Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

Appressorium-mediated plant infection by Magnaporthe oryzae is regulated by a Pmk1-dependent hierarchical transcriptional network

Abstract

Rice blast is a devastating disease caused by the fungal pathogen Magnaporthe oryzae that threatens rice production around the world. The fungus produces a specialized infection cell, called the appressorium, that enables penetration through the plant cell wall in response to surface signals from the rice leaf. The underlying biology of plant infection, including the regulation of appressorium formation, is not completely understood. Here we report the identification of a network of temporally coregulated transcription factors that act downstream of the Pmk1 mitogen-activated protein kinase pathway to regulate gene expression during appressorium-mediated plant infection. We show that this tiered regulatory mechanism involves Pmk1-dependent phosphorylation of the Hox7 homeobox transcription factor, which regulates genes associated with induction of major physiological changes required for appressorium development—including cell-cycle control, autophagic cell death, turgor generation and melanin biosynthesis—as well as controlling a additional set of virulence-associated transcription factor–encoding genes. Pmk1-dependent phosphorylation of Mst12 then regulates gene functions involved in septin-dependent cytoskeletal re-organization, polarized exocytosis and effector gene expression, which are necessary for plant tissue invasion. Identification of this regulatory cascade provides new potential targets for disease intervention.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Global comparative transcriptional profile analysis to define the response of M. oryzae to surface hydrophobicity and the presence/absence of the Pmk1 MAPK.
Fig. 2: Functional analysis and comparative global transcriptional profile analysis in response to the presence/absence of the M. oryzae TF Mst12.
Fig. 3: Defining the hierarchy of transcriptional control during appressorium development by M. oryzae.
Fig. 4: Characterization of the Hox7 homeobox TF and its role in the regulation of gene expression during appressorium development by M. oryzae.
Fig. 5: Phosphoproteomic analysis reveals Pmk1-dependent phosphorylation of Hox7 in M. oryzae.
Fig. 6: Purified recombinant GST–Pmk1 phosphorylates MBP–Hox7 and SUMO–Mst12 in vitro.
Fig. 7: Pmk1-dependent phosphorylation of Hox7 is required for appressorium development.

Similar content being viewed by others

Data availability

The RNA-seq data described in this study have been submitted to the European Nucleotide Archive (ENA): appressorial RNA-seq data under the accession number PRJEB36580 and mycelial RNA-seq data under the accession number PRJEB44745. The ChIP–seq data described in this study have been submitted to Gene Expression Omnibus under the accession number GSE182534. The proteomic data described in this study have been deposited into the ProteomeXchange Consortium via PRIDE (Perez-Riverol et al., 2019) partner repository with the dataset identifier PXD025700. The PRM data have been made publicly available through PanoramaWeb (https://panoramaweb.org/84ne1U.url) and the corresponding ProteomeXchange ID for the data is PXD028052. The M. oryzae genome database used in this study was http://fungi.ensembl.org/Magnaporthe_oryzae/Info/Index. All M. oryzae strains generated in this study are freely available on request from the corresponding authors. Source data are provided with this paper.

Code availability

Scripts for the analysis and prediction of the peaks of ChIP–seq experiments have been publicly deposited in GitHub at https://github.com/threadmapper/sequence-under-peaks.

References

  1. Talbot, N. J. On the trail of a cereal killer: exploring the biology of Magnaporthe grisea. Annu. Rev. Microbiol. 57, 177–202 (2003).

    CAS  PubMed  Google Scholar 

  2. DeZwaan, T. M., Carroll, A. M., Valent, B. & Sweigard, J. A. Magnaporthe grisea Pth11p is a novel plasma membrane protein that mediates appressorium differentiation in response to inductive substrate cues. Plant Cell 11, 2013–2030 (1999).

    CAS  PubMed  PubMed Central  Google Scholar 

  3. Hamer, J. E., Howard, R. J., Chumley, F. G. & Valent, B. A mechanism for surface attachment in spores of a plant pathogenic fungus. Science 239, 288–190 (1988).

    CAS  PubMed  Google Scholar 

  4. de Jong, J. C., McCormack, B. J., Smirnoff, N. & Talbot, N. J. Glycerol generates turgor in rice blast. Nature 389, 244–244 (1997).

    Google Scholar 

  5. Howard, R. J., Ferrari, M. A., Roach, D. H. & Money, N. P. Penetration of hard substrates by a fungus employing enormous turgor pressures. Proc. Natl Acad. Sci. USA 88, 11281–11284 (1991).

    CAS  PubMed  PubMed Central  Google Scholar 

  6. Kershaw, M. J. & Talbot, N. J. Genome-wide functional analysis reveals that infection-associated fungal autophagy is necessary for rice blast disease. Proc. Natl Acad. Sci. USA 106, 15967–15972 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Saunders, D. G., Aves, S. J. & Talbot, N. J. Cell cycle-mediated regulation of plant infection by the rice blast fungus. Plant Cell 22, 497–507 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  8. Veneault-Fourrey, C., Barooah, M., Egan, M., Wakley, G. & Talbot, N. J. Autophagic fungal cell death is necessary for infection by the rice blast fungus. Science 312, 580–583 (2006).

    CAS  PubMed  Google Scholar 

  9. Marroquin-Guzman, M., Sun, G. & Wilson, R. A. Glucose-ABL1-TOR signaling modulates cell cycle tuning to control terminal appressorial cell differentiation. PLoS Genet. 13, e1006557 (2017).

    PubMed  PubMed Central  Google Scholar 

  10. Marroquin-Guzman, M. & Wilson, R. A. GATA-dependent glutaminolysis drives appressorium formation in Magnaporthe oryzae by suppressing TOR inhibition of cAMP/PKA signaling. PLoS Pathog. 11, e1004851 (2015).

    PubMed  PubMed Central  Google Scholar 

  11. Sun, G., Qi, X. & Wilson, R. A. A feed-forward subnetwork emerging from integrated TOR- and cAMP/PKA-signaling architecture reinforces Magnaporthe oryzae appressorium morphogenesis. Mol. Plant Microbe Interact. 32, 593–607 (2019).

    CAS  PubMed  Google Scholar 

  12. Ryder, L. S. et al. NADPH oxidases regulate septin-mediated cytoskeletal remodeling during plant infection by the rice blast fungus. Proc. Natl Acad. Sci. USA 110, 3179–3184 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  13. Dagdas, Y. F. et al. Septin-mediated plant cell invasion by the rice blast fungus, Magnaporthe oryzae. Science 336, 1590–1595 (2012).

    CAS  PubMed  Google Scholar 

  14. Rocha, R. O., Elowsky, C., Pham, N. T. T. & Wilson, R. A. Spermine-mediated tight sealing of the Magnaporthe oryzae appressorial pore-rice leaf surface interface. Nat. Microbiol. 5, 1472–1480 (2020).

    CAS  PubMed  Google Scholar 

  15. Sakulkoo, W. et al. A single fungal MAP kinase controls plant cell-to-cell invasion by the rice blast fungus. Science 359, 1399–1403 (2018).

    CAS  PubMed  Google Scholar 

  16. Giraldo, M. C. et al. Two distinct secretion systems facilitate tissue invasion by the rice blast fungus Magnaporthe oryzae. Nat. Commun. 4, 1996 (2013).

    PubMed  Google Scholar 

  17. Xu, J. R. & Hamer, J. E. MAP kinase and cAMP signaling regulate infection structure formation and pathogenic growth in the rice blast fungus Magnaporthe grisea. Genes Dev. 10, 2696–2706 (1996).

    CAS  PubMed  Google Scholar 

  18. Saunders, D. G., Dagdas, Y. F. & Talbot, N. J. Spatial uncoupling of mitosis and cytokinesis during appressorium-mediated plant infection by the rice blast fungus Magnaporthe oryzae. Plant Cell 22, 2417–2428 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  19. Dean, R. A. et al. The genome sequence of the rice blast fungus Magnaporthe grisea. Nature 434, 980–986 (2005).

    CAS  PubMed  Google Scholar 

  20. Thines, E., Weber, R. W. & Talbot, N. J. MAP kinase and protein kinase A-dependent mobilization of triacylglycerol and glycogen during appressorium turgor generation by Magnaporthe grisea. Plant Cell 12, 1703–1718 (2000).

    CAS  PubMed  PubMed Central  Google Scholar 

  21. Becker, J., Liermann, J. C., Opatz, T., Anke, H. & Thines, E. GKK1032A2, a secondary metabolite from Penicillium sp. IBWF-029-96, inhibits conidial germination in the rice blast fungus Magnaporthe oryzae. J. Antibiot. 65, 99–102 (2012).

    CAS  Google Scholar 

  22. Park, G., Xue, C., Zheng, L., Lam, S. & Xu, J. R. MST12 regulates infectious growth but not appressorium formation in the rice blast fungus Magnaporthe grisea. Mol. Plant Microbe Interact. 15, 183–192 (2002).

    CAS  PubMed  Google Scholar 

  23. Soanes, D. M., Chakrabarti, A., Paszkiewicz, K. H., Dawe, A. L. & Talbot, N. J. Genome-wide transcriptional profiling of appressorium development by the rice blast fungus Magnaporthe oryzae. PLoS Pathog. 8, e1002514 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  24. Oses-Ruiz, M. & Talbot, N. J. Cell cycle-dependent regulation of plant infection by the rice blast fungus Magnaporthe oryzae. Commun. Integr. Biol. 10, e1372067 (2017).

    PubMed  PubMed Central  Google Scholar 

  25. Oses-Ruiz, M., Sakulkoo, W., Littlejohn, G. R., Martin-Urdiroz, M. & Talbot, N. J. Two independent S-phase checkpoints regulate appressorium-mediated plant infection by the rice blast fungus Magnaporthe oryzae. Proc. Natl Acad. Sci. USA 114, E237–E244 (2017).

    CAS  PubMed  Google Scholar 

  26. Kulkarni, R. D., Thon, M. R., Pan, H. & Dean, R. A. Novel G-protein-coupled receptor-like proteins in the plant pathogenic fungus Magnaporthe grisea. Genome Biol. 6, R24 (2005).

    PubMed  PubMed Central  Google Scholar 

  27. Chumley, F. G. & Valent, B. Genetic analysis of melanin deficient, nonpathogenic mutants of Magnaporthe grisea. Mol. Plant Microbe Interact. 3, 135–143 (1990).

    CAS  Google Scholar 

  28. Skamnioti, P. & Gurr, S. J. Magnaporthe grisea Cutinase2 mediates appressorium differentiation and host penetration and is required for full virulence. Plant Cell 19, 2674–2689 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  29. Johnson, K. L., Jones, B. J., Bacic, A. & Schultz, C. J. The fasciclin-like arabinogalactan proteins of Arabidopsis. A multigene family of putative cell adhesion molecules. Plant Physiol. 133, 1911–1925 (2003).

    CAS  PubMed  PubMed Central  Google Scholar 

  30. Seifert, G. J. Fascinating fasciclins: A surprisingly widespread family of proteins that mediate interactions between the cell exterior and the cell surface. Int. J. Mol. Sci. https://doi.org/10.3390/ijms19061628 (2018).

  31. Dong, B. et al. MgAtg9 trafficking in Magnaporthe oryzae. Autophagy 5, 946–953 (2009).

    CAS  PubMed  Google Scholar 

  32. Petersen, T. N., Brunak, S., von Heijne, G. & Nielsen, H. SignalP 4.0: discriminating signal peptides from transmembrane regions. Nat. Methods 8, 785–786 (2011).

    CAS  PubMed  Google Scholar 

  33. Haijiao, L. et al. A novel Magnaporthe oryzae gene MCG1, encoding an extracellular globular protein, affects conidial germination and appressorial formation. Int. J. Agric. Technol. 7, 1647–1660 (2011).

    Google Scholar 

  34. Skamnioti, P., Henderson, C., Zhang, Z., Robinson, Z. & Gurr, S. J. A novel role for catalase B in the maintenance of fungal cell-wall integrity during host invasion in the rice blast fungus Magnaporthe grisea. Mol. Plant Microbe Interact. 20, 568–580 (2007).

    CAS  PubMed  Google Scholar 

  35. Mosquera, G., Giraldo, M. C., Khang, C. H., Coughlan, S. & Valent, B. Interaction transcriptome analysis identifies Magnaporthe oryzae BAS1-4 as biotrophy-associated secreted proteins in rice blast disease. Plant Cell 21, 1273–1290 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  36. Lu, J., Cao, H., Zhang, L., Huang, P. & Lin, F. Systematic analysis of Zn2Cys6 transcription factors required for development and pathogenicity by high-throughput gene knockout in the rice blast fungus. PLoS Pathog. 10, e1004432 (2014).

    PubMed  PubMed Central  Google Scholar 

  37. Sweigard, J. A., Carroll, A. M., Farrall, L., Chumley, F. G. & Valent, B. Magnaporthe grisea pathogenicity genes obtained through insertional mutagenesis. Mol. Plant Microbe Interact. 11, 404–412 (1998).

    CAS  PubMed  Google Scholar 

  38. Felenbok, B. The ethanol utilization regulon of Aspergillus nidulans: the alcA–alcR system as a tool for the expression of recombinant proteins. J. Biotechnol. 17, 11–17 (1991).

    CAS  PubMed  Google Scholar 

  39. Kim, S. et al. Homeobox transcription factors are required for conidiation and appressorium development in the rice blast fungus Magnaporthe oryzae. PLoS Genet. 5, e1000757 (2009).

    PubMed  PubMed Central  Google Scholar 

  40. Howard, R. J. & Valent, B. Breaking and entering: host penetration by the fungal rice blast pathogen Magnaporthe grisea. Annu. Rev. Microbiol. 50, 491–512 (1996).

    CAS  PubMed  Google Scholar 

  41. Kwon, S. et al. Role of the histone acetyltransferase Rtt109 in development and pathogenicity of the rice blast fungus. Mol. Plant Microbe Interact. 31, 1200–1210 (2018).

    CAS  PubMed  Google Scholar 

  42. Lane, S., Zhou, S., Pan, T., Dai, Q. & Liu, H. The basic helix-loop-helix transcription factor Cph2 regulates hyphal development in Candida albicans partly via Tec1. Mol. Cell. Biol. 21, 6418–6428 (2001).

    CAS  PubMed  PubMed Central  Google Scholar 

  43. McLeod, M. et al. Cpc2, a fission yeast homologue of mammalian RACK1 protein, interacts with Ran1 (Pat1) kinase to regulate cell cycle progression and meiotic development. Mol. Cell. Biol. 20, 4016–4027 (2000).

    CAS  PubMed  PubMed Central  Google Scholar 

  44. Mitchell, T. K. & Dean, R. A. The cAMP-dependent protein kinase catalytic subunit is required for appressorium formation and pathogenesis by the rice blast pathogen Magnaporthe grisea. Plant Cell 7, 1869–1878 (1995).

    CAS  PubMed  PubMed Central  Google Scholar 

  45. Xu, J.-R., Urban, M., Sweigard, J. A. & Hamer, J. E. The CPKA gene of Magnaporthe grisea is essential for appressorial penetration. Mol. Plant Microbe Interact. 10, 187–194 (1997).

    CAS  Google Scholar 

  46. Menke, F. L. et al. Tobacco transcription factor WRKY1 is phosphorylated by the MAP kinase SIPK and mediates HR-like cell death in tobacco. Mol. Plant Microbe Interact. 18, 1027–1034 (2005).

    CAS  PubMed  Google Scholar 

  47. Pearson, J. C., Lemons, D. & McGinnis, W. Modulating Hox gene functions during animal body patterning. Nat. Rev. Genet. 6, 893–904 (2005).

    CAS  PubMed  Google Scholar 

  48. McGinnis, W., Garber, R. L., Wirz, J., Kuroiwa, A. & Gehring, W. J. A homologous protein-coding sequence in Drosophila homeotic genes and its conservation in other metazoans. Cell 37, 403–408 (1984).

    CAS  PubMed  Google Scholar 

  49. Bürglin, T. R. & Affolter, M. Homeodomain proteins: an update. Chromosoma 125, 497–521 (2016).

    PubMed  Google Scholar 

  50. Xuan, F. et al. Homeobox C9 suppresses Beclin1-mediated autophagy in glioblastoma by directly inhibiting the transcription of death-associated protein kinase 1. Neuro Oncol. 18, 819–829 (2016).

    CAS  PubMed  Google Scholar 

  51. Mao, L. et al. HOXC9 links cell-cycle exit and neuronal differentiation and is a prognostic marker in neuroblastoma. Cancer Res. 71, 4314–4324 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  52. Wilson, R. A., Gibson, R. P., Quispe, C. F., Littlechild, J. A. & Talbot, N. J. An NADPH-dependent genetic switch regulates plant infection by the rice blast fungus. Proc. Natl Acad. Sci. USA 107, 21902–21907 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  53. Celenza, J. L. & Carlson, M. Mutational analysis of the Saccharomyces cerevisiae SNF1 protein kinase and evidence for functional interaction with the SNF4 protein. Mol. Cell. Biol. 9, 5034–5044 (1989).

    CAS  PubMed  PubMed Central  Google Scholar 

  54. Wang, Z., Wilson, W. A., Fujino, M. A. & Roach, P. J. Antagonistic controls of autophagy and glycogen accumulation by Snf1p, the yeast homolog of AMP-activated protein kinase, and the cyclin-dependent kinase Pho85p. Mol. Cell. Biol. 21, 5742–5752 (2001).

    CAS  PubMed  PubMed Central  Google Scholar 

  55. Castillo-Lluva, S., Alvarez-Tabarés, I., Weber, I., Steinberg, G. & Pérez-Martin, J. Sustained cell polarity and virulence in the phytopathogenic fungus Ustilago maydis depends on an essential cyclin-dependent kinase from the Cdk5/Pho85 family. J. Cell Sci. 120, 1584–1595 (2007).

    CAS  PubMed  Google Scholar 

  56. Huang, D., Friesen, H. & Andrews, B. Pho85, a multifunctional cyclin-dependent protein kinase in budding yeast. Mol. Microbiol. 66, 303–314 (2007).

    CAS  PubMed  Google Scholar 

  57. Shapiro, R. S. et al. Pho85, Pcl1, and Hms1 signaling governs Candida albicans morphogenesis induced by high temperature or Hsp90 compromise. Curr. Biol. 22, 461–470 (2012).

    CAS  PubMed  Google Scholar 

  58. Zeng, X.-Q. et al. Crosstalk between SNF1 pathway and the peroxisome-mediated lipid metabolism in Magnaporthe oryzae. PLoS ONE 9, e103124 (2014).

    PubMed  PubMed Central  Google Scholar 

  59. Valent, B. & Chumley, F. G. Molecular genetic analysis of the rice blast fungus, Magnaporthe grisea. Annu. Rev. Phytopathol. 29, 443–467 (1991).

    CAS  PubMed  Google Scholar 

  60. Talbot, N. J., Ebbole, D. J. & Hamer, J. E. Identification and characterization of MPG1, a gene involved in pathogenicity from the rice blast fungus Magnaporthe grisea. Plant Cell 5, 1575–1590 (1993).

    CAS  PubMed  PubMed Central  Google Scholar 

  61. Trapnell, C. et al. Transcript assembly and quantification by RNA-Seq reveals unannotated transcripts and isoform switching during cell differentiation. Nat. Biotechnol. 28, 511–515 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  62. Anders, S. & Huber, W. Differential expression analysis for sequence count data. Genome Biol. 11, 106 (2010).

    Google Scholar 

  63. Bray, N. L., Pimentel, H., Melsted, P. & Pachter, L. Near-optimal probabilistic RNA-seq quantification. Nat. Biotechnol. 34, 525–527 (2016).

    CAS  PubMed  Google Scholar 

  64. Mithoe, S. C. et al. Attenuation of pattern recognition receptor signaling is mediated by a MAP kinase kinase kinase. EMBO Rep. 17, 441–454 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  65. Guo, H. et al. Phosphorylation-regulated activation of the Arabidopsis RRS1-R/RPS4 immune receptor complex reveals two distinct effector recognition mechanisms. Cell Host Microbe 27, 769–781.

  66. Zhang, Y. et al. Model-based analysis of ChIP-Seq (MACS). Genome Biol. 9, R137 (2008).

  67. Robinson, J. T. et al. Integrative genomics viewer. Nat. Biotechnol. 29, 24–26 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  68. Berrow, N. S. et al. A versatile ligation-independent cloning method suitable for high-throughput expression screening applications. Nucleic Acids Res. 35, e45 (2007).

    PubMed  PubMed Central  Google Scholar 

  69. Turra, D., Segorbe, D. & Di Pietro, A. Protein kinases in plant-pathogenic fungi: conserved regulators of infection. Annu. Rev. Phytopathol. 52, 267–288 (2014).

    CAS  PubMed  Google Scholar 

  70. Zhao, X. & Xu, J. R. A highly conserved MAPK-docking site in Mst7 is essential for Pmk1 activation in Magnaporthe grisea. Mol. Microbiol. 63, 881–894 (2007).

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

This project was supported by a European Research Council Advanced Investigator award (to N.J.T.) under the European Union’s Seventh Framework Programme FP7/2007-2013/ERC Grant Agreement 294702 GENBLAST, BBSRC grant BB/N009959/1, and by the Gatsby Charitable Foundation. We thank D. MacLean for help with statistical analysis and C. Dean (John Innes Centre) for her group’s guidance with the ChIP–seq analysis.

Author information

Authors and Affiliations

Authors

Contributions

M.O.-R. and N.J.T. conceptualized the project. Experimental analyses were carried out by M.O.-R., N.C-M., M.M-U., A.B.E., M.N., I.E., X.Y., M.J.K., X.Y., C.M. and G.R.L. F.L.H.M designed, and P.D. and F.L.H.M. carried out, the phosphoproteomic analysis. G.V.-P. and B.V. generated the Δrpp3 mutant. Bioinformatic analysis was performed by M.O.-R., D.M.S., B.T., J.C. and V.W. The paper was written by M.O.-R. and N.J.T., with contributions from all authors.

Corresponding authors

Correspondence to Míriam Osés-Ruiz or Nicholas J. Talbot.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information Nature Microbiology thanks Lászlo Nagy, Antonio Di Pietro and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. Peer reviewer reports are available.

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Extended data

Extended Data Fig. 1 The Pmk1 MAP kinase signalling pathway regulates appressorium development in response to surface hydrophobicity.

a. Micrographs to show development of a wild type M. oryzae strain Guy11 expressing histone H1–RFP nuclear marker and septin Sep3–GFP, inoculated on a hydrophobic (HP) or hydrophilic (HL) surface (Scale bar = 10 µm). b. Bar chart to show proportion of Guy11 germlings containing, 1, 2, 3, 4 or more nuclei following incubation for 24 h on HP or HL surfaces. (Grey: HP; White: HL; Circles represent replicates for HP surface; Triangles represent replicates for HL surface) (n = 135 conidia examined in 3 biological replicates; each biological replicate is colour coded; data is presented as Mean ±SEM; Multiple unpaired t-test; HP versus HL; 1 nucleus P = 0.014037). c. Live-cell imaging to show nuclear number and the presence/absence of conidial autophagic cell death of Guy11 on HP, ∆pmk1 mutant on HP, and Guy11 on HL surfaces respectively. Each strain expressed the H1–RFP nuclear marker. d. Rice blast disease symptoms of Guy11 and the ∆pmk1 mutant. Rice seedlings of cultivar CO-39 were spray-inoculated with M. oryzae conidial suspensions of equal concentrations and incubated for 5 days.

Source data

Extended Data Fig. 2 Expression of melanin biosynthesis pathway genes during appressorium development by M. oryzae.

a. Micrograph to show appressorium development by Guy11 and ∆mst12 mutant after incubation for 24 h on HP surface (Bar = 10 µm). b. Rice blast disease symptoms caused by Guy11 and ∆mst12 mutant. Rice seedlings of cultivar CO-39 were spray-inoculated with M. oryzae conidial suspensions of equal concentrations and incubated for 5 days. c. Table to show names and accession numbers of melanin biosynthesis enzymes of M. oryzae. d. Line graph to show mean of normalized counts of gene expression of melanin biosynthesis enzyme-encoding genes during a time course appressorium development for Guy11, ∆pmk1 and ∆mst12 mutants incubated on HP surface between 0 h and 24 h. e. Heatmap to show levels of transcript abundance of genes involved in melanin biosynthesis in a ∆pmk1 mutant compared to Guy11 in conidia germinated on HP surfaces between 0 h and 24 h. f. Heatmap to show levels of transcript abundance of genes involved in melanin biosynthesis in a ∆mst12 mutant compared to Guy11 incubated on HP surface between 0 h and 24 h. Levels of expression are represented as moderated logarithmic fold change (mod_lfc) (blue= downregulated in mutants versus Guy11; red= upregulated in mutants versus Guy11).

Extended Data Fig. 3 Mst12-dependent gene expression and sub-cellular localization of a subset of M. oryzae effectors during appressorium development.

a. Heatmap showing temporal pattern of transcript abundance of 436 Mst12-dependent genes predicted to encode secreted proteins during appressorium development between 0 h and 24 h on a HP surface. b. Heatmap showing levels of transcript abundance of 7 known effector-encoding genes2 in a ∆mst12 mutant during appressorium development. (blue= downregulated in ∆mst12 mutant versus Guy11; red= upregulated in ∆mst12 mutant versus Guy11). Levels of expression are represented as moderated logarithmic fold change (mod_lfc) c. Table to show name and accession number of 7 known effector genes differentially regulated in ∆mst12 compared to Guy11. d. Live-cell imaging of Septin5-GFP expression in appressoria of ∆mst12 and pmk1as mutants 24 h after conidial germination on a HP surface. The pmk1AS mutant was incubated ± 5 µM NA-PP1 added at 6-8 h, once appressorium development was underway. 1NA-PP1 is a specific inhibitor of the analogue-sensitive allele Pmk1 expressed by this mutant3 (Bar = 10 µm). e. Micrographs to show expression of effector Bas2pGFP in a pmk1AS mutant during appressorium formation, 24 h after conidia were inoculated on rice leaf sheath of blast-susceptible rice cultivar Mokoto, in the presence or absence (control) of 5 µM 1Na-PP1 (Bar = 10 µm). f. Micrographs to show cellular localization of exocyst and polarity proteins Exo70-GFP, Sec5-GFP, Sec6-GFP, Spa2-GFP and Mlc1-GFP in the appressorium pore of Guy11 and ∆mst12 following incubation on HP surface for 24 h (Bar = 5 µm). (g-k). Bar charts to show defects in localization of exocyst components during appressorium development of ∆mst12 compared to Guy11 on HP surface at 24 h (n = 180 conidia examined in 3 biological replicates; each biological replicate is colour coded; data are presented as Mean ±SEM).

Source data

Extended Data Fig. 4 ChIP–seq analysis of the Mst12 TF.

a. Micrographs to show cellular localization of Mst12–GFP during appressorium development following incubation on HP surface for 24 h (Bar = 10 µm). b. Rice blast disease symptoms of Guy11, ∆mst12 mutant and complemented strain ∆mst12: Mst12–GFP (T3 and T7 are two independent transformants). Rice seedlings of cultivar CO-39 were spray-inoculated with conidial suspensions of equal concentrations of each M. oryzae strain and incubated for 5 days. c. Table to show number of differentially regulated genes (padj <0.01, mod_lfc>1 or mod_lfc < -1) Guy11 and the ∆mst12 mutant during mycelium growth determined by RNA-seq analysis. d. Table to show over-represented motifs in peaks determined in Mst12 ChIP–seq experiment by Find Individual Motif Occurrence (FIMO – see https://meme-suite.org/meme/doc/fimo.html). Over-representation of each motif in specific sets of DEGs (up and downregulated in mycelium, or expressed during appressorium formation) was analysed using Fisher’s exact test and P values are indicated. The percentage value represents the proportion of the total pool of DEGs represented by that each set of motif-defined gene sets.

Extended Data Fig. 5 Appressorium development and pathogenicity assay of clade 4-associated TF null mutants.

a. Box and Whisker plots to show the number of rice blast disease lesions per 5 m of leaf tissue. Twenty-one-day-old rice seedlings of rice cultivar Co-39 were inoculated with uniform conidial suspensions (5 × 104 conidia ml-1) of ∆rpp1, ∆rpp2, ∆rpp4 and ∆rpp5 mutants and the isogenic wild type M. oryzae strain Guy11 or ∆ku70. Data points are shown in whisker plots which show 25th/75th percentiles, the median and the minimum and maximum values by the ends of the whiskers. A two-tailed non-parametric Mann–Whitney statistical test was conducted to determine significant differences. The plots show the results from 3 biological replications of the experiment and data points are colour coded for each replicate. Error bars show the standard deviation. b. Light micrographs showing appressorium development of Guy11, ∆aclR, ∆rpp1, ∆rpp2, ∆rpp4, ∆rpp5, ∆rpp3, ∆rpp3∆pig1 and ∆hox7 mutants germinated on HP surfaces and observed 24 h following conidial germination (Bar = 10 µm).

Source data

Extended Data Fig. 6 Hox7 operates downstream of Pmk1 MAP kinase to regulate a subset of cellular pathways required for appressorium development and plant infection.

a. Bar chart to show the proportion of germlings of Guy11 and ∆hox7 that develop appressoria (round data points), re-germinated aberrant appressorial swellings (squares) and undifferentiated germlings (triangles) following conidial germination on an HP surface for 24 h (n = 371 conidia examined over 3 biological replicates; biological replicates are colour coded; bars represent Mean ± SEM). b. Table to show the total number of DEGs, upregulated genes (padj <0.01; mod-lfc >1) and downregulated genes (padj <0.01; mod-lfc < -1) of ∆pmk1 versus Guy11, ∆mst12 versus Guy11 and ∆hox7 versus Guy11 at 14 h during appressorium development. c. Heatmap showing levels of relative transcript abundance between ∆pmk1 versus Guy11, ∆mst12 versus Guy11 and ∆hox7 versus Guy11 of a common set of 709 genes. For all heatmaps, the levels of expression are represented as moderated logarithmic fold change (mod_lfc) (blue= downregulated in mutants versus Guy11; red= upregulated in mutants versus Guy11. d. Heatmap showing levels of relative transcript abundance between ∆pmk1 versus Guy11 and ∆hox7 versus Guy11 of 1942 M. oryzae genes. e. Table to show the most representative cellular pathways found among 1942 genes with overlapping expression profiles in ∆pmk1 and ∆hox7. f. Bar charts to show mean normalized counts of gene expression of cyclin, CDK-related, and DNA damage response (DDR) pathway-related genes during appressorium development in Guy11, ∆pmk1, ∆mst12 and ∆hox7 mutants. g. Heatmap to show relative transcript abundance of autophagy-related genes in Δpmk1, ∆mst12 and ∆hox7 mutants compared to Guy11.

Source data

Extended Data Fig. 7 Complementation of Δhox7 mutant and ChIP–seq analysis.

a. Micrographs showing the extent of ∆hox7 complementation upon re-introduction of HOX7 under control of three native promoter lengths (2 kb, 1.5 kb and 1 kb), as shown in the schematic diagram. Conidia were inoculated on HP surface and incubated for 24 h (Bar = 10 µm). b. Bar chart to show the frequency of appressorium formation and range of aberrant infection structures formed by ∆hox7 mutants expressing HOX7 under control of 1 kb promoter (∆hox7:M11, ∆hox7:M13), 1.5 kb promoter (∆hox7:M24) and 2 kb promoter fragment (∆hox7:M32, ∆hox7:M35), n = 813 conidia examined in 3 biological replicates; each biological replicate is colour coded; the data are presented as Mean ± SEM) (legend X-axis- Re-G.App.N.M.: Re-germinated appressoria, non-melanised; B.M.App.: Branched melanised appressoria; App.: Appressoria). c. Micrographs to show appressorium development by Guy11, ∆hox7 and ∆hox7:Hox7-3xFLAG (expressed under control of 2 kb native promoter fragment). Bar = 10 µm. d. Table to show the total number of DEGs, upregulated genes (padj <0.01; mod-lfc >1) and downregulated genes (padj <0.01; mod-lfc < -1) of ∆pmk1 versus Guy11, ∆mst12 versus Guy11 and ∆hox7 versus Guy11 during mycelium development determined by RNA-seq analysis. e. Table to show over-represented motifs in peaks determined in Hox7 ChIP–seq experiment by FIMO (https://meme-suite.org/meme/doc/fimo.html). Over-representation of each motif in specific sets of DEGs (up and downregulated in mycelium, or expressed during appressorium formation) was analysed using Fisher’s exact test and P values are indicated. The percentage value represents the proportion of the total pool of DEGs represented by that each set of motif-defined gene sets.

Source data

Extended Data Fig. 8 Hox7 conservation across fungi.

a. Diagram to show phosphorylated serine residues identified by phosphoproteomic analysis in Hox7 corresponding to serine 126, 158 and 254. KN represents the PFAM domain homeobox KN (PF05920). b. Phylogenetic neighbour-joining tree to show conservation across fungal and oomycete species Aspergillus nidulans, Phythopthora sojae, Phythopthora ramorum, Neurospora crassa, Cryptococcus neoformans, Saccharomyces cerevisiae, Fusarium graminearun, Magnaporthe oryzae and Podospora anserina. c. Amino acid alignment of HOX7 homologues identified using the Homeobox TFs InterPro term IPR001356 in the Fungal Transcription Factor Database http://ftfd.snu.ac.kr of Aspergillus nidulans, Phythopthora sojae, Phythopthora ramorum, Neurospora crassa, Cryptococcus neoformans, Saccharomyces cerevisiae, Fusarium graminearun, Magnaporthe oryzae and Podospora anserina.

Extended Data Fig. 9 Phosphoproteomic analysis reveals Pmk1-dependent phosphorylation of Hox7 in M. oryzae.

a. Diagram to show experimental design for PRM analysis to measure relative normalized intensities (R. N. I.) of peptides associated with serine 126, serine 158 and serine 254 residues of Hox7. The pmk1as conditional mutant was incubated on HP and germinated for 4 h (Baseline), 6 h and 8 h in the presence (+1NA-PP1 crimson bars) or absence (-1NA-PP1 green bars) of the ATP analogue Naphthyl-PP1. Falcon tubes represent sample collection. b. PRM showing R. N. I. of peptide associated with phosphorylated serine at 126 of Hox7 during appressorium development in pmk1as conditional mutant in the presence or absence of 1NA-PP1. Differences were assessed by a two-tailed unpaired t-test, using 2 biological replicates and 2 technical replicates per biological replicate, Mean ± SD. c. PRM showing R. N. I. of peptide associated with phosphorylated serine at 158 of Hox7 during appressorium development in pmk1as conditional mutant in the presence and absence of NA-PP1. Differences were assessed by a two-tailed unpaired t-test, 8 h (P = 0.0316), 2 biological replicates and 2 technical replicates per biological replicate, Mean ±SD. d. PRM showing R. N. I. of peptide associated with phosphorylated serine at 254 of Hox7 during appressorium development in pmk1as conditional mutant in the presence and absence of NA-PP1. Differences were assessed by a two-tailed unpaired t-test, 2 biological replicates and 2 technical replicates per biological replicate, Mean ±SD. Black asterisks below bar charts indicate that some peptides could not be detected in a biological or technical replicate. Red asterisks correspond to the significance of the P value.

Source data

Extended Data Fig. 10 Pmk1 activation by MEK2DD, phenotypes of Δhox7 mutants expressing phosphomimetic alleles of Hox7 and temporal analysis of HOX7 gene expression.

a. Western blot analysis of in vitro phosphorylation experiment of Pmk1 MAPK (GST- Pmk1)- with Hox7 (6xHis-SUMO-Hox7t-342) and Mst12 (6xHis-MBP-Mst12) in the presence or absence of the activated MEKDD.. Proteins were immunoblotted with appropriate antisera (listed on the right). Arrows indicate expected band sizes. Phosphorylation of the Pmk1 MAPK was detected by α-pTEpY antibody in the presence of MEKDD. His-tagged proteins were detected with α-His antibody. b. Micrographs to show representative phenotypes of germlings of Δhox7 mutants expressing phosphomimetic alleles of Hox7 incubated on HP surfaces for 24 h, as quantified in Fig. 7b (Bar= 10 µm). c. Line graph fo show temporal analysis of HOX7 transcript abundance in wild type Guy11 (blue), and ∆pmk1 (green) and ∆mst12 (purple) mutants during a time course of appressorium development measured by RNA-seq analysis.

Source data

Supplementary information

Supplementary Information

Supplementary Figs. 1–10 and Source data for Supplementary Fig. 7. Supplementary Table 7. Gene name, accession number, reported mutant phenotype and reference of each Clade 4 TF. Supplementary Table 11. Peptides used in the PRM assay. Supplementary Table 13. Primers used in this study.

Reporting Summary

Supplementary Table 1

RNA-seq analysis of a time course of appressorium development in Guy11 on an HL versus HP surface.

Supplementary Table 2

RNA-seq analysis of a time course of appressorium development in Guy11 versus ∆pmk1 on an HP surface.

Supplementary Table 3

DEGs as classified in the Venn diagram in Fig. 1d.

Supplementary Table 4

RNA-seq analysis of a time course of appressorium development in Guy11 versus ∆mst12 on an HP surface

Supplementary Table 5

DEGs as classified in the Venn diagram in Fig. 2e.

Supplementary Table 6

RNA-seq and ChIP–seq analysis of mycelium of Guy11 versus ∆mst12.

Supplementary Table 8

RNA-seq analysis of appressorium development in Guy11 versus ∆hox7 on an HP surface at 14h.

Supplementary Table 9

DEGs as classified in the Venn diagram in Fig. 5c.

Supplementary Table 10

RNA-seq and ChIP–seq analysis of mycelium of Guy11 versus the ∆hox7 mutant.

Supplementary Table 12

Spectral count data for Pmk1 in vitro phosphorylation assay of Hox7 and Mst12.

Peer Review File

Source data

Source Data Fig. 3

Numerical data for the graphs in Fig. 3h

Source Data Fig. 5

Numerical data for the graphs in Fig. 5b,c,e–g.

Source Data Fig. 7

Numerical data for the graph in Fig. 7b.

Source Data Extended Data Fig. 1

Numerical data for the graphs in Extended Data Fig. 1b.

Source Data Extended Data Fig. 3

Numerical data for the graphs in Extended Data Fig. 3g–k.

Source Data Extended Data Fig. 5

Numerical data for the graphs in Extended Data Fig. 5a.

Source Data Extended Data Fig. 6

Numerical data for the graphs in Extended Data Fig. 6a.

Source Data Extended Data Fig. 7

Numerical data for the graphs in Extended Data Fig. 7b.

Source Data Extended Data Fig. 9

Numerical data for the graphs in Extended Data Fig. 9b–d.

Source Data Extended Data Fig. 10

Un-cropped gels and autoradiographs for Extended Data Fig. 10a.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Osés-Ruiz, M., Cruz-Mireles, N., Martin-Urdiroz, M. et al. Appressorium-mediated plant infection by Magnaporthe oryzae is regulated by a Pmk1-dependent hierarchical transcriptional network. Nat Microbiol 6, 1383–1397 (2021). https://doi.org/10.1038/s41564-021-00978-w

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41564-021-00978-w

This article is cited by

Search

Quick links

Nature Briefing Microbiology

Sign up for the Nature Briefing: Microbiology newsletter — what matters in microbiology research, free to your inbox weekly.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing: Microbiology