#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

Evidence for a Transketolase-Mediated Metabolic Checkpoint Governing Biotrophic Growth in Rice Cells by the Blast Fungus


The blast fungus Magnaporthe oryzae destroys rice and wheat harvests and could compromise global food security. Following penetration into the rice cell, M. oryzae elaborates bulbous invasive hyphae that grow in living rice cells for most of the infection cycle without causing disease symptoms. Little is known about the physiological processes governing this important biotrophic stage of fungal growth. Here, we used gene functional analysis to show how the primary metabolic enzyme transketolase is essential for hyphal growth in rice cells. Loss of transketolase did not affect the ability of the fungus to gain entry into rice cells, but invasive hyphal growth was curtailed in transketolase null mutants. Biotrophic growth was restored in transketolase mutants by the addition of exogenous ATP. We conclude that M. oryzae metabolism is dedicated to metabolizing glucose through transketolase in planta in order to provide ATP as a trigger for biotrophic growth and infection. This work is significant because it reveals important—but previously unknown—metabolic strategies employed by M. oryzae to facilitate rice infection. These strategies might be open to abrogation by chemical or biological means and are likely relevant to other rapidly proliferating intracellular pathogens.


Vyšlo v časopise: Evidence for a Transketolase-Mediated Metabolic Checkpoint Governing Biotrophic Growth in Rice Cells by the Blast Fungus. PLoS Pathog 10(9): e32767. doi:10.1371/journal.ppat.1004354
Kategorie: Research Article
prolekare.web.journal.doi_sk: https://doi.org/10.1371/journal.ppat.1004354

Souhrn

The blast fungus Magnaporthe oryzae destroys rice and wheat harvests and could compromise global food security. Following penetration into the rice cell, M. oryzae elaborates bulbous invasive hyphae that grow in living rice cells for most of the infection cycle without causing disease symptoms. Little is known about the physiological processes governing this important biotrophic stage of fungal growth. Here, we used gene functional analysis to show how the primary metabolic enzyme transketolase is essential for hyphal growth in rice cells. Loss of transketolase did not affect the ability of the fungus to gain entry into rice cells, but invasive hyphal growth was curtailed in transketolase null mutants. Biotrophic growth was restored in transketolase mutants by the addition of exogenous ATP. We conclude that M. oryzae metabolism is dedicated to metabolizing glucose through transketolase in planta in order to provide ATP as a trigger for biotrophic growth and infection. This work is significant because it reveals important—but previously unknown—metabolic strategies employed by M. oryzae to facilitate rice infection. These strategies might be open to abrogation by chemical or biological means and are likely relevant to other rapidly proliferating intracellular pathogens.


Zdroje

1. WilsonRA, TalbotNJ (2009) Under pressure: investigating the biology of plant infection by Magnaporthe oryzae. Nat Rev Microbiol 7: 185–195.

2. FernandezJ, WilsonRA (2012) Why no feeding frenzy? Mechanisms of nutrient acquisition and utilization during Infection by the rice blast fungus Magnaporthe oryzae. Mol Plant-Microbe Interact 25: 1286–1293.

3. FisherMC, HenkDA, BriggsCJ, BrownsteinJS, MadoffLC, et al. (2012) Emerging fungal threats to animal, plant and ecosystem health. Nature 484: 186–194.

4. KupferschmidtK (2012) Attack of the clones. Science 337: 636–638.

5. DagdasYF, YoshinoK, DagdasG, RyderLS, BielskaE, et al. (2012) Septin-mediated plant cell invasion by the rice blast fungus, Magnaporthe oryzae. Science 336: 1590–1595.

6. RyderLS, DagdasYF, MentlakTA, KershawMJ, ThorntonCR, et al. (2013) NADPH oxidases regulate septin-mediated cytoskeletal remodeling during plant infection by the rice blast fungus. Proc Natl Acad Sci USA 110: 3179–3184.

7. SaundersDG, DagdasYF, TalbotNJ (2010) Spatial uncoupling of mitosis and cytokinesis during appressorium-mediated plant Infection by the rice blast fungus Magnaporthe oryzae. Plant Cell 22: 2417–2428.

8. Veneault-FourreyC, BarooahM, EganM, WakleyG, TalbotNJ (2006) Autophagic fungal cell death is necessary for infection by the rice blast fungus. Science 312: 580–583.

9. SaundersDG, AvesSJ, TalbotNJ (2010) Cell cycle-mediated regulation of plant infection by the rice blast fungus. Plant Cell 22: 497–507.

10. HowardRJ, ValentB (1996) Breaking and entering: Host penetration by the fungal rice blast pathogen Magnaporthe grisea. Annu Rev Microbiol 50: 491–512.

11. de JongJC, McCormackBJ, SmirnoffN, TalbotNJ (1997) Glycerol generates turgor in rice blast. Nature 389: 244–244.

12. KankanalaP, CzymmekK, ValentB (2007) Roles for rice membrane dynamics and plasmodesmata during biotrophic invasion by the blast fungus. Plant Cell 19: 706–724.

13. ThinesE, WeberRW, TalbotNJ (2000) MAP kinase and protein kinase A-dependent mobilization of triacylglycerol and glycogen during appressorium turgor generation by Magnaporthe grisea. Plant Cell 12: 1703–1718.

14. WangZY, JenkinsonJM, HolcombeLJ, SoanesDM, Veneault-FourreyC, et al. (2005) The molecular biology of appressorium turgor generation by the rice blast fungus Magnaporthe grisea. Biochem Soc Trans 33: 384–388.

15. WangZY, SoanesDM, KershawMJ, TalbotNJ (2007) Functional analysis of lipid metabolism in Magnaporthe grisea reveals a requirement for peroxisomal fatty acid beta-oxidation during appressorium-mediated plant infection. Mol Plant-Microbe Interact 20: 475–491.

16. WangZY, ThorntonCR, KershawMJ, DebaoL, TalbotNJ (2003) The glyoxylate cycle is required for temporal regulation of virulence by the plant pathogenic fungus Magnaporthe grisea. Mol Microbiol 47: 1601–1612.

17. BhambraGK, WangZY, SoanesDM, WakleyGE, TalbotNJ (2006) Peroxisomal carnitine acetyl transferase is required for elaboration of penetration hyphae during plant infection by Magnaporthe grisea. Mol Microbiol 61: 46–60.

18. Ramos-PamplonaM, NaqviNI (2006) Host invasion during rice-blast disease requires carnitine-dependent transport of peroxisomal acetyl-CoA. Mol Microbiol 61: 61–75.

19. BhadauriaV, BannizaS, VandenbergA, SelvarajG, WeiY (2012) Peroxisomal alanine: glyoxylate aminotransferase AGT1 is indispensable for appressorium function of the rice blast pathogen, Magnaporthe oryzae. PLOS One 7: e36266.

20. PatkarRN, Ramos-PamplonaM, GuptaAP, FanY, NaqviNI (2012) Mitochondrial β-oxidation regulates organellar integrity and is necessary for conidial germination and invasive growth in Magnaporthe oryzae. Mol Microbiol 86: 1345–1363.

21. WilsonRA, JenkinsonJM, GibsonRP, LittlechildJA, WangZY, et al. (2007) Tps1 regulates the pentose phosphate pathway, nitrogen metabolism and fungal virulence. EMBO J 26: 3673–3685.

22. WilsonRA, GibsonRP, QuispeCF, LittlechildJA, TalbotNJ (2010) An NADPH-dependent genetic switch regulates plant infection by the rice blast fungus. Proc Natl Acad Sci USA 107: 21902–21907.

23. FernandezJ, WrightJD, HartlineD, QuispeCF, MadayiputhiyaN, et al. (2012) Principles of carbon catabolite repression in the rice blast fungus: Tps1, Nmr1–3, and a MATE–Family Pump regulate glucose metabolism during Infection. PLoS Genet 8: e1002673.

24. FernandezJ, WilsonRA (2014) Characterizing roles for the glutathione reductase, thioredoxin reductase and thioredoxin peroxidase-encoding genes of Magnaporthe oryzae during rice blast disease. PLoS One 9: e87300.

25. FernandezJ, WilsonRA (2014) Cells in cells: morphogenetic and metabolic strategies conditioning rice infection by the blast fungus Magnaporthe oryzae. Protoplasma 251: 37–47.

26. CollerHA (2011) The essence of quiescence. 334: 1074–1075.

27. KlosinskaMM, CrutchfieldCA, BradleyPH, RabinowitzJD, BroachJR (2012) Yeast cells can access distinct quiescent states. Genes Dev 25: 336–349.

28. Berg JM, Tymoczko JL, Stryer L (2011) Biochemistry. Basingstoke, NY: W.H. Freeman & Co Ltd.

29. DeanRA, TalbotNJ, EbboleDJ, FarmanML, MitchellTK, et al. (2005) The genome sequence of the rice blast fungus Magnaporthe grisea. Nature 434: 980–986.

30. LuC, BrauerMJ, BotsteinD (2009) Slow growth induces heat-shock resistance in normal and respiratory-deficient yeast. Mol Biol Cell 20: 891–903.

31. CollerHA, SangL, RobertsJM (2006) A new description of cellular quiescence. PLoS Biol 4: e83.

32. BarbetNC, SchneiderU, HelliwellSB, StansfieldI, TuiteMF, et al. (1996) TOR controls translation initiation and early G1 progression in yeast. Mol Biol Cell 7: 25–42.

33. LoewithR, HallMN (2011) Target of rapamycin (TOR) in nutrient signaling and growth control. Genetics 189: 1177–1201.

34. De VirgilioC (2012) The essence of yeast quiescence. FEMS Microbiol Rev 36: 306–339.

35. HardwickJS, KuruvillaFG, TongJK, ShamjiAF, SchreiberSL (1999) Rapamycin-modulated transcription defines the subset of nutrient-sensitive signaling pathways directly controlled by the Tor proteins. Proc Natl Acad Sci U S A 96: 14866–14870.

36. Ramírez-ValleF, BraunsteinS, ZavadilJ, FormentiSC, SchneiderRJ (2008) eIF4GI links nutrient sensing by mTOR to cell proliferation and inhibition of autophagy. J Cell Biol 181: 293–307.

37. FernandezJ, YangKT, CornwellKM, WrightJD, WilsonRA (2013) Growth in rice cells requires de novo purine biosynthesis by the blast fungus Magnaporthe oryzae. Sci Rep 3: 2398.

38. ShanerNC, CampbellRE, SteinbachPA, GiepmansBN, PalmerAE, et al. (2004) Improved monomeric red, orange and yellow fluorescent proteins derived from Discosoma sp. red fluorescent protein. Nat Biotechnol 22: 1567–1572.

39. FranceschettiM, BuenoE, WilsonRA, TuckerSL, Gómez-MenaC, et al. (2011) Fungal virulence and development is regulated by alternative pre-mRNA 3′ end processing in Magnaporthe oryzae. PLoS Pathog 7: e1002441.

40. PollakN, DölleC, ZieglerM (2007) The power to reduce: pyridine nucleotides – small molecules with a multitude of functions. Biochem J 402: 205–218.

41. YingW (2008) NAD+/NADH and NADP+/NADPH in cellular functions and cell death: regulation and biological consequences. Antioxidants & redox signaling 10: 179–206.

42. DennisPB, JaeschkeA, SaitohM, FowlerB, KozmaSC, et al. (2001) Mammalian TOR: a homeostatic ATP sensor. Science 294: 1102–1105.

43. GwinnDM, ShackelfordDB, EganDF, MihaylovaMM, MeryA, et al. (2008) AMPK phosphorylation of raptor mediates a metabolic checkpoint. Mol Cell 30: 214–226.

44. TakuboK, NagamatsuG, KobayashiCI, Nakamura-IshizuA, KobayashiH, et al. (2013) Regulation of glycolysis by Pdk functions as a metabolic checkpoint for cell-cycle quiescence in hematopoietic stem cells. Cell Stem Cell 12: 49–61.

45. NakashimaA, MarukiY, ImamuraY, KondoC, KawamataT, et al. (2008) The Yeast Tor Signaling Pathway Is Involved in G2/M Transition via Polo- Kinase. PLoS ONE 3 (5) e2223.

46. GréenA, LönnA, PetersonKH, OllingerK, RundquistI (2010) Translocation of histone H1 subtypes between chromatin and cytoplasm during mitosis in normal human fibroblasts. Cytometry A 77 (5) 478–484.

47. BhattacharyaD, MazumderA, MiriamSA, ShivashankarGV (2006) EGFP-tagged core and linker histones diffuse via distinct mechanisms within living cells. Biophys J 91: 2326–2336.

48. SchulzeA, HarrisAL (2012) How cancer metabolism is tuned for proliferation and vulnerable to disruption. Nature 491: 364–373.

49. O'NeillLAJ, HardieDG (2013) Metabolism of inflammation limited by AMPK and pseudo-starvation. Nature 493: 346–355.

50. GerrietsVA, RathmellJC (2012) Metabolic pathways in T cell fate and function. Trends Immunol 33: 168–173.

51. WangR, GreenDR (2012) Metabolic checkpoints in activated T cells. Nat Immunol 13: 907–915.

52. Diaz-MoralliS, Tarrado-CastellarnauM, AlendaC, CastellsA, CascanteM (2011) Transketolase-like 1 expression is modulated during colorectal cancer progression and metastasis formation. PLOS One 6: e25323.

53. LangbeinS, ZerilliM, Zur HausenA, StaigerW, Rensch-BoschertK, et al. (2006) Expression of transketolase TKTL1 predicts colon and urothelial cancer patient survival: Warburg effect reinterpreted. Br J Cancer 94: 578–585.

54. BorosLG, PuigjanerJ, CascanteM, LeeWN, BrandesJL, et al. (1997) Oxythiamine and dehydroepiandrosterone inhibit the nonoxidative synthesis of ribose and tumor cell proliferation. Cancer Res 57: 4242–4248.

55. Comín-AnduixB, BorenJ, MartinezS, MoroC, CentellesJJ, et al. (2001) The effect of thiamine supplementation on tumour proliferation. A metabolic control analysis study. Eur J Biochem 268: 4177–4182.

56. DuMX, SimJ, FangL, YinZ, KohS, et al. (2004) Identification of novel small-molecule inhibitors for human transketolase by high-throughput screening with fluorescent intensity (FLINT) assay. J Biomol Screen 9: 427–433.

57. WullschlegerS, LoewithR, HallMN (2006) TOR signaling in growth and metabolism. Cell 124: 471–484.

58. TalbotNJ, EbboleDJ, HamerJE (1993) Identification and characterization of MPG1, a gene involved in pathogenicity from the rice blast fungus Magnaporthe grisea. Plant Cell 5: 1575–1590.

59. WilsonRA, FernandezJ, QuispeCF, GradnigoJ, SengA, et al. (2012) Towards defining nutrient conditions encountered by the rice blast fungus during host infection. PLoS ONE 7: e47392.

60. ChiMH, ParkSY, KimS, LeeYH (2009) A novel pathogenicity gene is required in the rice blast fungus to suppress the basal defenses of the host. PLoS Pathogen 5: e1000401.

61. BajadS, ShulaevV (2011) LC-MS-based metabolomics. Methods Mol Biol 708: 213–228.

Štítky
Hygiena a epidemiológia Infekčné lekárstvo Laboratórium

Článok vyšiel v časopise

PLOS Pathogens


2014 Číslo 9
Najčítanejšie tento týždeň
Najčítanejšie v tomto čísle
Kurzy

Zvýšte si kvalifikáciu online z pohodlia domova

Získaná hemofilie - Povědomí o nemoci a její diagnostika
nový kurz

Eozinofilní granulomatóza s polyangiitidou
Autori: doc. MUDr. Martina Doubková, Ph.D.

Všetky kurzy
Prihlásenie
Zabudnuté heslo

Zadajte e-mailovú adresu, s ktorou ste vytvárali účet. Budú Vám na ňu zasielané informácie k nastaveniu nového hesla.

Prihlásenie

Nemáte účet?  Registrujte sa

#ADS_BOTTOM_SCRIPTS#