1887

Abstract

Plant diseases caused by fungal pathogens are typically initiated by molecular interactions between ‘effector’ molecules released by a pathogen and receptor molecules on or within the plant host cell. In many cases these effector-receptor interactions directly determine host resistance or susceptibility. The search for fungal effector proteins is a developing area in fungal-plant pathology, with more than 165 distinct confirmed fungal effector proteins in the public domain. For a small number of these, novel effectors can be rapidly discovered across multiple fungal species through the identification of known effector homologues. However, many have no detectable homology by standard sequence-based search methods. This study employs a novel comparison method (RemEff) that is capable of identifying protein families with greater sensitivity than traditional homology-inference methods, leveraging a growing pool of confirmed fungal effector data to enable the prediction of novel fungal effector candidates by protein family association. Resources relating to the RemEff method and data used in this study are available from https://figshare.com/projects/Effector_protein_remote_homology/87965.

Funding
This study was supported by the:
  • Grains Research and Development Corporation (Award CUR00023)
    • Principle Award Recipient: ApplicableNot
  • This is an open-access article distributed under the terms of the Creative Commons Attribution License.
Loading

Article metrics loading...

/content/journal/mgen/10.1099/mgen.0.000637
2021-09-01
2024-03-19
Loading full text...

Full text loading...

/deliver/fulltext/mgen/7/9/mgen000637.html?itemId=/content/journal/mgen/10.1099/mgen.0.000637&mimeType=html&fmt=ahah

References

  1. Thomma B, Nürnberger T, Joosten M. Of pamps and effectors: The blurred PTI-ETI dichotomy. Plant Cell 2011; 23:4–15 [View Article] [PubMed]
    [Google Scholar]
  2. De Wit PJGM, Mehrabi R, Van den Burg HA, Stergiopoulos I. Fungal effector proteins: Past, present and future. Mol Plant Pathol 2009; 10:735–747 [View Article] [PubMed]
    [Google Scholar]
  3. Thrall PH, Barrett LG, Dodds PN, Burdon JJ. Epidemiological and evolutionary outcomes in gene-for-gene and matching allele models. Front Plant Sci 2016; 6:1084 [View Article] [PubMed]
    [Google Scholar]
  4. Vleeshouwers V, Oliver RP. Effectors as tools in disease resistance breeding against biotrophic, hemibiotrophic, and necrotrophic plant pathogens. Mol Plant Microbe Interact 2014; 27:196–206 [View Article]
    [Google Scholar]
  5. Oome S, Van den Ackerveken G. Comparative and functional analysis of the widely occurring family of Nep1-like proteins. Mol Plant Microbe Interact 2014; 27:1081–1094 [View Article]
    [Google Scholar]
  6. Chen H, Kovalchuk A, Keriö S, Asiegbu FO. Distribution and bioinformatic analysis of the cerato-platanin protein family in Dikarya. Mycologia 2013; 105:1479–1488 [View Article] [PubMed]
    [Google Scholar]
  7. Olombrada M, Lázaro-Gorines R, López-Rodríguez JC, Martínez-Del-Pozo Á, Oñaderra M et al. Fungal ribotoxins: A review of potential biotechnological applications. Toxins (Basel) 2017; 9: [View Article] [PubMed]
    [Google Scholar]
  8. Bertazzoni S, Williams AH, Jones DA, Syme RA, Tan KC et al. Accessories make the outfit: accessory chromosomes and other dispensable DNA regions in plant-pathogenic fungi. Mol Plant Microbe Interact 2018; 31:779–788 [View Article] [PubMed]
    [Google Scholar]
  9. Hane JK, Rouxel T, Howlett BJ, Kema GH, Goodwin SB et al. A novel mode of chromosomal evolution peculiar to filamentous Ascomycete fungi. Genome Biol 2011; 12:R45 [View Article] [PubMed]
    [Google Scholar]
  10. Rouxel T, Grandaubert J, Hane JK, Hoede C, van de Wouw AP et al. Effector diversification within compartments of the Leptosphaeria maculans genome affected by Repeat-Induced Point mutations. Nat Commun 2011; 2:202 [View Article] [PubMed]
    [Google Scholar]
  11. Sánchez-Vallet A, Fouché S, Fudal I, Hartmann FE, Soyer JL et al. The genome biology of effector gene evolution in filamentous plant pathogens. Annu Rev Phytopathol 2018; 56:21–40 [View Article] [PubMed]
    [Google Scholar]
  12. Testa AC, Oliver RP, Hane JK. OcculterCut: A comprehensive survey of AT-rich regions in fungal genomes. Genome Biol Evol 2016; 8:2044–2064 [View Article] [PubMed]
    [Google Scholar]
  13. Torres DE, Oggenfuss U, Croll D, Seidl MF. Genome evolution in fungal plant pathogens: Looking beyond the two-speed genome model. Fungal Biology Reviews 2020; 34:136–143 [View Article]
    [Google Scholar]
  14. Adhikari BN, Hamilton JP, Zerillo MM, Tisserat N, Lévesque CA et al. Comparative genomics reveals insight into virulence strategies of plant pathogenic Oomycetes. PLoS One 2013; 8:e75072 [View Article] [PubMed]
    [Google Scholar]
  15. Fang Y, Coelho MA, Shu H, Schotanus K, Thimmappa BC et al. Long transposon-rich centromeres in an oomycete reveal divergence of centromere features in Stramenopila-Alveolata-Rhizaria lineages. PLoS Genet 2020; 16:e1008646 [View Article] [PubMed]
    [Google Scholar]
  16. Anderson RG, Deb D, Fedkenheuer K, McDowell JM. Recent Progress in RXLR Effector Research. Mol Plant Microbe Interact 2015; 28:1063–1072 [View Article] [PubMed]
    [Google Scholar]
  17. Amaro T, Thilliez GJA, Motion GB, Huitema E. A Perspective on CRN proteins in the genomicsage: evolution, classification, delivery and function revisited. Front Plant Sci 2017; 8:99 [View Article] [PubMed]
    [Google Scholar]
  18. de Guillen K, Lorrain C, Tsan P, Petre B, Saveleva N et al. Structural genomics applied to the rust fungus Melampsora larici-populina reveals two candidate effector proteins adopting cystine knot and NTF2-like protein folds. Sci Rep 2019; 9:18084 [View Article] [PubMed]
    [Google Scholar]
  19. Jones DA, Bertazzoni S, Turo CJ, Syme RA, Hane JK. Bioinformatic prediction of plant–pathogenicity effector proteins of fungi. Curr Opin Microbiol 2018; 46:43–49 [View Article] [PubMed]
    [Google Scholar]
  20. Testa AC, Hane JK, Ellwood SR, Oliver RP. CodingQuarry: Highly accurate hidden Markov model gene prediction in fungal genomes using RNA-seq transcripts. BMC Genomics 2015; 16:170 [View Article] [PubMed]
    [Google Scholar]
  21. Lu S, Gillian Turgeon B, Edwards MC. A ToxA-like protein from Cochliobolus heterostrophus induces light-dependent leaf necrosis and acts as a virulence factor with host selectivity on maize. Fungal Genet Biol 2015; 81:12–24 [View Article] [PubMed]
    [Google Scholar]
  22. Schmidt SM, Lukasiewicz J, Farrer R, Dam P van, Bertoldo C et al. Comparative genomics of Fusarium oxysporum f. sp. Melonis reveals the secreted protein recognized by the Fom-2 resistance gene in melon. New Phytol 2016; 209:307–318 [View Article] [PubMed]
    [Google Scholar]
  23. de Guillen K, Ortiz-Vallejo D, Gracy J, Fournier E, Kroj T et al. Structure analysis uncovers a highly diverse but structurally conserved effector family in phytopathogenic fungi. PLoS Pathog 2015; 11:e1005228 [View Article] [PubMed]
    [Google Scholar]
  24. Spanu PD. Cereal immunity against powdery mildews targets RNase-like proteins associated with haustoria (RALPH) effectors evolved from a common ancestral gene. New Phytol 2017; 213:969–971 [View Article]
    [Google Scholar]
  25. Kale SD, Gu B, Capelluto DGS, Dou D, Feldman E et al. External lipid PI3P mediates entry of eukaryotic pathogen effectors into plant and animal host cells. Cell 2010; 142:284–295 [View Article] [PubMed]
    [Google Scholar]
  26. Galagan JE, Selker EU. RIP: The evolutionary cost of genome defense. Trends Genet 2004; 20:417–423 [View Article] [PubMed]
    [Google Scholar]
  27. Kaas Q, Yu R, Jin A-H, Dutertre S, Craik DJ. ConoServer: Updated content, knowledge, and discovery tools in the conopeptide database. Nucleic Acids Res 2012; 40:D325–D330 [View Article] [PubMed]
    [Google Scholar]
  28. Saucedo AL, Flores-Solis D, Vega RCR, la de, Ramírez-Cordero B et al. New tricks of an old pattern structural versatility of scorpion toxins with common cysteine spacing. J Biol Chem 2012; 287:12321–12330 [View Article] [PubMed]
    [Google Scholar]
  29. Ballance GM, Lamari L, Bernier CC. Purification and characterization of a host-selective necrosis toxin from Pyrenophora tritici-repentis. Physiol Mol Plant Path 1989; 35:203–213 [View Article]
    [Google Scholar]
  30. Tuori RP, Wolpert TJ, Ciuffetti LM. Purification and immunological characterization of toxic components from cultures of Pyrenophora tritici-repentis. Mol Plant Microbe Interact 1995; 8:41–48 [View Article] [PubMed]
    [Google Scholar]
  31. Friesen TL, Stukenbrock EH, Liu Z, Meinhardt S, Ling H et al. Emergence of a new disease as a result of interspecific virulence gene transfer. Nat Genet 2006; 38:953–956 [View Article] [PubMed]
    [Google Scholar]
  32. McDonald MC, Ahren D, Simpfendorfer S, Milgate A, Solomon PS. The discovery of the virulence gene ToxA in the wheat and barley pathogen Bipolaris sorokiniana. Mol Plant Pathol 2018; 19:432–439 [View Article] [PubMed]
    [Google Scholar]
  33. McDonald MC, Taranto AP, Hill E, Schwessinger B, Liu Z et al. Transposon-mediated horizontal transfer of the host-specific virulence protein toxa between three fungal wheat pathogens. mBio 2019; 10: [View Article] [PubMed]
    [Google Scholar]
  34. Moolhuijzen P, See PT, Hane JK, Shi G, Liu Z et al. Comparative genomics of the wheat fungal pathogen Pyrenophora tritici-repentis reveals chromosomal variations and genome plasticity. BMC Genomics 2018; 19:279 [View Article] [PubMed]
    [Google Scholar]
  35. Friesen TL, Holmes DJ, Bowden RL, Faris JD. ToxA is present in the US bipolaris sorokiniana population and is a snificant virulence factor on wheat harboring Tsn1. Plant Disease 2018; 102:2446–2452 [View Article]
    [Google Scholar]
  36. Ciuffetti LM, Tuori RP, Gaventa JM. A single gene encodes a selective toxin causal to the development of tan spot of wheat. Plant Cell 1997; 9:135–144 [View Article] [PubMed]
    [Google Scholar]
  37. Sarma GN, Manning VA, Ciuffetti LM, Karplus PA. Structure of Ptr ToxA: an RGD-containing host-selective toxin from Pyrenophora tritici-repentis. Plant Cell 2005; 17:3190–3202 [View Article] [PubMed]
    [Google Scholar]
  38. Liu Z, Friesen TL, Ling H, Meinhardt SW, Oliver RP et al. The Tsn1 –ToxA interaction in the wheat–Stagonospora nodorum pathosystem parallels that of the wheat–tan spot system. Genome 2006; 49:1265–1273 [View Article] [PubMed]
    [Google Scholar]
  39. Tai YS, Bragg J, Meinhardt SW. Functional characterization of ToxA and molecular identification of its intracellular targeting protein in wheat. American J of Plant Physiology 2007; 2:76–89 [View Article]
    [Google Scholar]
  40. Lu S, Faris JD, Sherwood R, Friesen TL, Edwards MC. A dimeric PR-1-type pathogenesis-related protein interacts with ToxA and potentially mediates ToxA-induced necrosis in sensitive wheat: PR-1 potentially mediates ToxA-induced necrosis. Mol Plant Pathol 2014; 15:650–663 [View Article] [PubMed]
    [Google Scholar]
  41. Manning VA, Chu AL, Steeves JE, Wolpert TJ, Ciuffetti LM. A host-selective toxin of Pyrenophora tritici-repentis, Ptr ToxA, induces photosystem changes and reactive oxygen species accumulation in sensitive wheat. Mol Plant Microbe Interact 2009; 22:665–676 [View Article] [PubMed]
    [Google Scholar]
  42. Meinhardt SW, Cheng W, Kwon CY, Donohue CM, Rasmussen JB. Role of the Arginyl-Glycyl-Aspartic motif in the action of Ptr ToxA produced by Pyrenophora tritici-repentis. Plant Physiol 2002; 130:1545–1551 [View Article] [PubMed]
    [Google Scholar]
  43. Di X, Cao L, Hughes RK, Tintor N, Banfield MJ et al. Structure–function analysis of the Fusarium oxysporum Avr2 effector allows uncoupling of its immune-suppressing activity from recognition. New Phytol 2017; 216:897–914 [View Article] [PubMed]
    [Google Scholar]
  44. Nyarko A, Singarapu KK, Figueroa M, Manning VA, Pandelova I et al. Solution NMR structures of pyrenophora Tritici-repentis ToxB and its inactive homolog reveal potential determinants of toxin activity. J Biol Chem 2014; 289:25946–25956 [View Article] [PubMed]
    [Google Scholar]
  45. Andrie RM, Schoch CL, Hedges R, Spatafora JW, Ciuffetti LM. Homologs of ToxB, a host-selective toxin gene from Pyrenophora tritici-repentis, are present in the genome of sister-species Pyrenophora bromi and other members of the Ascomycota. Fungal Genet Biol 2008; 45:363–377 [View Article] [PubMed]
    [Google Scholar]
  46. Balesdent MH, Attard A, Kühn ML, Rouxel T. New avirulence genes in the phytopathogenic fungus Leptosphaeria maculans. Phytopathology 2002; 92:1122–1133 [View Article] [PubMed]
    [Google Scholar]
  47. Fudal I, Ross S, Gout L, Blaise F, Kuhn ML et al. Heterochromatin-like regions as ecological niches for avirulence genes in the Leptosphaeria maculans genome: map-based cloning of AvrLm6. Mol Plant Microbe Interact 2007; 20:459–470 [View Article] [PubMed]
    [Google Scholar]
  48. Grandaubert J, Lowe RG, Soyer JL, Schoch CL, Van de Wouw AP et al. Transposable element-assisted evolution and adaptation to host plant within the Leptosphaeria maculans-Leptosphaeria biglobosa species complex of fungal pathogens. BMC Genomics 2014; 15:891 [View Article] [PubMed]
    [Google Scholar]
  49. Shiller J, Van de Wouw AP, Taranto AP, Bowen JK, Dubois D et al. A large family of AvrLm6-like genes in the apple and pear scab pathogens, Venturia inaequalis and Venturia pirina. Front Plant Sci 2015; 6:980 [View Article] [PubMed]
    [Google Scholar]
  50. Lacadena J, Álvarez-García E, Carreras-Sangrà N, Herrero-Galán E, Alegre-Cebollada J et al. Fungal ribotoxins: Molecular dissection of a family of natural killers. FEMS Microbiol Rev 2007; 31:212–237 [View Article] [PubMed]
    [Google Scholar]
  51. Glück A, Wool IG. Determination of the 28 S Ribosomal RNA identity element (G4319) for Alpha-sarcin and the relationship of recognition to the selection of the catalytic site. J Mol Biol 1996; 256:838–848 [View Article] [PubMed]
    [Google Scholar]
  52. Martı́nez-Ruiz A, Kao R, Davies J, Martı́nez del Pozo Á. Ribotoxins are a more widespread group of proteins within the filamentous fungi than previously believed. Toxicon 1999; 37:1549–1563 [View Article] [PubMed]
    [Google Scholar]
  53. Citores L, Ragucci S, Ferreras JM, Di Maro A, Iglesias R. Ageritin, a ribotoxin from poplar mushroom (Agrocybe aegerita) with defensive and antiproliferative activities. ACS Chem Biol 2019; 14:1319–1327 [View Article] [PubMed]
    [Google Scholar]
  54. Landi N, Pacifico S, Ragucci S, Iglesias R, Piccolella S et al. Purification, characterization and cytotoxicity assessment of Ageritin: The first ribotoxin from the basidiomycete mushroom Agrocybe aegerita. Biochimica et Biophysica Acta (BBA) - General Subjects 2017; 5:1113–1121 [View Article]
    [Google Scholar]
  55. Herrero‐Galán E, Lacadena J, Pozo Á, del M, Boucias DG et al. The insecticidal protein hirsutellin A from the mite fungal pathogen Hirsutella thompsonii is a ribotoxin. Proteins 2008; 72:217–228 [View Article]
    [Google Scholar]
  56. Kettles GJ, Bayon C, Sparks CA, Canning G, Kanyuka K et al. Characterization of an antimicrobial and phytotoxic ribonuclease secreted by the fungal wheat pathogen Zymoseptoria tritici. New Phytol 2018; 217:320–331 [View Article] [PubMed]
    [Google Scholar]
  57. Pedersen C, Ver Loren van Themaat E, McGuffin LJ, Abbott JC, Burgis TA et al. Structure and evolution of Barley powdery mildew effector candidates. BMC Genomics 2012; 13:694 [View Article] [PubMed]
    [Google Scholar]
  58. Praz CR, Bourras S, Zeng F, Sánchez‐Martín J, Menardo F et al. AvrPm2 encodes an RNase-like avirulence effector which is conserved in the two different specialized forms of wheat and rye powdery mildew fungus. New Phytol 2017; 213:1301–1314 [View Article] [PubMed]
    [Google Scholar]
  59. Pennington HG, Jones R, Kwon S, Bonciani G, Thieron H et al. The fungal ribonuclease-like effector protein CSEP0064/BEC1054 represses plant immunity and interferes with degradation of host ribosomal RNA. PLoS Pathog 2019; 15:e1007620 [View Article] [PubMed]
    [Google Scholar]
  60. Deng CH, Plummer KM, Jones DAB, Mesarich CH, Shiller J et al. Comparative analysis of the predicted secretomes of Rosaceae scab pathogens venturia inaequalis and V. Pirina reveals expanded effector families and putative determinants of host range. BMC Genomics 2017; 18:339 [View Article] [PubMed]
    [Google Scholar]
  61. Islam SMA, Kearney CM, Baker E. Classes, databases, and prediction methods of pharmaceutically and commercially important cystine-stabilized peptides. Toxins (Basel) 2018; 10:251 [View Article] [PubMed]
    [Google Scholar]
  62. Linial M, Rappoport N, Ofer D. Overlooked short toxin-like proteins: A shortcut to drug design. Toxins (Basel) 2017; 9: [View Article] [PubMed]
    [Google Scholar]
  63. Robinson SD, Norton RS. Conotoxin gene superfamilies. Mar Drugs 2014; 12:6058–6101 [View Article] [PubMed]
    [Google Scholar]
  64. Asgari E, McHardy AC, Mofrad MRK. Probabilistic variable-length segmentation of protein sequences for discriminative motif discovery (DiMotif) and sequence embedding (ProtVecX. Sci Rep 2019; 9:3577 [View Article] [PubMed]
    [Google Scholar]
  65. Negi SS, Schein CH, Ladics GS, Mirsky H, Chang P et al. Functional classification of protein toxins as a basis for bioinformatic screening. Sci Rep 2017; 7:13940 [View Article] [PubMed]
    [Google Scholar]
  66. Park J, Teichmann SA, Hubbard T, Chothia C. Intermediate sequences increase the detection of homology between sequences11Edited by J. J Mol Biol 1997; 273:349–354 [View Article] [PubMed]
    [Google Scholar]
  67. Chen J, Liu B, Huang D. Protein remote homology detection based on an ensemble learning approach [Research article]. Biomed Res Int 2016; 2016:5813645 [View Article] [PubMed]
    [Google Scholar]
  68. Li S, Chen J, Liu B. Protein remote homology detection based on bidirectional long short-term memory. BMC Bioinformatics 2017; 18:443 [View Article] [PubMed]
    [Google Scholar]
  69. Rangwala H, Karypis G. Profile-based direct kernels for remote homology detection and fold recognition. Bioinformatics 2005; 21:4239–4247 [View Article] [PubMed]
    [Google Scholar]
  70. Altschul SF, Madden TL, Schäffer AA, Zhang J, Zhang Z et al. Gapped BLAST and PSI-BLAST: A new generation of protein database search programs. Nucleic Acids Res 1997; 25:3389–3402 [View Article] [PubMed]
    [Google Scholar]
  71. Mistry J, Finn RD, Eddy SR, Bateman A, Punta M. Challenges in homology search: Hmmer3 and convergent evolution of coiled-coil regions. Nucleic Acids Res 2013; 41:e121 [View Article] [PubMed]
    [Google Scholar]
  72. Steinegger M, Meier M, Mirdita M, Vöhringer H, Haunsberger SJ et al. HH-suite3 for fast remote homology detection and deep protein annotation. BMC Bioinformatics 2019; 20:473 [View Article] [PubMed]
    [Google Scholar]
  73. Ma J, Wang Z, Wang Z, Xu J. MRFalign: Protein homology detection through alignment of markov random fields. PLoS Comput Biol 2014; 10:e1003500 [View Article] [PubMed]
    [Google Scholar]
  74. Szklarczyk R, Wanschers BF, Cuypers TD, Esseling JJ, Riemersma M et al. Iterative orthology prediction uncovers new mitochondrial proteins and identifies C12orf62 as the human ortholog of COX14, a protein involved in the assembly of cytochrome coxidase. Genome Biol 2012; 13:R12 [View Article] [PubMed]
    [Google Scholar]
  75. Wagner I, Volkmer M, Sharan M, Villaveces JM, Oswald F et al. morFeus: A web-based program to detect remotely conserved orthologs using symmetrical best hits and orthology network scoring. BMC Bioinformatics 2014; 15:263 [View Article] [PubMed]
    [Google Scholar]
  76. Tatusov RL, Koonin E, Lipman DJ. A genomic perspective on protein families. Science 1997; 278:631–637 [View Article] [PubMed]
    [Google Scholar]
  77. Enright AJ, Van Dongen S, Ouzounis CA. An efficient algorithm for large-scale detection of protein families. Nucleic Acids Res 2002; 30:1575–1584 [View Article] [PubMed]
    [Google Scholar]
  78. Petegrosso R, Li Z, Srour MA, Saad Y, Zhang W et al. Scalable remote homology detection and fold recognition in massive protein networks. Proteins 2019; 87:478–491 [View Article]
    [Google Scholar]
  79. Chen L, Li X, Li C, Swoboda GA, Young CA et al. Two distinct Epichloë species symbiotic with Achnatherum inebrians, drunken horse grass. Mycologia 2015; 107:863–873 [View Article] [PubMed]
    [Google Scholar]
  80. Gao Q, Jin K, Ying SH, Zhang Y, Xiao G et al. Genome sequencing and comparative transcriptomics of the model entomopathogenic fungi Metarhizium anisopliae and M. acridum. PLoS Genet 2011; 7:e1001264 [View Article] [PubMed]
    [Google Scholar]
  81. Pan J. Ether Bridge Formation and Chemical Diversification in Loline Alkaloid Biosynthesis University of Kentucky; 2014
    [Google Scholar]
  82. Schardl CL, Young CA, Hesse U, Amyotte SG, Andreeva K et al. Plant-symbiotic fungi as chemical engineers: Multi-genome analysis of the Clavicipitaceae reveals dynamics of alkaloid loci. PLoS Genet 2013; 9:e1003323 [View Article] [PubMed]
    [Google Scholar]
  83. Schardl CL, Young CA, Moore N, Krom N, Dupont PY et al. Chapter ten genomes of plant-associated Clavicipitaceae. In Fungi 2014; Vol. 70:291–327 [View Article]
    [Google Scholar]
  84. Dang H, Pryor B, Peever T, Lawrence CB. The Alternaria genomes database: A comprehensive resource for a fungal genus comprised of saprophytes, plant pathogens, and allergenic species. BMC Genomics 2015; 16:239 [View Article] [PubMed]
    [Google Scholar]
  85. Chiapello H, Mallet L, Guérin C, Aguileta G, Amselem J et al. Deciphering genome content and evolutionary relationships of isolates from the fungus Magnaporthe oryzae attacking different host plants. Genome Biol Evol 2015; 7:2896–2912 [View Article] [PubMed]
    [Google Scholar]
  86. Babnigg G, Giometti CS. A database of unique protein sequence identifiers for proteome studies. Proteomics 2006; 6:4514–4522 [View Article] [PubMed]
    [Google Scholar]
  87. Cock PJA, Antao T, Chang JT, Chapman BA, Cox CJ et al. Biopython: Freely available Python tools for computational molecular biology and bioinformatics. Bioinformatics 2009; 25:1422–1423 [View Article] [PubMed]
    [Google Scholar]
  88. Steinegger M, Söding J. MMseqs2 enables sensitive protein sequence searching for the analysis of massive data sets. Nat Biotechnol 2017; 35:1026–1028 [View Article] [PubMed]
    [Google Scholar]
  89. Wright ES. DECIPHER: Harnessing local sequence context to improve protein multiple sequence alignment. BMC Bioinformatics 2015; 16:322 [View Article] [PubMed]
    [Google Scholar]
  90. Keul F, Hess M, Goesele M, Hamacher K. PFASUM: A substitution matrix from Pfam structural alignments. BMC Bioinformatics 2017; 18:293 [View Article] [PubMed]
    [Google Scholar]
  91. Urban M, Cuzick A, Seager J, Wood V, Rutherford K et al. PHI-base: The pathogen–host interactions database. Nucleic Acids Res 2020; 48:D613–D620 [View Article] [PubMed]
    [Google Scholar]
  92. Armenteros JJA, Tsirigos KD, Sønderby CK, Petersen TN, Winther O et al. SignalP 5.0 improves signal peptide predictions using deep neural networks. Nat Biotechnol 2019; 37:420–423 [View Article] [PubMed]
    [Google Scholar]
  93. Bendtsen JD, Nielsen H, von Heijne G, Brunak S. Improved prediction of signal peptides: SignalP 3.0. J Mol Biol 2004; 340:783–795 [View Article] [PubMed]
    [Google Scholar]
  94. Petersen TN, Brunak S, Heijne G von, Nielsen H. SignalP 4.0: Discriminating signal peptides from transmembrane regions. Nat Methods 2011; 8:785–786 [View Article] [PubMed]
    [Google Scholar]
  95. Savojardo C, Martelli PL, Fariselli P, Casadio R. DeepSig: Deep learning improves signal peptide detection in proteins. Bioinformatics 2018; 34:1690–1696 [View Article] [PubMed]
    [Google Scholar]
  96. Käll L, Krogh A, Sonnhammer ELL. A combined transmembrane topology and signal peptide prediction method. J Mol Biol 2004; 338:1027–1036 [View Article] [PubMed]
    [Google Scholar]
  97. Krogh A, Larsson B, von Heijne G, Sonnhammer EL. Predicting transmembrane protein topology with a hidden Markov model: Application to complete genomes. J Mol Biol 2001; 305:567–580 [View Article] [PubMed]
    [Google Scholar]
  98. Emms DM, Kelly S. OrthoFinder: Solving fundamental biases in whole genome comparisons dramatically improves orthogroup inference accuracy. Genome Biol 2015; 16:157 [View Article] [PubMed]
    [Google Scholar]
  99. Hagberg AA, Schult DA, Swart PJ. Exploring network structure, dynamics, and function using NetworkX. Varoquaux G, Vaught T, Millman J. eds In Proceedings of the 7th Python in Science Conference 2008 pp 11–15
    [Google Scholar]
  100. McKinney W. Data structures for statistical computing in python. van der Walt S, Millman J. eds In Proceedings of the 9th Python in Science Conference 2010 pp 56–61 [View Article]
    [Google Scholar]
  101. Virtanen P, Gommers R, Oliphant TE, Haberland M, Reddy T et al. SCIPY 1.0: Fundamental algorithms for scientific computing in Python. Nat Methods 2020; 17:261–272 [View Article] [PubMed]
    [Google Scholar]
  102. Hauser M, Steinegger M, Söding J. MMseqs software suite for fast and deep clustering and searching of large protein sequence sets. Bioinformatics 2016; 32:1323–1330 [View Article] [PubMed]
    [Google Scholar]
  103. Malliaros FD, Vazirgiannis M. Clustering and community detection in directed networks: A survey. Physics Reports 2013; 533:95–142 [View Article]
    [Google Scholar]
  104. Peixoto TP. The Graph-Tool Python Library 2014 [View Article]
    [Google Scholar]
  105. Schneider TD, Stephens RM. Sequence logos: A new way to display consensus sequences. Nucleic Acids Res 1990; 18:6097–6100 [View Article] [PubMed]
    [Google Scholar]
  106. Sievers F, Higgins DG. Clustal Omega for making accurate alignments of many protein sequences. Protein Sci 2018; 27:135–145 [View Article] [PubMed]
    [Google Scholar]
  107. Tareen A, Kinney JB. Logomaker: Beautiful sequence logos in Python. Bioinformatics 2020; 36:2272–2274 [View Article] [PubMed]
    [Google Scholar]
  108. Hunter JD. Matplotlib: A 2D graphics environment. Comput Sci Eng 2007; 9:90–95 [View Article]
    [Google Scholar]
  109. Mosquera G, Giraldo MC, Khang CH, Coughlan S, Valent B. Interaction transcriptome analysis identifies magnaporthe oryzae bas1-4 as biotrophy-associated secreted proteins in rice blast disease. Plant Cell 2009; 21:1273–1290 [View Article] [PubMed]
    [Google Scholar]
  110. Sharpee W, Oh Y, Yi M, Franck W, Eyre A et al. Identification and characterization of suppressors of plant cell death (SPD) effectors from Magnaporthe oryzae. Mol Plant Pathol 2017; 18:850–863 [View Article] [PubMed]
    [Google Scholar]
  111. Lievens B, Houterman PM, Rep M. Effector gene screening allows unambiguous identification of Fusarium oxysporum f. sp. Lycopersici races and discrimination from other formae speciales. FEMS Microbiol Lett 2009; 300:201–215 [View Article] [PubMed]
    [Google Scholar]
  112. Ebert MK. Effector Biology of the Sugar Beet Pathogen Cercospora Beticola Wageningen University; 2018 [View Article]
    [Google Scholar]
  113. Ghanbarnia K, Fudal I, Larkan NJ, Links MG, Balesdent M-H et al. Rapid identification of the Leptosphaeria maculans avirulence gene AvrLm2 using an intraspecific comparative genomics approach. Mol Plant Pathol 2015; 16:699–709 [View Article] [PubMed]
    [Google Scholar]
  114. Rep M, Does HCVD, Meijer M, Wijk R, Houterman PM et al. A small, cysteine-rich protein secreted by Fusarium oxysporum during colonization of xylem vessels is required for I-3-mediated resistance in tomato. Molecular Microbiology 2004; 53:1373–1383 [View Article]
    [Google Scholar]
  115. Ben M’Barek S, Cordewener JHG, Tabib Ghaffary SM, van der Lee TAJ, Liu Z et al. FPLC and liquid-chromatography mass spectrometry identify candidate necrosis-inducing proteins from culture filtrates of the fungal wheat pathogen Zymoseptoria tritici. Fungal Genet Biol 2015; 79:54–62 [View Article] [PubMed]
    [Google Scholar]
  116. Laugé R, Joosten M, Van den Ackerveken G, Van den Broek HWJ, De Wit P. The in planta-produced extracellular proteins ECP1 and ECP2 of Cladosporium fulvum are virulence factors. MPMI 1997; 10:725–734 [View Article]
    [Google Scholar]
  117. Schmidt SM, Kuhn H, Micali C, Liller C, Kwaaitaal M et al. Interaction of a Blumeria graminis f. sp. hordei effector candidate with a barley ARF-GAP suggests that host vesicle trafficking is a fungal pathogenicity target: Blumeria graminis effector candidates. Mol Plant Pathol 2014; 15:535–549 [View Article] [PubMed]
    [Google Scholar]
  118. Bolton MD, Esse HP, Vossen JH, Jonge RD, Stergiopoulos I et al. The novel Cladosporium fulvum lysin motif effector Ecp6 is a virulence factor with orthologues in other fungal species. Molecular Microbiology 2008; 69:119–136 [View Article]
    [Google Scholar]
  119. Marshall R, Kombrink A, Motteram J, Loza-Reyes E, Lucas J et al. Analysis of two in planta expressed LysM effector homologs from the fungus Mycosphaerella graminicola reveals novel functional properties and varying contributions to virulence on wheat. Plant Physiol 2011; 156:756–769 [View Article] [PubMed]
    [Google Scholar]
  120. Irieda H, Inoue Y, Mori M, Yamada K, Oshikawa Y et al. Conserved fungal effector suppresses PAMP-triggered immunity by targeting plant immune kinases. Proc Natl Acad Sci U S A 2019; 116:496–505 [View Article] [PubMed]
    [Google Scholar]
  121. Yoshino K, Irieda H, Sugimoto F, Yoshioka H, Okuno T et al. Cell death of Nicotiana benthamiana is induced by secreted protein NIS1 of Colletotrichum orbiculare and is suppressed by a homologue of CgDN3. Mol Plant Microbe Interact 2012; 25:625–636 [View Article] [PubMed]
    [Google Scholar]
  122. Chuma I, Isobe C, Hotta Y, Ibaragi K, Futamata N et al. Multiple translocation of the AVR-Pita effector gene among chromosomes of the riceblast fungus Magnaporthe oryzae and related species. PLoS Pathog 2011; 7:e1002147 [View Article] [PubMed]
    [Google Scholar]
  123. Dai Y, Jia Y, Correll J, Wang X, Wang Y. Diversification and evolution of the avirulence gene AVR-Pita1 in field isolates of Magnaporthe oryzae. Fungal Genet Biol 2010; 47:973–980 [View Article] [PubMed]
    [Google Scholar]
  124. Ramachandran SR, Yin C, Kud J, Tanaka K, Mahoney AK et al. Effectors from wheat rust fungi suppress multiple plant defense responses. Phytopathology 2017; 107:75–83 [View Article] [PubMed]
    [Google Scholar]
  125. Mueller AN, Ziemann S, Treitschke S, Aßmann D, Doehlemann G. Compatibility in the Ustilago maydis-maize interaction requires inhibition of host cysteine proteases by the fungal effector Pit2. PLoS Pathog 2013; 9:e1003177 [View Article] [PubMed]
    [Google Scholar]
  126. Schweizer G, Münch K, Mannhaupt G, Schirawski J, Kahmann R et al. Positively selected effector genes and their contribution to virulence in the smut fungus Sporisorium reilianum. Genome Biol Evol 2018; 10:629–645 [View Article] [PubMed]
    [Google Scholar]
  127. Bailey BA, Apel-Birkhold PC, Luster DG. Expression of NEP1 by Fusarium oxysporum f. Sp erythroxyli after gene replacement and overexpression using polyethylene glycol-mediated transformation. Phytopathology 2002; 92:833–841 [View Article]
    [Google Scholar]
  128. Garcia O, Macedo JAN, Tibúrcio R, Zaparoli G, Rincones J et al. Characterization of necrosis and ethylene-inducing proteins (NEP) in the basidiomycete Moniliophthora perniciosa, the causal agent of witches’ broom in Theobroma cacao. Mycol Res 2007; 111:443–455 [View Article] [PubMed]
    [Google Scholar]
  129. Staats M, van Baarlen P, Schouten A, van Kan JAL, Bakker FT. Positive selection in phytotoxic protein-encoding genes of Botrytis species. Fungal Genet Biol 2007; 44:52–63 [View Article] [PubMed]
    [Google Scholar]
  130. Wang JY, Cai Y, Gou JY, Mao YB, Xu YH et al. VdNEP, an Elicitor from Verticillium dahliae, Induces cotton plant wilting. Appl Environ Microbiol 2004; 70:4989–4995 [View Article] [PubMed]
    [Google Scholar]
  131. Balesdent M-H, Fudal I, Ollivier B, Bally P, Grandaubert J et al. The dispensable chromosome of Leptosphaeria maculans shelters an effector gene conferring avirulence towards Brassica rapa. New Phytol 2013; 198:887–898 [View Article]
    [Google Scholar]
  132. Wouw APV de, Lowe RGT, Elliott CE, Dubois DJ, Howlett BJ. An avirulence gene, AvrLmJ1, from the blackleg fungus, Leptosphaeria maculans, confers avirulence to Brassica juncea cultivars. Mol Plant Pathol 2014; 15:523–530 [View Article] [PubMed]
    [Google Scholar]
  133. Mesarich CH, Griffiths SA, van der Burgt A, Okmen B, Beenen HG et al. Transcriptome sequencing uncovers the Avr5 avirulence gene of the tomato leaf mold pathogen Cladosporium fulvum. Mol Plant Microbe Interact 2014; 27:846–857 [View Article] [PubMed]
    [Google Scholar]
  134. Ridout CJ, Skamnioti P, Porritt O, Sacristan S, Jones JDG et al. Multiple avirulence paralogues in cereal powdery mildew fungi may contribute to parasite fitness and defeat of plant resistance. Plant Cell 2006; 18:2402–2414 [View Article] [PubMed]
    [Google Scholar]
  135. Akum FN, Steinbrenner J, Biedenkopf D, Imani J, Kogel K-H. The Piriformospora indica effector PIIN_08944 promotes the mutualistic Sebacinalean symbiosis. Front Plant Sci 2015; 6:906 [View Article] [PubMed]
    [Google Scholar]
  136. Wawra S, Fesel P, Widmer H, Timm M, Seibel J et al. The fungal-specific β-glucan-binding lectin FGB1 alters cell-wall composition and suppresses glucan-triggered immunity in plants. Nat Commun 2016; 7:13188 [View Article] [PubMed]
    [Google Scholar]
  137. Chen S, Songkumarn P, Venu RC, Gowda M, Bellizzi M et al. Identification and characterization of In planta–expressed secreted effector proteins from Magnaporthe oryzae that induce cell death in rice. MPMI 2012; 26:191–202 [View Article]
    [Google Scholar]
  138. Mentlak TA, Kombrink A, Shinya T, Ryder LS, Otomo I et al. Effector-mediated suppression of chitin-triggered immunity by Magnaporthe oryzae is necessary for rice blast disease. Plant Cell 2012; 24:322–335 [View Article] [PubMed]
    [Google Scholar]
  139. Liu T, Song T, Zhang X, Yuan H, Su L et al. Unconventionally secreted effectors of two filamentous pathogens target plant salicylate biosynthesis. Nat Commun 2014; 5:4686 [View Article] [PubMed]
    [Google Scholar]
  140. Poppe S, Dorsheimer L, Happel P, Stukenbrock EH. Rapidly evolving genes are key players in host specialization and virulence of the fungal Wheat pathogen Zymoseptoria tritici (Mycosphaerella graminicola. PLoS Pathog 2015; 11:e1005055 [View Article] [PubMed]
    [Google Scholar]
  141. Kunjeti SG, Iyer G, Johnson E, Li E, Broglie KE et al. Identification of Phakopsora pachyrhizi candidate effectors with virulence activity in a distantly related pathosystem. Front Plant Sci 2016; 7:269 [View Article] [PubMed]
    [Google Scholar]
  142. Liu Z, Zhang Z, Faris JD, Oliver RP, Syme R et al. The Cysteine rich Necrotrophic effector SnTox1 produced by Stagonospora nodorum triggers susceptibility of wheat lines harboring Snn1. PLoS Pathog 2012; 8:e1002467 [View Article] [PubMed]
    [Google Scholar]
  143. Houterman PM, Cornelissen BJC, Rep M. Suppression of plant resistance gene-based immunity by a fungal effector. PLoS Pathog 2008; 4:e1000061 [View Article] [PubMed]
    [Google Scholar]
  144. Stephenson SA, Hatfield J, Rusu AG, Maclean DJ, Manners JM. CgDN3: an essential pathogenicity gene of Colletotrichum gloeosporioides necessary to avert a hypersensitive-like response in the host Stylosanthes guianensis. Mol Plant Microbe Interact 2000; 13:929–941 [View Article]
    [Google Scholar]
  145. Vargas WA, Sanz-Martín JM, Rech GE, Armijos-Jaramillo VD, Rivera LP et al. A fungal effector with host nuclear localization and DNA-binding properties is required for maize anthracnose development. MPMI 2015; 29:83–95 [View Article]
    [Google Scholar]
  146. Catanzariti A-M, Dodds PN, Lawrence GJ, Ayliffe MA, Ellis JG. Haustorially expressed secreted proteins from flax rust are highly enriched for avirulence elicitors. Plant Cell 2006; 18:243–256 [PubMed]
    [Google Scholar]
  147. Dodds PN, Lawrence GJ, Catanzariti AM, Ayliffe MA, Ellis JG. The Melampsora lini Avrl567 avirulence genes are expressed in Haustoria and their products are recognized inside plant cells. Plant Cell 2004; 16:755–768 [View Article] [PubMed]
    [Google Scholar]
  148. Lyu X, Shen C, Fu Y, Xie J, Jiang D et al. A small secreted virulence-related protein is essential for the necrotrophic interactions of Sclerotinia sclerotiorum with its host plants. PLoS Pathog 2016; 12:e1005435 [View Article] [PubMed]
    [Google Scholar]
  149. Kirsten S, Navarro-Quezada A, Penselin D, Wenzel C, Matern A et al. Necrosis-inducing proteins of Rhynchosporium commune, effectors in quantitative disease resistance. Mol Plant Microbe Interact 2012; 25:1314–1325 [View Article] [PubMed]
    [Google Scholar]
  150. Zhang L, Ni H, Du X, Wang S, Ma X-W et al. The Verticillium-specific protein VdSCP7 localizes to the plant nucleus and modulates immunity to fungal infections. New Phytol 2017; 215:368–381 [View Article]
    [Google Scholar]
  151. Joosten MHAJ, Cozijnsen TJ, De Wit PJGM. Host resistance to a fungal tomato pathogen lost by a single base-pair change in an avirulence gene. Nature 1994; 367:384–386 [View Article] [PubMed]
    [Google Scholar]
  152. Plissonneau C, Daverdin G, Ollivier B, Blaise F, Degrave A et al. A game of hide and seek between avirulence genes AvrLm4-7 and AvrLm3 in Leptosphaeria maculans. New Phytol 2016; 209:1613–1624 [View Article]
    [Google Scholar]
  153. Rice P, Longden I, Bleasby A. EMBOSS: the european molecular Biology open software suite. Trends Genet 2000; 16:276–277 [View Article] [PubMed]
    [Google Scholar]
  154. Katoh K, Standley DM. MAFFT multiple sequence alignment software version 7: Improvements in performance and usability. Mol Biol Evol 2013; 30:772–780 [View Article] [PubMed]
    [Google Scholar]
  155. Stergiopoulos I, Kourmpetis YA, Slot JC, Bakker FT, De Wit P et al. In silico characterization and molecular evolutionary analysis of a novel superfamily of fungal effector proteins. Mol Biol Evol 2012; 29:3371–3384 [View Article]
    [Google Scholar]
  156. Cao L, Blekemolen MC, Tintor N, Cornelissen BJC, Takken FLW. The Fusarium oxysporum Avr2-Six5 effector pair alters plasmodesmatal exclusion selectivity to facilitate Cell-to-Cell movement of Avr2. Mol Plant 2018; 11:691–705 [View Article] [PubMed]
    [Google Scholar]
  157. Petit‐Houdenot Y, Degrave A, Meyer M, Blaise F, Ollivier B et al. A two genes – for – one gene interaction between Leptosphaeria maculans and Brassica napus. New Phytol 2019; 223:397–411 [View Article]
    [Google Scholar]
  158. Wang C, Liu Y, Liu L, Wang Y, Yan J et al. The biotrophy-associated secreted protein 4 (BAS4) participates in the transition of Magnaporthe oryzae from the biotrophic to the necrotrophic phase. Saudi J Biol Sci 2019; 26:795–807 [View Article] [PubMed]
    [Google Scholar]
  159. Khang CH, Berruyer R, Giraldo MC, Kankanala P, Park S-Y et al. Translocation of Magnaporthe oryzae effectors into rice cells and their subsequent cell-to-cell movement. Plant Cell 2010; 22:1388–1403 [View Article] [PubMed]
    [Google Scholar]
  160. Manning VA, Hamilton SM, Karplus PA, Ciuffetti LM. The Arg-Gly-Asp–containing, solvent-exposed loop of Ptr ToxA is required for internalization. Mol Plant Microbe Interact 2008; 21:315–325 [View Article] [PubMed]
    [Google Scholar]
  161. Figueroa M, Manning VA, Pandelova I, Ciuffetti LM. Persistence of the host-selective toxin PTR ToxB in the apoplast. Mol Plant Microbe Interact 2015; 28:1082–1090 [View Article] [PubMed]
    [Google Scholar]
  162. Giraldo MC, Dagdas YF, Gupta YK, Mentlak TA, Yi M et al. Two distinct secretion systems facilitate tissue invasion by the rice blast fungus Magnaporthe oryzae. Nat Commun 2013; 4:1996 [View Article] [PubMed]
    [Google Scholar]
  163. Owji H, Nezafat N, Negahdaripour M, Hajiebrahimi A, Ghasemi Y. A comprehensive review of signal peptides: Structure, roles, and applications. Eur J Cell Biol 2018; 97:422–441 [View Article] [PubMed]
    [Google Scholar]
  164. Duffy J, Patham B, Mensa-Wilmot K. Discovery of functional motifs in h-regions of trypanosome signal sequences. Biochemical Journal 2010; 426:135–145 [View Article]
    [Google Scholar]
  165. Pérez-Cañadillas JM, Santoro J, Campos-Olivas R, Lacadena J, Martínez del Pozo A et al. The highly refined solution structure of the cytotoxic ribonuclease alpha-sarcin reveals the structural requirements for substrate recognition and ribonucleolytic activity. J Mol Biol 2000; 299:1061–1073 [View Article] [PubMed]
    [Google Scholar]
  166. Viegas A, Herrero-Galán E, Oñaderra M, Macedo AL, Bruix M. Solution structure of hirsutellin A-new insights into the active site and interacting interfaces of ribotoxins. FEBS J 2009; 276:2381–2390 [View Article] [PubMed]
    [Google Scholar]
  167. Yang X, Moffat K. Insights into specificity of cleavage and mechanism of cell entry from the crystal structure of the highly specific Aspergillus ribotoxin, restrictocin. Structure 1996; 4:837–852 [View Article] [PubMed]
    [Google Scholar]
  168. Bourras S, McNally KE, Ben-David R, Parlange F, Roffler S et al. Multiple Avirulence loci and allele-specific effector recognition control the Pm3 race-specific resistance of wheat to powdery mildew. Plant Cell 2015; 27:2991–3012 [View Article] [PubMed]
    [Google Scholar]
  169. McNally KE, Menardo F, Lüthi L, Praz CR, Müller MC et al. Distinct domains of the AVRPM3A2/F2 avirulence protein from wheat powdery mildew are involved in immune receptor recognition and putative effector function. New Phytol 2018; 218:681–695 [View Article] [PubMed]
    [Google Scholar]
  170. Paulus JK, van der Hoorn RAL. Tricked or trapped-Two decoy mechanisms in host-pathogen interactions. PLoS Pathog 2018; 14:e1006761 [View Article] [PubMed]
    [Google Scholar]
  171. Feldman D, Yarden O, Hadar Y. Seeking the roles for fungal small-secreted proteins in affecting saprophytic lifestyles. Front Microbiol 2020; 11:455 [View Article] [PubMed]
    [Google Scholar]
  172. Hane JK, Paxman J, Jones DAB, Oliver RP, de Wit P. “CATAStrophy”, a genome-informed trophic classification of Filamentous plant pathogens – how many different types of Filamentous plant pathogens are there?. Front Microbiol 2020; 10:3088 [View Article] [PubMed]
    [Google Scholar]
  173. Kuo H-C, Hui S, Choi J, Asiegbu FO, Valkonen JPT et al. Secret lifestyles of Neurospora crassa. Sci Rep 2015; 4:5135 [View Article] [PubMed]
    [Google Scholar]
  174. Fry BG, Roelants K, Champagne DE, Scheib H, Tyndall JDA et al. The toxicogenomic multiverse: Convergent recruitment of proteins into animal venoms. Annu Rev Genomics Hum Genet 2009; 10:483–511 [View Article] [PubMed]
    [Google Scholar]
http://instance.metastore.ingenta.com/content/journal/mgen/10.1099/mgen.0.000637
Loading
/content/journal/mgen/10.1099/mgen.0.000637
Loading

Data & Media loading...

Supplements

Loading data from figshare Loading data from figshare
This is a required field
Please enter a valid email address
Approval was a Success
Invalid data
An Error Occurred
Approval was partially successful, following selected items could not be processed due to error