (pp. 1–12). St. Paul, MN: AmericanPhytopathological Society (APS Press).

CATANZARITI, A.M., DODDS, P.N., LAWRENCE, G.J., AYLIFFE, M.A., &ELLIS, J.G. (2006). Haustorially expressed secreted proteins from flaxrust are highly enriched for avirulence elicitors. Plant Cell, 18, 243–256.

CATANZARITI, A.-M., DODDS, P.N., VE, T., KOBE, B., ELLIS, J.G., &STASKAWICZ, B.J. (2010). The AVRM effector from flax rust has astructured c-terminal domain and interacts directly with the M resistanceprotein. Mol. Plant–Microbe Interact., 23, 49–57.

CHAUHAN, R.S., FARMAN, M.L., ZHANG, H.B., & LEONG, S.A. (2002).Genetic and physical mapping of a rice blast resistance locus, Pi-Co39(t),that corresponds to the avirulence gene Avr1-Co39 of Magnaporthegrisea. Mol. Genet. Genomics, 267, 603–612.

CHEN, Q.H., WANG, Y.C., & ZHENG, X.B. (2006). Genetic analysisand molecular mapping of the avirulence gene Pre1, a gene for host-species specificity in the blast fungus Magnaporthe grisea. Genome, 49,873–881.

CHISHOLM, S.T., COAKER, G., DAY, B., & STASKAWICZ, B.J. (2006).Host–microbe interactions: Shaping the evolution of the plant immuneresponse. Cell, 124, 803–814.

COLLEMARE, J., PIANFETTI, M., HOULLE, A.E., MORIN, D., CAMBORDE,L., GAGEY, M.J. et al. (2008). Magnaporthe grisea avirulence geneAce1 belongs to an infection-specific gene cluster involved in secondarymetabolism. New Phytol., 179, 196–208.

COLLINS, N.C., THORDAL-CHRISTENSEN, H., LIPKA, V., BAU, S.,KOMBRINK, E., QIU, J.L. et al. (2003). SNARE-protein-mediated diseaseresistance at the plant cell wall. Nature, 425, 973–977.

COUCH, B.C., & KOHN, L.M. (2002). A multilocus gene genealogy con-cordant with host preference indicates segregation of a new species,Magnaporthe oryzae, from M. grisea. Mycologia, 94, 683–693.

CUNNAC, S., LINDEBERG, M., & COLLMER, A. (2009). Pseudomonassyringae type III secretion system effectors: Repertoires in search offunctions. Curr. Opin. Microbiol., 12, 53–60.

DANGL, J.L., & JONES, J.D. (2001). Plant pathogens and integrated defenseresponses to infection. Nature, 411, 826–833.

DE JONGE, R., & THOMMA, B.P.H.J. (2009). Fungal lysM effectors:extinguishers of host immunity? Trends Microbiol. 17, 151–157.

DE KOCK, M.J.D., BRANDWAGT, B.F., BONNEMA, G., DE WIT, P.J.G.M.,& LINDHOUT, P. (2005). The tomato Orion locus comprises a uniqueclass of Hcr9 genes. Mol. Breeding, 15, 409–422.

DE TORRES, M., MANSFIELD, J.W., GRABOV, N., BROWN, I.R.,AMMOUNEH, H., TSIAMIS, G. et al. (2006). Pseudomonas syringaeeffector AVRPTOB suppresses basal defense in Arabidopsis. Plant J.,47, 368–382.

DE WIT, P.J. (2007). How plants recognize pathogens and defend them-selves. Cell Mol. Life Sci., 64, 2726–2732.

DE WIT, P.J., LAUGE, R., HONEE, G., JOOSTEN, M.H., VOSSEN, P.,KOOMAN-GERSMANN, M. et al. (1997). Molecular and biochemi-cal basis of the interaction between tomato and its fungal pathogenCladosporium fulvum. Antonie van Leeuwenhoek, 71, 137–141.

DE WIT, P.J., MEHRABI, R., VAN DEN BURG, H.A., & STERGIOPOULOS,I. (2009). Fungal effector proteins: Past, present and future. Mol. PlantPathol., 10, 735–747.

DEAKIN, W.J., & BROUGHTON, W.J. (2009). Symbiotic use of pathogenicstrategies: Rhizobial protein secretion systems. Nat. Rev. Microbiol.,7, 312–320.

DEAN, R.A., TALBOT, N.J., EBBOLE, D.J., FARMAN, M.L., MITCHELL,T.K., ORBACH, M.J. et al. (2005). The genome sequence of the rice blastfungus Magnaporthe grisea. Nature, 434, 980–986.

DESLANDES, L., OLIVIER, J., PEETERS, N., FENG, D.X.,KHOUNLOTHAM, M., BOUCHER, C. et al. (2003). Physical inter-action between RRS1-R, a protein conferring resistance to bacterial wilt,and PopP2, a type III effector targeted to the plant nucleus. Proc. Natl.Acad. Sci. USA, 100, 8024–8029.

Dow

nloa

ded

by [

Can

adia

n A

gric

ultu

re L

ibra

ry, A

gric

ultu

re a

nd A

gri-

Food

Can

ada]

at 1

3:23

08

Mar

ch 2

012

Page 18: Fungal and oomycete effectors – strategies to subdue a host · Can. J. Plant Pathol. (2011), 33(4): 425–446 Minireview/Minisynthèse Fungal and oomycete effectors – strategies

S. Ali and G. Bakkeren 442

DODDS, P.N., LAWRENCE, G.J., CATANZARITI, A.M., AYLIFFE, M.A., &ELLIS, J.G. (2004). The Melampsora lini Avrl567 avirulence genes areexpressed in haustoria and their products are recognized inside plant cells.Plant Cell, 16, 755–768.

DODDS, P.N., LAWRENCE, G.J., CATANZARITI, A.M., TEH, T., WANG,C.I., AYLIFFE, M.A. et al. (2006). Direct protein interaction underliesgene-for-gene specificity and coevolution of the flax resistance genes andflax rust avirulence genes. Proc. Natl. Acad. Sci. USA, 103, 8888–8893.

DOEHLEMANN, G., REISSMANN, S., ASSMANN, D., FLECKENSTEIN, M.,& KAHMANN, R. (2011). Two linked genes encoding a secreted effectorand a membrane protein are essential for Ustilago maydis-induced tumourformation. Mol. Microbiol. 81, 751–766.

DOEHLEMANN, G., VAN DER LINDE, K., ASSMANN, D., SCHWAMMBACH,D., HOF, A., MOHANTY, A. et al. (2009). Pep1, a secreted effectorprotein of Ustilago maydis, is required for successful invasion of plantcells.PLoS Pathog 5, e1000290.

DOEHLEMANN, G., WAHL, R., HORST, R.J., VOLL, L.M., USADEL, B.,POREE, F. et al. (2008). Reprogramming a maize plant: Transcriptionaland metabolic changes induced by the fungal biotroph Ustilago maydis.Plant J., 56, 181–195.

DONG, S., QUTOB, D., TEDMAN-JONES, J., KUFLU, K., WANG, Y.,TYLER, B.M. et al. (2009). The Phytophthora sojae avirulence locusAvr3c encodes a multi-copy RXLR effector with sequence polymor-phisms among pathogen strains. PloS One, 4, e5556.

DOU, D., KALE, S.D., WANG, X., CHEN, Y., WANG, Q., JIANG, R.H. et al.(2008a). Conserved C-terminal motifs required for avirulence and sup-pression of cell death by Phytophthora sojae effector Avr1b. Plant Cell,20, 1118–1133.

DOU, D., KALE, S.D., WANG, X., JIANG, R.H., BRUCE, N.A.,ARREDONDO, F.D. et al. (2008b). RXLR-mediated entry ofPhytophthora sojae effector AVR1b into soybean cells does notrequire pathogen-encoded machinery. Plant Cell, 20, 1930–1947.

DUPLESSIS, S., CUOMO, C.A., LIN, Y.-C., AERTS, A., TISSERANT, E.,VENEAULT-FOURREY, C. et al. (2011). Obligate biotrophy featuresunraveled by the genomic analysis of rust fungi. Proc. Natl. Acad. Sci.USA, 108, 9166–9171.

ELLIS, J., CATANZARITI, A.M., & DODDS, P. (2006). The problem of howfungal and oomycete avirulence proteins enter plant cells. Trends PlantSci., 11, 61–63.

ELLIS, J., LAWRENCE, G., AYLIFFE, M., ANDERSON, P., COLLINS, N.,FINNEGAN, J. et al. (1997). Advances in the molecular genetic analysisof the flax–flax rust interaction. Annu. Rev. Phytopathol., 35, 271–291.

ELLIS, J.G., LAWRENCE, G.J., & DODDS, P.N. (2007). Further analysis ofgene-for-gene disease resistance specificity in flax. Mol. Plant Pathol., 8,103–109.

ERBS, G., & NEWMAN, M.A. (2003). The role of lipopolysaccharides ininduction of plant defense responses. Mol. Plant. Pathol., 4, 421–425.

FARMAN, M.L., ETO, Y., NAKAO, T., TOSA, Y., NAKAYASHIKI, H.,MAYAMA, S. et al. (2002). Analysis of the structure of the Avr1-Co39 avirulence locus in virulent rice-infecting isolates of Magnaporthegrisea. Mol. Plant–Microbe Interact., 15, 6–16.

FARMAN, M.L., & LEONG, S.A. (1998). Chromosome walking to the Avr1-Co39 avirulence gene of Magnaporthe grisea: Discrepancy between thephysical and genetic maps. Genetics, 150, 1049–1058.

FELIX, G., & BOLLER, T. (2003). Molecular sensing of bacteria in plants.The highly conserved RNA-binding motif RNP-1 of bacterial cold shockproteins is recognized as an elicitor signal in tobacco. J. Biol. Chem., 278,6201–6208.

FELIX, G., DURAN, J.D., VOLKO, S., & BOLLER, T. (1999). Plants havea sensitive perception system for the most conserved domain of bacterialflagellin. Plant J., 18, 265–276.

FELLE, H.H. (1998). The apoplastic pH of the Zea mays root cortex as mea-sured with pH-sensitive microelectrodes: Aspects of regulation. J. Exp.Bot., 49, 987–995.

FITT, B., BRUN, H., BARBETTI, M., & RIMMER, S. (2006). World-wideimportance of phoma stem canker (Leptosphaeria maculans and L. biglo-bosa) on oilseed rape (Brassica napus) Eur. J. Plant. Pathol., 114,3–15.

FLOR, H.H. (1942). Inheritance of pathogenicity in melampsora lini.Phytopathology 32, 653–669.

FRIESEN, T.L., MEINHARDT, S.W., & FARIS, J.D. (2007). TheStagonospora nodorum–wheat pathosystem involves multiple proteina-ceous host-selective toxins and corresponding host sensitivity genes thatinteract in an inverse gene-for-gene manner. Plant J., 51, 681–692.

FU, Z.Q., GUO, M., JEONG, B.R., TIAN, F., ELTHON, T.E., CERNY, R.L.et al. (2007). A type III effector ADP-ribosylates RNA-binding proteinsand quells plant immunity. Nature, 447, 284–288.

FUDAL, I., BOHNERT, H.U., THARREAU, D., & LEBRUN, M.-H. (2005).Transposition of mine, a composite retrotransposon, in the avirulencegene Ace1 of the rice blast fungus Magnaporthe grisea. Fungal Genet.Biol., 42, 761.

FUDAL, I., ROSS, S., BRUN, H., BESNARD, A.-L., ERMEL, M., KUHN,M.-L. et al. (2009). Repeat-induced point mutation (RIP) as an alterna-tive mechanism of evolution toward virulence in Leptosphaeria maculans.Mol. Plant–Microbe Interact., 22, 932–941.

FUDAL, I., ROSS, S., GOUT, L., BLAISE, F., KUHN, M.-L., ECKERT, M.R.et al. (2007). Heterochromatin-like regions as ecological niches for avir-ulence genes in the Leptosphaeria maculans genome: Map-based cloningof AvrLm6. Mol. Plant–Microbe Interact., 20, 459–470.

GAN, P.H.P., RAFIQI, M., ELLIS, J.G., JONES, D.A., HARDHAM, A.R.,& DODDS, P.N. (2010). Lipid binding activities of flax rust AVRM andAVRL567 effectors. Plant Sig. Beh., 5, 1272–1275.

GIJZEN, M., & NURNBERGER, T. (2006). Nep1-like proteins from plantpathogens: Recruitment and diversification of the NPP1 domain acrosstaxa. Phytochemistry, 67, 1800–1807.

GILROY, E.M., TAYLOR, R.M., HEIN, I., BOEVINK, P., SADANANDOM,A., & BIRCH, P.R.J. (2011). CMPG1-dependent cell death follows per-ception of diverse pathogen elicitors at the host plasma membrane andis suppressed by Phytophthora infestans RXLR effector Avr3a. NewPhytologist, 190, 653–666.

GLAWE, D.A. (2008). The powdery mildews: A review of the world’s mostfamiliar (yet poorly known) plant pathogens. Annu. Rev. Phytopathol., 46,27–51.

GODFREY, D., BOHLENIUS, H., PEDERSEN, C., ZHANG, Z., EMMERSEN,J., & THORDAL-CHRISTENSEN, H. (2010). Powdery mildew and rustfungal effector candidates share N-terminal Y/F/WXC-motif. BMCGenomics, 11, 317.

GOHRE, V., & ROBATZEK, S. (2008). Breaking the barriers: Microbialeffector molecules subvert plant immunity. Annu. Rev. Phytopathol., 46,189–215.

GOUT, L., FUDAL, I., KUHN, M.-L., BLAISE, F., ECKERT, M., CATTOLICO,L. et al. (2006). Lost in the middle of nowhere: The Avrlm1 avirulencegene of the dothideomycete Leptosphaeria maculans. Mol. Microbiol.,60, 67–80.

GURLEBECK, D., THIEME, F., & BONAS, U. (2006). Type III effectorproteins from the plant pathogen Xanthomonas and their role in theinteraction with the host plant. J. Plant Physiol., 163, 233–235.

GUST, A.A., BISWAS, R., LENZ, H.D., RAUHUT, T., RANF, S.,KEMMERLING, B. et al. (2007). Bacteria-derived peptidoglycans consti-tute pathogen-associated molecular patterns triggering innate immunityin Arabidopsis. J. Biol. Chem., 282, 32338–32348.

HAAS, B.J., KAMOUN, S., ZODY, M.C., JIANG, R.H., HANDSAKER, R.E.,CANO, L.M. et al. (2009). Genome sequence and analysis of the Irishpotato famine pathogen Phytophthora infestans. Nature, 461, 393–398.

HAHN, M., JUNGLING, S., & KNOGGE, W. (1993). Cultivar-specificelicitation of barley defense reactions by the phytotoxic peptideNIP1 from Rhynchosporium secalis. Mol. Plant–Microbe Interact., 6,745–754.

Dow

nloa

ded

by [

Can

adia

n A

gric

ultu

re L

ibra

ry, A

gric

ultu

re a

nd A

gri-

Food

Can

ada]

at 1

3:23

08

Mar

ch 2

012

Page 19: Fungal and oomycete effectors – strategies to subdue a host · Can. J. Plant Pathol. (2011), 33(4): 425–446 Minireview/Minisynthèse Fungal and oomycete effectors – strategies

Fungal and oomycete effectors 443

HALTERMAN, D.A., & WISE, R.P. (2004). A single-amino acid substitutionin the sixth leucine-rich repeat of barley MLA6 and MLA13 allevi-ates dependence on RAR1 for disease resistance signaling. Plant J., 38,215–226.

HALTERMAN, D., ZHOU, F., WEI, F., WISE, R.P., & SCHULZE, L.P. (2001).The MLA6 coiled-coil, NBS-LRR protein confers AVRMLA6-dependentresistance specificity to Blumeria graminis f. sp. hordei in barley andwheat. Plant J., 25, 335–348.

HAMMERSCHMIDT, R. (1999). Phytoalexins: What have we learned after60 years? Annu. Rev. Phytopathol. 37, 285–306.

HANN, D.R., & RATHJEN, J.P. (2007). Early events in the pathogenicity ofPseudomonas syringae on Nicotiana benthamiana. Plant J., 49, 607–618.

HE, P., SHAN, L., LIN, N.C., MARTIN, G.B., KEMMERLING, B.,NURNBERGER, T. et al. (2006). Specific bacterial suppressors of MAMPsignaling upstream of mapkkk in Arabidopsis innate immunity. Cell, 125,563–575.

HEATH, M.C. (2002). Cellular interactions between biotrophic fungalpathogens and host or nonhost plants. Can. J. Plant Pathol., 24, 259–264.

HOUTERMAN, P.M., SPEIJER, D., DEKKER, H.L., DE KOSTER, C.G.,CORNELISSEN, B.J.C., & REP, M. (2007). The mixed xylem sap pro-teome of Fusarium oxysporum-infected tomato plants. Mol. Plant Pathol.,8, 215–221.

HOUTERMAN, P.M., CORNELISSEN, B.J., & REP, M. (2008). Suppressionof plant resistance gene-based immunity by a fungal effector. PLoSPathogens, 4, e1000061.

HOUTERMAN, P.M., MA, L., VAN OOIJEN, G., DE VROOMEN, M.J.,CORNELISSEN, B.J.C., TAKKEN, F.L.W. et al. (2009). The effector pro-tein AVR2 of the xylem-colonizing fungus Fusarium oxysporum activatesthe tomato resistance protein I-2 intracellularly. Plant J., 58, 970–978.

HUANG, C.-C., & LINDHOUT, P. (1997). Screening for resistance in wildlycopersicon species to Fusarium oxysporum f. sp. lycopersici race 1 andrace 2. Euphytica, 93, 145–153.

HUANG, G., GAO, B., MAIER, T., ALLEN, R., DAVIS, E.L., BAUM, T.J.et al. (2003). A profile of putative parasitism genes expressed in theoesophageal gland cells of the root-knot nematode Meloidogyne incog-nita. Mol. Plant–Microbe Interact., 16, 376–381.

HUCKELHOVEN, R. (2007). Cell wall-associated mechanisms of diseaseresistance and susceptibility. Annu. Rev. Phytopathol., 45, 101–127.

JAMIR, Y., GUO, M., OH, H.S., PETNICKI-OCWIEJA, T., CHEN, S., TANG,X. et al. (2004). Identification of Pseudomonas syringae type III effectorsthat can suppress programmed cell death in plants and yeast. Plant J.,37, 554–565.

JENSEN, J., JØRGENSEN, J.H., JENSEN, H.P., GIESE, H., & DOLL, H.(1980). Linkage of the hordein loci Hor1 and Hor2 with the powderymildew resistance loci Ml-k and Ml-a on barley chromosome 5. Theor.Appl. Genet., 58, 27–31.

JHA, G., RAJESHWARI, R., & SONTI, R.V. (2007). Functional interplaybetween two Xanthomonas oryzae pv. oryzae secretion systems in modu-lating virulence on rice. Mol. Plant–Microbe Interact., 20, 31–40.

JIA, Y., MCADAMS, S.A., BRYAN, G.T., HERSHEY, H.P., & VALENT, B.(2000). Direct interaction of resistance gene and avirulence gene productsconfers rice blast resistance. EMBO J., 19, 4004–4014.

JIANG, R.H., TRIPATHY, S., GOVERS, F., & TYLER, B.M. (2008). RXLReffector reservoir in two Phytophthora species is dominated by a singlerapidly evolving superfamily with more than 700 members. Proc. Natl.Acad. Sci. USA, 105, 4874–4879.

JONES, J.D., & DANGL, J.L. (2006). The plant immune system. Nature, 444,323–329.

JOOSTEN, M., COZIJNSEN, T.J., & DE WIT, P.J. (1994). Host resistance to afungal tomato pathogen lost by a single base-pair change in an avirulencegene. Nature, 367, 384–386.

JOOSTEN, M., & DE WIT, P.J. (1999). The tomato–Cladosporium fulvuminteraction: A versatile experimental system to study plant–pathogeninteractions. Annu. Rev. Phytopathol., 37, 335–367.

JOSHI, B.N., SAINANI, M.N., BASTAWADE, K.B., DESHPANDE, V.V.,GUPTA, V.S., & RANJEKAR, P.K. (1999). Pearl millet cysteine proteaseinhibitor. Eur. J. Biochem., 265, 556–563.

KALE, S.D., GU, B., CAPELLUTO, D.G., DOU, D., FELDMAN, E.,RUMORE, A. et al. (2010). External lipid PI3P mediates entry of eukary-otic pathogen effectors into plant and animal host cells. Cell, 142,284–295.

KAMOUN, S. (2006). A catalogue of the effector secretome of plantpathogenic oomycetes. Annu. Rev. Phytopathol., 44, 41–60.

KAMOUN, S. (2007). Groovy times: Filamentous pathogen effectorsrevealed. Curr. Opin. Plant Biol., 10, 358–365.

KAMOUN, S., VAN WEST, P., DE JONG, A.J., DE GROOT, K.E.,VLEESHOUWERS, V.G., & GOVERS, F. (1997). A gene encoding a pro-tein elicitor of Phytophthora infestans is down-regulated during infectionof potato. Mol. Plant–Microbe Interact., 10, 13–20.

KAMPER, J., KAHMANN, R., BOLKER, M., MA, L.-J., BREFORT, T.,SAVILLE, B.J. et al. (2006). Insights from the genome of the biotrophicfungal plant pathogen Ustilago maydis. Nature, 444, 97–101.

KANG, S., SWEIGARD, J.A., & VALENT, B. (1995). The PWL hostspecificity gene family in the blast fungus Magnaporthe grisea. Mol.Plant–Microbe Interact., 8, 939–948.

KANG, S., LEBRUN, M.H., FARRALL, L., & VALENT, B. (2001).Gain of virulence caused by insertion of a pot3 transposon in aMagnaporthe grisea avirulence gene. Mol. Plant–Microbe Interact., 14,671–674.

KANNEGANTI, T.-D., HUITEMA, E., CAKIR, C., & KAMOUN, S. (2006).Synergistic interactions of the plant cell death pathways induced byPhytophthora infestans NEP1-like protein PINPP1.1 and INF1 elicitin.Mol. Plant–Microbe Interact., 19, 854–863.

KASCHANI, F., SHABAB, M., BOZKURT, T., SHINDO, T., SCHORNACK,S., GU, C. et al. (2010). An effector-targeted protease contributes todefense against Phytophthora infestans and is under diversifying selectionin natural hosts. Plant Physiol., 154, 1794–1804.

KATO, H., YAMAMOTO, M., YAMAGUCHI-OZAKI, T., KADOUCHI, H.,IWAMOTO, Y., NAKAYASHIKI, H. et al. (2000). Pathogenicity, matingability and DNA restriction fragment length polymorphisms of pyriculariapopulations isolated from gramineae, bambusideae and zingiberaceaeplants. J. Gen. Plant Pathol., 66, 30–47.

KAY, S., HAHN, S., MAROIS, E., HAUSE, G., & BONAS, U. (2007). A bac-terial effector acts as a plant transcription factor and induces a cell sizeregulator. Science, 318, 648–651.

KELLEY, B.S., LEE, S.J., DAMASCENO, C.M., CHAKRAVARTHY, S.,KIM, B.D., MARTIN, G.B. et al. (2010). A secreted effector protein(SNE1) from Phytophthora infestans is a broadly acting suppressor ofprogrammed cell death. Plant J., 62, 357–366.

KEMEN, E., KEMEN, A.C., RAFIQI, M., HEMPEL, U., MENDGEN, K.,HAHN, M. et al. (2005). Identification of a protein from rust fungitransferred from haustoria into infected plant cells. Mol. Plant–MicrobeInteract., 18, 1130–1139.

KHANG, C.H., BERRUYER, R., GIRALDO, M.C., KANKANALA, P., PARK,S.-Y., CZYMMEK, K. et al. (2010). Translocation of Magnaporthe oryzaeeffectors into rice cells and their subsequent cell-to-cell movement. PlantCell, 22, 1388–1403.

KHANG, C.H., PARK, S.Y., LEE, Y.H., VALENT, B., & KANG, S. (2008).Genome organization and evolution of the Avr-Pita avirulence gene fam-ily in the Magnaporthe grisea species complex. Mol. Plant–MicrobeInteract., 21, 658–670.

KNOGGE, W., & MARIE, C., (1997). Molecular characterization of fungalavirulence., In I.R. CRUTE, E.B. HOLUB & J.J. BURDAN (Ed.), The gene-for-gene relationship in plant-parasite interactions., (pp. 329–346). NewYork: CAB International.

KOBAYASHI, I., KOBAYASHI, Y., & HARHAM, A.R. (1997). Inhibition ofrust-induced hypersensitive response in flax cells by the microtubuleinhibitor oryzalin. Aust. J. Plant Physiol., 24, 733–740.

Dow

nloa

ded

by [

Can

adia

n A

gric

ultu

re L

ibra

ry, A

gric

ultu

re a

nd A

gri-

Food

Can

ada]

at 1

3:23

08

Mar

ch 2

012

Page 20: Fungal and oomycete effectors – strategies to subdue a host · Can. J. Plant Pathol. (2011), 33(4): 425–446 Minireview/Minisynthèse Fungal and oomycete effectors – strategies

S. Ali and G. Bakkeren 444

KOLMER, J.A. (1989). Virulence and race dynamics of Puccinia recon-dita f. sp. tritici in Canada during 1956–1987. Phytopathology, 79,349–356.

KRASILEVA, K.V., DAHLBECK, D., & STASKAWICZ, B.J. (2010).Activation of an Arabidopsis resistance protein is specified by the inplanta association of its leucine-rich repeat domain with the cognateoomycete effector. Plant Cell, 22, 2444–2458.

KRUGER, W.M., PRITSCH, C., CHAO, S., & MUEHLBAUER, G.J. (2002).Functional and comparative bioinformatic analysis of expressed genesfrom wheat spikes infected with Fusarium graminearum. Mol. Plant–Microbe Interact., 15, 445–455.

KUNZE, G., ZIPFEL, C., ROBATZEK, S., NIEHAUS, K., BOLLER, T., &FELIX, G. (2004). The N terminus of bacterial elongation factor Tu elicitsinnate immunity in Arabidopsis plants. Plant Cell, 16, 3496–3507.

LAHAYE, T., & BONAS, U. (2001). Molecular secrets of bacterial type IIIeffector proteins. Trends Plant Sci., 6, 479–485.

LAUGE, R., GOODWIN, P.H., DE WIT, P.J., & JOOSTEN, M.H.(2000). Specific HR-associated recognition of secreted proteins fromCladosporium fulvum occurs in both host and non-host plants. Plant J.,23, 735–745.

LAUGE, R., JOOSTEN, M.H.A.J., VAN DE ACKERVEKEN, G.F.J.M., VAN

DEN BROEK, H.W.J., & DE WIT, P.J. (1997). The in planta-producedextracellular proteins ECP1 and ECP2 of Cladosporium fulvum arevirulence factors. Mol. Plant–Microbe Interact., 10, 725–734.

LAWRENCE, G.J., DODDS, P.N., & ELLIS, J.G. (2007). Rust of flax andlinseed caused by Melampsora lini. Mol. Plant Pathol., 8, 349–364.

LAWRENCE, G.J., DODDS, P.N. & ELLIS, J.G. (2010). Transformationof the flax rust fungus, Melampsora lini: selection via silencing of anavirulence gene. Plant J., 61, 364–369.

LEACH, J.E., VERA CRUZ, C.M., BAI, J., & LEUNG, H. (2001). Pathogenfitness penalty as a predictor of durability of disease resistance genes.Annu. Rev. Phytopathol., 39, 187–224.

LEHNACKERS, H., & KNOGGE, W. (1990). Cytological studies on theinfection of barley cultivars with known resistance genotypes byRhynchosporium secalis. Can. J. Bot., 68, 1953–1961.

LI, W., WANG, B., WU, J., LU, G., HU, Y., ZHANG, X. et al. (2009).The Magnaporthe oryzae avirulence gene Avrpiz-t encodes a predictedsecreted protein that triggers the immunity in rice mediated by the blastresistance gene Piz-t. Mol. Plant–Microbe Interact., 22, 411–420.

LIBAULT, M., WAN, J., CZECHOWSKI, T., UDVARDI, M., & STACEY,G. (2007). Identification of 118 Arabidopsis transcription factor and30 ubiquitin-ligase genes responding to chitin, a plant-defense elicitor.Mol. Plant–Microbe Interact., 20, 900–911.

LINDEBERG, M., CARTINHOUR, S., MYERS, C.R., SCHECHTER, L.M.,SCHNEIDER, D.J., & COLLMER, A. (2006). Closing the circle on the dis-covery of genes encoding Hrp regulon members and type III secretionsystem effectors in the genomes of three model Pseudomonas syringaestrains. Mol. Plant–Microbe Interact., 19, 1151–1158.

LIU, Z.H., FARIS, J.D., MEINHARDT, S.W., ALI, S., RASMUSSEN, J.B.,& FRIESEN, T.L. (2004). Genetic and physical mapping of a gene con-ditioning sensitivity in wheat to a partially purified host-selective toxinproduced by Stagonospora nodorum. Phytopathology, 94, 1056–1060.

LIU, Z., FRIESEN, T.L., LING, H., MEINHARDT, S.W., OLIVER, R.P.,RASMUSSEN, J.B. et al. (2006). The Tsn1–ToxA interaction in the wheat–Stagonospora nodorum pathosystem parallels that of the wheat–tan spotsystem. Genome, 49, 1265–1273.

LUCAS, J.A. (1998). Plant pathology and plant pathogens. Oxford, UK.:Blackwell Sci.

LUDERER, R., DE KOCK, M.J.D., DEES, R.H.L., DE WIT, P.J., & JOOSTEN,M.H.A.J. (2002). Functional analysis of cysteine residues of ECP elicitorproteins of the fungal tomato pathogen Cladosporium fulvum. Mol. PlantPathol., 3, 91–95.

MA, L.-J., VAN DER DOES, H.C., BORKOVICH, K.A., COLEMAN, J.J.,DABOUSSI, M.-J., DI PIETRO, A. et al. (2010). Comparative genomics

reveals mobile pathogenicity chromosomes in Fusarium. Nature, 464,367–373.

MANNING, V.A., HARDISON, L.K., & CIUFFETTI, L.M. (2007). PTRTOXA interacts with a chloroplast-localized protein. Mol. Plant–MicrobeInteract., 20, 168–177.

MANNING, V.A., CHU, A.L., STEEVES, J.E., WOLPERT, T.J., &CIUFFETTI, L.M. (2009). A host-selective toxin of Pyrenophora tritici-repentis, PTR TOXA, induces photosystem changes and reactive oxygenspecies accumulation in sensitive wheat. Mol. Plant–Microbe Interact., 2,665–676.

MARMEISSE, R., VAN DE ACKERVEKEN, G.F.J.M., GOOSEN, T., DE WIT,P.J., & VAN DEN BROEK, H.W.J. (1993). Disruption of the avirulencegene Avr9 in two races of the tomato pathogen Cladosporium fulvumcauses virulence on tomato genotypes with the complementary resistancegene Cf9. Mol. Plant–Microbe Interact., 6, 412–417.

MARTIN, F., AERTS, A., AHREN, D., BRUN, A., DANCHIN, E.G.J.,DUCHAUSSOY, F. et al. (2008). The genome of Laccaria bicolor providesinsights into mycorrhizal symbiosis. Nature, 452, 88–92.

MARTIN, F., & SELOSSE, M.-A. (2008). The Laccaria genome: A symbiontblueprint decoded. New Phytol., 180, 296–310.

MCDONALD, B.A., & LINDE, C. (2002). Pathogen population genetics,evolutionary potential, and durable resistance. Annu. Rev. Phytopathol.,40, 349–379.

MELOTTO, M., UNDERWOOD, W., KOCZAN, J., NOMURA, K., & HE,S.Y. (2006). Plant stomata function in innate immunity against bacterialinvasion. Cell, 126, 969.

MEYER, A., PUHLER, A., & NIEHAUS, K. (2001). The lipopolysaccharidesof the phytopathogen Xanthomonas campestris pv. campestris induce anoxidative burst reaction in cell cultures of Nicotiana tabacum. Planta,213, 214–222.

MIKI, S., MATSUI, K., KITO, H., OTSUKA, K., ASHIZAWA, T., YASUDA,N. et al. (2009). Molecular cloning and characterization of the Avr-Pialocus from a Japanese field isolate of Magnaporthe oryzae. Mol. PlantPathol., 10, 361–374.

MORRISSEY, J.P., & OSBOURN, A.E. (1999). Fungal resistance to plantantibiotics as a mechanism of pathogenesis. Microbiol. Mol. Biol. Rev.,63, 708–724.

MOSQUERA, G., GIRALDO, M.C., KHANG, C.H., COUGHLAN, S.,& VALENT, B. (2009). Interaction transcriptome analysis identifiesMagnaporthe oryzae bas1-4 as biotrophy-associated secreted proteins inrice blast disease. Plant Cell, 21, 1273–1290.

MUDGETT, M.B. (2005). New insights to the function of phytopathogenicbacterial type III effectors in plants. Annu. Rev. Plant Biol., 56, 509–531.

MUELLER, O., KAHMANN, R., AGUILAR, G., TREJO-AGUILAR, B., WU,A., & DE VRIES, R.P. (2008). The secretome of the maize pathogenUstilago maydis. Fungal Genet. Biol., 45, S63–S70.

MYSORE, K.S., & RYU, C.M. (2004). Nonhost resistance: How much do weknow? Trends Plant Sci., 9, 97–104.

NOWARA, D., GAY, A., LACOMME, C., SHAW, J., RIDOUT, C.,DOUCHKOV, D. et al. (2010). HIGS: Host-induced gene silencing in theobligate biotrophic fungal pathogen Blumeria graminis. Plant Cell, 22,3130–3141.

OH, H.S., KVITKO, B.H., MORELLO, J.E., & COLLMER, A. (2007).Pseudomonas syringae lytic transglycosylases coregulated with the typeIII secretion system contribute to the translocation of effector proteins intoplant cells. J. Bacteriol., 189, 8277–8289.

OH, S.-K., YOUNG, C., LEE, M., OLIVA, R., BOZKURT, T.O., CANO, L.M.et al. (2009). In planta expression screens of Phytophthora infestansRXLR effectors reveal diverse phenotypes, including activation of thesolanum bulbocastanum disease resistance protein Rpi-blb2. Plant Cell,21, 2928–2947.

OORT, A.J.P. (1944). Onderzoekingen over stuifbrand II. Overgevoeligheidvan tarwe voor stuifbrand, Ustilago tritici. Hypersensitiveness of wheatto loose smut. Tijdschrift Plantenziekten, 50, 73–106.

Dow

nloa

ded

by [

Can

adia

n A

gric

ultu

re L

ibra

ry, A

gric

ultu

re a

nd A

gri-

Food

Can

ada]

at 1

3:23

08

Mar

ch 2

012

Page 21: Fungal and oomycete effectors – strategies to subdue a host · Can. J. Plant Pathol. (2011), 33(4): 425–446 Minireview/Minisynthèse Fungal and oomycete effectors – strategies

Fungal and oomycete effectors 445

ORBACH, M.J., FARRALL, L., SWEIGARD, J.A., CHUMLEY, F.G., &VALENT, B. (2000). A telomeric avirulence gene determines efficacy forthe rice blast resistance gene Pi-ta. Plant Cell, 12, 2019–2032.

OSBOURN, A.E., BOWYER, P., & DANIELS, M.J. (1996). Saponin detoxifi-cation by plant pathogenic fungi. Adv. Exp. Med. Biol., 404, 547–555.

OTTMANN, C., LUBERACKI, B., KÜFNER, I., KOCH, W., BRUNNER,F.d.r., WEYAND, M. et al. (2009). A common toxin fold mediatesmicrobial attack and plant defense. Proc. Natl. Acad. Sci. USA, 106,10359–10364.

PARLANGE, F., DAVERDIN, G., FUDAL, I., KUHN, M.L., BALESDENT,M.H., BLAISE, F. et al. (2009). Leptosphaeria maculans avirulencegene Avrlm4-7 confers dual recognition specificity by the RLM4 andRLM7 resistance genes of oilseed rape, and circumvents RLM4-mediatedrecognition through a single amino acid change. Mol. Microbiol., 71,851–863.

PEDRAS, M.S.C., & AHIAHONU, P.W.K. (2005). Metabolism and detox-ification of phytoalexins and analogs by phytopathogenic fungi.Phytochemistry, 66, 391.

PEMBERTON, C.L., & SALMOND, G.P.C. (2004). The NEP1-like proteins, agrowing family of microbial elicitors of plant necrosis. Mol. Plant Pathol.,5, 353–359.

PLETT, J.M., KEMPPAINEN, M., KALE, S.D., KOHLER, A., LEGUÉ, V.,BRUN, A. et al. (2011). A secreted effector protein of Laccaria bicolor isrequired for symbiosis development. Curr. Biol., 21, 1197–1203.

PRYCE-JONES, E., CARVER, T.I.M., & GURR, S.J. (1999). The roles ofcellulase enzymes and mechanical force in host penetration by Erysiphegraminis f. sp. hordei. Physiol. Mol. Plant Pathol., 55, 175–182.

QUTOB, D., KAMOUN, S., & GIJZEN, M. (2002). Expression of aPhytophthora sojae necrosis-inducing protein occurs during transitionfrom biotrophy to necrotrophy. Plant J., 32, 361–373.

QUTOB, D., TEDMAN-JONES, J., DONG, S., KUFLU, K., PHAM, H., WANG,Y. et al. (2009). Copy number variation and transcriptional polymor-phisms of Phytophthora sojae RXLR effector genes Avr1a and Avr3a.PLoS ONE, 4, e5066.

RAFIQI, M., GAN, P.H.P., RAVENSDALE, M., LAWRENCE, G.J., ELLIS,J.G., JONES, D.A. et al. (2010). Internalization of flax rust avirulenceproteins into flax and tobacco cells can occur in the absence of thepathogen. Plant Cell, 22, 2017–2032.

REHMANY, A.P., GORDON, A., ROSE, L.E., ALLEN, R.L., ARMSTRONG,M.R., WHISSON, S.C. et al. (2005). Differential recognition of highlydivergent downy mildew avirulence gene alleles by RPP1 resistance genesfrom two Arabidopsis lines. Plant Cell, 17, 1839–1850.

REN, Y.Y., & WEST, C.A. (1992). Elicitation of diterpene biosynthesis inrice (Oryza sativa L.) by chitin. Plant Physiol., 99, 1169–1178.

REP, M., MEIJER, M., HOUTERMAN, P.M., VAN DER DOES, H.C., &CORNELISSEN, B.J. (2005). Fusarium oxysporum evades I-3-mediatedresistance without altering the matching avirulence gene. Mol. Plant–Microbe Interact., 18, 15–23.

REP, M., VAN DER DOES, H.C., MEIJER, M., VAN WIJK, R., HOUTERMAN,P.M., DEKKER, H.L. et al. (2004). A small, cysteine-rich protein secretedby Fusarium oxysporum during colonization of xylem vessels is requiredfor I-3-mediated resistance in tomato. Mol. Microbiol., 53, 1373–1383.

RIDOUT, C.J., SKAMNIOTI, P., PORRITT, O., SACRISTAN, S., JONES, J.D.,& BROWN, J.K. (2006). Multiple avirulence paralogues in cereal pow-dery mildew fungi may contribute to parasite fitness and defeat of plantresistance. Plant Cell, 18, 2402–2414.

ROHE, M., GIERLICH, A., HERMANN, H., HAHN, M., SCHMIDT, B.,ROSAHL, S. et al. (1995). The race-specific elicitor, NIP1, from the barleypathogen, Rhynchosporium secalis, determines avirulence on host plantsof the RRS1 resistance genotype. EMBO J., 14, 4168–4177.

ROONEY, H.C., VAN ’T KLOOSTER, J.W., VAN DER HOORN, R.A.,JOOSTEN, M.H., JONES, J.D., & DE WIT, P.J. (2005). CladosporiumAVR2 inhibits tomato RCR3 protease required for Cf2-dependent diseaseresistance. Science, 308, 1783–1786.

ROSE, J.K., HAM, K.S., DARVILL, A.G., & ALBERSHEIM, P. (2002).Molecular cloning and characterization of glucanase inhibitor proteins:Coevolution of a counterdefense mechanism by plant pathogens. PlantCell, 14, 1329–1345.

ROUXEL, T., & BALESDENT, M.H. (2005). The stem canker (blackleg) fun-gus, Leptosphaeria maculans, enters the genomic era. Mol. Plant Pathol.,6, 225–241.

SARMA, G.N., MANNING, V.A., CIUFFETTI, L.M., & KARPLUS, P.A.(2005). Structure of PTR TOXA: An RGD-containing host-selective toxinfrom Pyrenophora tritici-repentis. Plant Cell, 17, 3190–3202.

SCHIRAWSKI, J., MANNHAUPT, G., MUNCH, K., BREFORT, T., SCHIPPER,K., DOEHLEMANN, G. et al. (2010). Pathogenicity determinants in smutfungi revealed by genome comparison. Science, 330, 1546–1548.

SHABAB, M., SHINDO, T., GU, C., KASCHANI, F., PANSURIYA, T.,CHINTHA, R. et al. (2008). Fungal effector protein AVR2 targets diversi-fying defense-related Cys proteases of tomato. Plant Cell, 20, 1169–1183.

SHAN, W., CAO, M., LEUNG, D., & TYLER, B.M. (2004). The Avr1b locusof Phytophthora sojae encodes an elicitor and a regulator required foravirulence on soybean plants carrying resistance gene RPS1b. Mol. Plant–Microbe Interact., 17, 394–403.

SHARP, J.K., MCNEIL, M., & ALBERSHEIM, P. (1984a). The pri-mary structures of one elicitor-active and seven elicitor-inactivehexa(beta-d-glucopyranosyl)-d-glucitols isolated from the mycelial wallsof Phytophthora megasperma f. sp. glycinea. J. Biol. Chem., 259,11321–11336.

SHARP, J.K., VALENT, B., & ALBERSHEIM, P. (1984b). Purification andpartial characterization of a beta-glucan fragment that elicits phytoalexinaccumulation in soybean. J. Biol. Chem., 259, 11312–11320.

SHEN, Q.H., SAIJO, Y., MAUCH, S., BISKUP, C., BIERI, S., KELLER,B. et al. (2007). Nuclear activity of MLA immune receptors linksisolate-specific and basal disease-resistance responses. Science, 315,1098–1103.

SHIU, S.H., & BLEECKER, A.B. (2003). Expansion of the receptor-likekinase/pelle gene family and receptor-like proteins in Arabidopsis. PlantPhysiol., 132, 530–543.

SINGH, R.P., & RAJARAM, S. (1992). Genetics of adult-plant resistance toleaf rust in ‘Frontana’ and three CIMMYT wheats. Genome, 35, 24–31.

SKAMNIOTI, P., PEDERSEN, C., AL-CHAARANI, G.R., HOLEFORS, A.,THORDAL-CHRISTENSEN, H., BROWN, J.K.M. et al. (2008). Genetics ofavirulence genes in Blumeria graminis f. sp. hordei and physical mappingof Avra22 and Avra12. Fungal Genet. Biol., 45, 243–252.

SKIBBE, D.S., DOEHLEMANN, G., FERNANDES, J., & WALBOT, V. (2010).Maize tumors caused by Ustilago maydis require organ-specific genes inhost and pathogen. Science, 328, 89–92.

SOHN, K.H., LEI, R., NEMRI, A., & JONES, J.D. (2007). The downy mildeweffector proteins ATR1 and ATR13 promote disease susceptibility inArabidopsis thaliana. Plant Cell, 19, 4077–4090.

SONG, J., WIN, J., TIAN, M., SCHORNACK, S., KASCHANI, F., ILYAS, M. etal. (2009). Apoplastic effectors secreted by two unrelated eukaryotic plantpathogens target the tomato defense protease RCR3. Proc. Natl. Acad.Sci. USA, 106, 1654–1659.

STASKAWICZ, B.J., DAHLBECK, D., & KEEN, N.T. (1984). Cloned avir-ulence gene of Pseudomonas syringae pv. glycinea determines race-specific incompatibility on glycine max (L.) Merr. Proc. Natl. Acad. Sci.USA, 81, 6024–6028.

Steiner-Lange, S., FISCHER, A., BOETTCHER, A., ROUHARA, I.,LIEDGENS, H., SCHMELZER, E. et al. (2003). Differential defense reac-tions in leaf tissues of barley in response to infection by Rhynchosporiumsecalis and to treatment with a fungal avirulence gene product. Mol.Plant–Microbe Interact., 16, 893–902.

STERGIOPOULOS, I., DE KOCK, M.J., LINDHOUT, P., & DE WIT, P.J.(2007). Allelic variation in the effector genes of the tomato pathogenCladosporium fulvum reveals different modes of adaptive evolution. Mol.Plant–Microbe Interact., 20, 1271–1283.

Dow

nloa

ded

by [

Can

adia

n A

gric

ultu

re L

ibra

ry, A

gric

ultu

re a

nd A

gri-

Food

Can

ada]

at 1

3:23

08

Mar

ch 2

012

Page 22: Fungal and oomycete effectors – strategies to subdue a host · Can. J. Plant Pathol. (2011), 33(4): 425–446 Minireview/Minisynthèse Fungal and oomycete effectors – strategies

S. Ali and G. Bakkeren 446

STERGIOPOULOS, I., & DE WIT, P.J. (2009). Fungal effector proteins.Annu. Rev. Phytopathol., 47, 233–263.

STERGIOPOULOS, I., VAN DEN BURG, H.A., ÖKMEN, B., BEENEN, H.G.,VAN LIERE, S., KEMA, G.H.J. et al. (2010). Tomato Cf resistance pro-teins mediate recognition of cognate homologous effectors from fungipathogenic on dicots and monocots. Proc. Natl. Acad. Sci. USA, 107,7610–7615.

SWEIGARD, J.A., CARROLL, A.M., KANG, S., FARRALL, L., CHUMLEY,F.G., & VALENT, B. (1995). Identification, cloning, and characterizationof PWL2, a gene for host species specificity in the rice blast fungus. PlantCell, 7, 1221–1233.

SZUREK, B., ROSSIER, O., HAUSE, G., & BONAS, U. (2002). Type III-dependent translocation of the Xanthomonas AVRBS3 protein into theplant cell. Mol. Microbiol., 46, 13–23.

TALBOT, N.J. (2003). On the trail of a cereal killer: Exploring the biologyof Magnaporthe grisea. Annu. Rev. Microbiol., 57, 177–202.

THOMMA, B.P., VAN ESSE, H.P., CROUS, P.W., & DE WIT, P.J.G.M.(2005). Cladosporium fulvum (syn. Passalora fulva), a highly specializedplant pathogen as a model for functional studies on plant pathogenicmycosphaerellaceae. Mol. Plant Pathol., 6, 379–393.

TIAN, M., HUITEMA, E., DA CUNHA, L., TORTO-ALALIBO, T., &KAMOUN, S. (2004). A Kazal-like extracellular serine protease inhibitorfrom Phytophthora infestans targets the tomato pathogenesis-relatedprotease P69B. J. Biol. Chem., 279, 26370–26377.

TIAN, M., & KAMOUN, S. (2005). A two disulfide bridge Kazal domainfrom Phytophthora exhibits stable inhibitory activity against serine pro-teases of the subtilisin family. BMC Biochem., 6, 15.

TIAN, M., WIN, J., SONG, J., VAN DER HOORN, R., VAN DER KNAAP, E., &KAMOUN, S. (2007). A Phytophthora infestans cystatin-like protein tar-gets a novel tomato papain-like apoplastic protease. Plant Physiol., 143,364–377.

TORTO, T.A., LI, S., STYER, A., HUITEMA, E., TESTA, A., GOW, N.A. etal. (2003). EST mining and functional expression assays identify extra-cellular effector proteins from the plant pathogen Phytophthora. GenomeRes., 13, 1675–1685.

TYLER, B.M., TRIPATHY, S., ZHANG, X., DEHAL, P., JIANG, R.H., AERTS,A. et al. (2006). Phytophthora genome sequences uncover evolutionaryorigins and mechanisms of pathogenesis. Science, 313, 1261–1266.

VALENT, B., FARRALL, L. & CHUMLEY, F.G. (1991). Magnaporthe griseagenes for pathogenicity and virulence identified through a series ofbackcrosses. Genetics, 127, 87–101.

VAN DEN ACKERVEKEN, G.F., VAN KAN, J.A., JOOSTEN, M.H.,MUISERS, J.M., VERBAKEL, H.M., & DE WIT, P.J. (1993).Characterization of two putative pathogenicity genes of the fungaltomato pathogen Cladosporium fulvum. Mol. Plant–Microbe Interact.,6, 210–215.

VAN DEN BURG, H.A., HARRISON, S.J., JOOSTEN, M.H., VERVOORT,J., & DE WIT, P.J. (2006). Cladosporium fulvum AVR4 protects fungalcell walls against hydrolysis by plant chitinases accumulating duringinfection. Mol. Plant–Microbe Interact., 19, 1420–1430.

VAN DEN HOOVEN, H.W., VAN DEN BURG, H.A., VOSSEN, P., BOEREN,S., DE WIT, P.J., & VERVOORT, J. (2001). Disulfide bond structure ofthe AVR9 elicitor of the fungal tomato pathogen Cladosporium fulvum:Evidence for a cystine knot. Biochemistry, 40, 3458–3466.

VAN DER HOORN, R.A., LAURENT, F., ROTH, R., & DE WIT, P.J. (2000).Agroinfiltration is a versatile tool that facilitates comparative analysesof AVR9/Cf-9-induced and AVR4/CF-4-induced necrosis. Mol. Plant–Microbe Interact., 13, 439–446.

VAN ESSE, H.P., VAN ’T KLOOSTER, J.W., BOLTON, M.D., YADETA, K.A.,VAN BAARLEN, P., BOEREN, S. et al. (2008). The Cladosporium fulvum

virulence protein AVR2 inhibits host proteases required for basal defense.Plant Cell, 20, 1948–1963.

VAN KAN, J.A., VAN DEN ACKERVEKEN, G.F., & DE WIT, P.J. (1991).Cloning and characterization of cDNA of avirulence gene Avr9 of thefungal pathogen Cladosporium fulvum, causal agent of tomato leaf mold.Mol. Plant–Microbe Interact., 4, 52–59.

VAN LOON, L.C., REP, M. & PIETERSE, C.M. (2006). Significanceof inducible defense-related proteins in infected plants. Annu. Rev.Phytopathol. 44, 135–162.

VAN ’T SLOT, K.A., GIERLICH, A., & KNOGGE, W. (2007). A singlebinding site mediates resistance- and disease-associated activities of theeffector protein NIP1 from the barley pathogen Rhynchosporium secalis.Plant Physiol., 144, 1654–1666.

VAN ’T SLOT, K.A.E., & KNOGGE, W. (2002). A dual role for microbialpathogen-derived effector proteins in plant disease and resistance. Crit.Rev. Plant Sci., 21, 229–271.

WANG, C.I., GUNCAR, G., FORWOOD, J.K., TEH, T., CATANZARITI, A.M.,LAWRENCE, G.J. et al. (2007). Crystal structures of flax rust avirulenceproteins AVRL567-A and -D reveal details of the structural basis for flaxdisease resistance specificity. Plant Cell, 19, 2898–2912.

WANG, Q., HAN, C., FERREIRA, A.O., YU, X., YE, W., TRIPATHY, S.et al. (2011). Transcriptional programming and functional interactionswithin the Phytophthora sojae RXLR effector repertoire. Plant Cell, 23,2064–2086.

WESTERINK, N., BRANDWAGT, B.F., DE WIT, P.J., & JOOSTEN, M.H.(2004). Cladosporium fulvum circumvents the second functional resis-tance gene homologue at the Cf4 locus (Hcr9-4E) by secretion of a stableavr4E isoform. Mol. Microbiol., 54, 533–545.

WEVELSIEP, L., KOGEL, K.H., & KNOGGE, W. (1991). Purificationand characterization of peptides from Rhynchosporium secalis inducingnecrosis in barley. Physiol. Mol. Plant Pathol., 39, 471–482.

WHISSON, S.C., BOEVINK, P.C., MOLELEKI, L., AVROVA, A.O.,MORALES, J.G., GILROY, E.M. et al. (2007). A translocation signal fordelivery of oomycete effector proteins into host plant cells. Nature, 450,115–118.

WOLPERT, T.J., DUNKLE, L.D. & CIUFFETTI, L.M. (2002). Host-selectivetoxins and avirulence determinants: What’s in a name? Annu. Rev.Phytopathol., 40, 251–285.

YARWOOD, C. (1957). Powdery mildews. Bot. Rev., 23, 235–301.YOSHIDA, K., SAITOH, H., FUJISAWA, S., KANZAKI, H., MATSUMURA,

H., YOSHIDA, K. et al. (2009). Association genetics reveals three novelavirulence genes from the rice blast fungal pathogen Magnaportheoryzae. Plant Cell, 21, 1573–1591.

YU, F., LYDIATE, D.J., & RIMMER, S.R. (2005). Identification of two novelgenes for blackleg resistance in Brassica napus. Theor. Appl. Genet., 110,969–979.

ZHANG, Z., HENDERSON, C., PERFECT, E., CARVER, T.L.W., THOMAS,B.J., SKAMNIOTI, P. et al. (2005). Of genes and genomes, needlesand haystacks: Blumeria graminis and functionality. Mol. Plant Pathol.,6, 561–575.

ZHENG, Y., ZHENG, W., LIN, F., ZHANG, Y., YI, Y., WANG, B. et al.(2011). Avr1-CO39 is a predominant locus governing the broad aviru-lence of Magnaporthe oryzae 2539 on cultivated rice (Oryza sativa L.).Mol. Plant–Microbe Interact., 24, 13–17.

ZHOU, E., JIA, Y., SINGH, P., CORRELL, J.C., & LEE, F.N. (2007).Instability of the Magnaporthe oryzae avirulence gene Avr-Pita altersvirulence. Fungal Genet. Biol., 44, 1024–1034.

ZIPFEL, C., ROBATZEK, S., NAVARRO, L., OAKELEY, E.J., JONES, J.D.,FELIX, G. et al. (2004). Bacterial disease resistance in Arabidopsisthrough flagellin perception. Nature, 428, 764–767.

Dow

nloa

ded

by [

Can

adia

n A

gric

ultu

re L

ibra

ry, A

gric

ultu

re a

nd A

gri-

Food

Can

ada]

at 1

3:23

08

Mar

ch 2

012