#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

The Genome of and the Basis of Host-Microsporidian Interactions


Microsporidia are obligate intracellular parasites with the smallest known eukaryotic genomes. Although they are increasingly recognized as economically and medically important parasites, the molecular basis of microsporidian pathogenicity is almost completely unknown and no genetic manipulation system is currently available. The fish-infecting microsporidian Spraguea lophii shows one of the most striking host cell manipulations known for these parasites, converting host nervous tissue into swollen spore factories known as xenomas. In order to investigate the basis of these interactions between microsporidian and host, we sequenced and analyzed the S. lophii genome. Although, like other microsporidia, S. lophii has lost many of the protein families typical of model eukaryotes, we identified a number of gene family expansions including a family of leucine-rich repeat proteins that may represent pathogenicity factors. Building on our comparative genomic analyses, we exploited the large numbers of spores that can be obtained from xenomas to identify potential effector proteins experimentally. We used complex-mix proteomics to identify proteins released by the parasite upon germination, resulting in the first experimental isolation of putative secreted effector proteins in a microsporidian. Many of these proteins are not related to characterized pathogenicity factors or indeed any other sequences from outside the Microsporidia. However, two of the secreted proteins are members of a family of RICIN B-lectin-like proteins broadly conserved across the phylum. These proteins form syntenic clusters arising from tandem duplications in several microsporidian genomes and may represent a novel family of conserved effector proteins. These computational and experimental analyses establish S. lophii as an attractive model system for understanding the evolution of host-parasite interactions in microsporidia and suggest an important role for lineage-specific innovations and fast evolving proteins in the evolution of the parasitic microsporidian lifecycle.


Vyšlo v časopise: The Genome of and the Basis of Host-Microsporidian Interactions. PLoS Genet 9(8): e32767. doi:10.1371/journal.pgen.1003676
Kategorie: Research Article
prolekare.web.journal.doi_sk: https://doi.org/10.1371/journal.pgen.1003676

Souhrn

Microsporidia are obligate intracellular parasites with the smallest known eukaryotic genomes. Although they are increasingly recognized as economically and medically important parasites, the molecular basis of microsporidian pathogenicity is almost completely unknown and no genetic manipulation system is currently available. The fish-infecting microsporidian Spraguea lophii shows one of the most striking host cell manipulations known for these parasites, converting host nervous tissue into swollen spore factories known as xenomas. In order to investigate the basis of these interactions between microsporidian and host, we sequenced and analyzed the S. lophii genome. Although, like other microsporidia, S. lophii has lost many of the protein families typical of model eukaryotes, we identified a number of gene family expansions including a family of leucine-rich repeat proteins that may represent pathogenicity factors. Building on our comparative genomic analyses, we exploited the large numbers of spores that can be obtained from xenomas to identify potential effector proteins experimentally. We used complex-mix proteomics to identify proteins released by the parasite upon germination, resulting in the first experimental isolation of putative secreted effector proteins in a microsporidian. Many of these proteins are not related to characterized pathogenicity factors or indeed any other sequences from outside the Microsporidia. However, two of the secreted proteins are members of a family of RICIN B-lectin-like proteins broadly conserved across the phylum. These proteins form syntenic clusters arising from tandem duplications in several microsporidian genomes and may represent a novel family of conserved effector proteins. These computational and experimental analyses establish S. lophii as an attractive model system for understanding the evolution of host-parasite interactions in microsporidia and suggest an important role for lineage-specific innovations and fast evolving proteins in the evolution of the parasitic microsporidian lifecycle.


Zdroje

1. KeelingP (2009) Five Questions about Microsporidia. PLoS Pathog 5: e1000489.

2. SinghT, BhatMM, KhanMA (2012) Microsporidiosis in the Silkworm, Bombyx mori L. (Lepidoptera: Bombycidae). Pertanika Journal of Tropical Agricultural Science 35: 387–406.

3. Speare DJ, Lovy J (2011) Loma salmonae related species. In: Woo PTK, Buchmann K, editors. Fish Parasites: Pathobiology and Protection. UK: CABI Publishing.

4. SchotteliusJ, SchmetzC, KockNP, SchulerT, SobottkaI, et al. (2000) Presentation by scanning electron microscopy of the life cycle of microsporidia of the genus Encephalitozoon. Microbes Infect 2: 1401–1406.

5. KatinkaMD, DupratS, CornillotE, MetenierG, ThomaratF, et al. (2001) Genome sequence and gene compaction of the eukaryote parasite Encephalitozoon cuniculi. Nature 414: 450–453.

6. CorradiN, PombertJF, FarinelliL, DidierES, KeelingPJ (2010) The complete sequence of the smallest known nuclear genome from the microsporidian Encephalitozoon intestinalis. Nat Commun 1: 77.

7. CorradiN, HaagKL, PombertJF, EbertD, KeelingPJ (2009) Draft genome sequence of the Daphnia pathogen Octosporea bayeri: insights into the gene content of a large microsporidian genome and a model for host-parasite interactions. Genome Biol 10: R106.

8. HeinzE, WilliamsTA, NakjangS, NoëlCJ, SwanDC, et al. (2012) The Genome of the Obligate Intracellular Parasite Trachipleistophora hominis: New Insights into Microsporidian Genome Dynamics and Reductive Evolution. PLoS Pathog 8: e1002979.

9. WilliamsBA, LeeRC, BecnelJJ, WeissLM, FastNM, et al. (2008) Genome sequence surveys of Brachiola algerae and Edhazardia aedis reveal microsporidia with low gene densities. BMC Genomics 9: 200.

10. KrylovDM, WolfYI, RogozinIB, KooninEV (2003) Gene loss, protein sequence divergence, gene dispensability, expression level, and interactivity are correlated in eukaryotic evolution. Genome Res 13: 2229–2235.

11. DöfleinF (1898) Studien zur Naturgeschichte der Protozoen. III. Über die Myxosporidien. Zool Jahrb Abt Anat 11: 281–350.

12. WeidnerE, ByrdW (1982) The microsporidian spore invasion tube. II. Role of calcium in the activation of invasion tube discharge. J Cell Biol 93: 970–975.

13. FreemanMA, YokoyamaH, OsadaA, YoshidaT, YamanobeA, et al. (2011) Spraguea (Microsporida: Spraguidae) infections in the nervous system of the Japanese anglerfish, Lophius litulon (Jordan), with comments on transmission routes and host pathology. J Fish Dis 34: 445–452.

14. CañásL, SampedroMP, FariñaAC (2010) Influence of host biological features on macroparasites of the two European anglerfish species, Lophius piscatorius and Lophius budegassa, off north and northwest Spain. J Parasitol 96: 191–193.

15. PutzRE, HoffmanGL, DunbarCE (1965) 2 New Species of Plistophora (Microsporides) from North American Fish with a Synopsis of Microsporidea of Freshwater and Euryhaline Fishes. Journal of Protozoology 12: 228–236.

16. HinkleG, MorrisonHG, SoginML (1997) Genes coding for reverse transcriptase, DNA-directed RNA polymerase, and chitin synthase from the microsporidian Spraguea lophii. Biol Bull 193: 250–251.

17. MansourL, CheikaliC, DesaunaisP, CoulonJP, DaubinJ, et al. (2004) Description of an ultrathin multiwire proportional chamber-based detector and application to the characterization of the Spraguea lophii (Microspora) two-dimensional genome fingerprint. Electrophoresis 25: 3365–3377.

18. BiderreC, PagesM, MetenierG, DavidD, BataJ, et al. (1994) On small genomes in eukaryotic organisms: molecular karyotypes of two microsporidian species (Protozoa) parasites of vertebrates. C R Acad Sci III 317: 399–404.

19. MansourL, Ben HassineOK, VivaresCP, CornillotE (2013) Spraguea lophii (Microsporidia) parasite of the teleost fish, Lophius piscatorius from Tunisian coasts: Evidence for an extensive chromosome length polymorphism. Parasitol Int 62: 66–74.

20. LiL, StoeckertCJJr, RoosDS (2003) OrthoMCL: identification of ortholog groups for eukaryotic genomes. Genome Res 13: 2178–2189.

21. KeelingPJ, CorradiN, MorrisonHG, HaagKL, EbertD, et al. (2010) The Reduced Genome of the Parasitic Microsporidian Enterocytozoon bieneusi Lacks Genes for Core Carbon Metabolism. Genome Biology and Evolution 2: 304–309.

22. CuomoCA, DesjardinsCA, BakowskiMA, GoldbergJ, MaAT, et al. (2012) Microsporidian genome analysis reveals evolutionary strategies for obligate intracellular growth. Genome Res 12: 2478–2488.

23. CornmanRS, ChenYP, SchatzMC, StreetC, ZhaoY, et al. (2009) Genomic analyses of the microsporidian Nosema ceranae, an emergent pathogen of honey bees. PLoS Pathog 5: e1000466.

24. AkiyoshiDE, MorrisonHG, LeiS, FengX, ZhangQ, et al. (2009) Genomic survey of the non-cultivatable opportunistic human pathogen, Enterocytozoon bieneusi. PLoS Pathog 5: e1000261.

25. El AlaouiH, BataJ, BauchartD, DoreJC, VivaresCP (2001) Lipids of three microsporidian species and multivariate analysis of the host-parasite relationship. J Parasitol 87: 554–559.

26. LangeBM, RujanT, MartinW, CroteauR (2000) Isoprenoid biosynthesis: the evolution of two ancient and distinct pathways across genomes. Proc Natl Acad Sci U S A 97: 13172–13177.

27. DrinnenbergIA, WeinbergDE, XieKT, MowerJP, WolfeKH, et al. (2009) RNAi in budding yeast. Science 326: 544–550.

28. EisenbergD, GillHS, PflueglGM, RotsteinSH (2000) Structure-function relationships of glutamine synthetases. Biochim Biophys Acta 1477: 122–145.

29. SuarezI, BodegaG, FernandezB (2002) Glutamine synthetase in brain: effect of ammonia. Neurochem Int 41: 123–142.

30. Huss HH (1995) Quality and quality changes in fresh fish. Food and agriculture organization of the united nations.

31. PhadtareS, AlsinaJ, InouyeM (1999) Cold-shock response and cold-shock proteins. Curr Opin Microbiol 2: 175–180.

32. SoanesDM, TalbotNJ (2010) Comparative genome analysis reveals an absence of leucine-rich repeat pattern-recognition receptor proteins in the kingdom Fungi. PLoS One 5: e12725.

33. ButlerG, RasmussenMD, LinMF, SantosMA, SakthikumarS, et al. (2009) Evolution of pathogenicity and sexual reproduction in eight Candida genomes. Nature 459: 657–662.

34. BaileyTL, ElkanC (1994) Fitting a mixture model by expectation maximization to discover motifs in biopolymers. Proc Int Conf Intell Syst Mol Biol 2: 28–36.

35. PuntaM, CoggillPC, EberhardtRY, MistryJ, TateJ, et al. (2012) The Pfam protein families database. Nucleic Acids Res 40: D290–301.

36. KroghA, LarssonB, von HeijneG, SonnhammerEL (2001) Predicting transmembrane protein topology with a hidden Markov model: application to complete genomes. J Mol Biol 305: 567–580.

37. EmanuelssonO, BrunakS, von HeijneG, NielsenH (2007) Locating proteins in the cell using TargetP, SignalP and related tools. Nat Protoc 2: 953–71.

38. KobeB, DeisenhoferJ (1995) A structural basis of the interactions between leucine-rich repeats and protein ligands. Nature 374: 183–186.

39. BertholdJ, SchenkovaK, RamosS, MiuraY, FurukawaM, et al. (2008) Characterization of RhoBTB-dependent Cul3 ubiquitin ligase complexes–evidence for an autoregulatory mechanism. Exp Cell Res 314: 3453–3465.

40. DeitschKW, LukehartSA, StringerJR (2009) Common strategies for antigenic variation by bacterial, fungal and protozoan pathogens. Nat Rev Microbiol 7: 493–503.

41. MarcelloL, BarryJD (2007) Analysis of the VSG gene silent archive in Trypanosoma brucei reveals that mosaic gene expression is prominent in antigenic variation and is favored by archive substructure. Genome Res 17: 1344–1352.

42. RautaPR, NayakB, DasS (2012) Immune system and immune responses in fish and their role in comparative immunity study: a model for higher organisms. Immunol Lett 148: 23–33.

43. GrabherrMG, HaasBJ, YassourM, LevinJZ, ThompsonDA, et al. (2011) Full-length transcriptome assembly from RNA-Seq data without a reference genome. Nat Biotechnol 29: 644–652.

44. LiB, DeweyCN (2011) RSEM: accurate transcript quantification from RNA-Seq data with or without a reference genome. BMC Bioinformatics 12: 323.

45. AltschulSF, GishW, MillerW, MyersEW, LipmanDJ (1990) Basic local alignment search tool. J Mol Biol 215: 403–410.

46. CorradiN, GangaevaA, KeelingPJ (2008) Comparative profiling of overlapping transcription in the compacted genomes of microsporidia Antonospora locustae and Encephalitozoon cuniculi. Genomics 91: 388–93.

47. WilliamsBA, SlamovitsCH, PatronNJ, FastNM, KeelingPJ (2005) A high frequency of overlapping gene expression in compacted eukaryotic genomes. Proc Natl Acad Sci U S A 102: 10936–10941.

48. KeelingPJ, SlamovitsCH (2004) Simplicity and complexity of microsporidian genomes. Eukaryot Cell 3: 1363–1369.

49. LeeRCH, GillEE, RoySW, FastNM (2010) Constrained Intron Structures in a Microsporidian. Molecular Biology and Evolution 27: 1979–1982.

50. GrisdaleC, BowersL, DidierE, FastN (2013) Transcriptome analysis of the parasite Encephalitozoon cuniculi: an in-depth examination of pre-mRNA splicing in a reduced eukaryote. BMC Genomics 14: 207.

51. PleshingerJ, WeidnerE (1985) The microsporidian spore invasion tube. IV. Discharge activation begins with pH-triggered Ca2+ influx. J Cell Biol 100: 1834–1838.

52. FrixioneE, RuizL, UndeenAH (1994) Monovalent cations induce microsporidian spore germination in vitro. J Eukaryot Microbiol 41: 464–468.

53. FrixioneE, RuizL, CerbonJ, UndeenAH (1997) Germination of Nosema algerae (Microspora) spores: conditional inhibition by D2O, ethanol and Hg2+ suggests dependence of water influx upon membrane hydration and specific transmembrane pathways. J Eukaryot Microbiol 44: 109–116.

54. DolgikhVV, SenderskiyIV, PavlovaOA, NaumovAM, BeznoussenkoGV (2011) Immunolocalization of an alternative respiratory chain in Antonospora (Paranosema) locustae spores: mitosomes retain their role in microsporidial energy metabolism. Eukaryot Cell 10: 588–593.

55. BrossonD, KuhnL, PrensierG, VivaresCP, TexierC (2005) The putative chitin deacetylase of Encephalitozoon cuniculi: a surface protein implicated in microsporidian spore-wall formation. FEMS Microbiol Lett 247: 81–90.

56. BrossonD, KuhnL, DelbacF, GarinJ, P. VivarèsC, et al. (2006) Proteomic analysis of the eukaryotic parasite Encephalitozoon cuniculi (microsporidia): a reference map for proteins expressed in late sporogonial stages. PROTEOMICS 6: 3625–3635.

57. UrchJE, Hurtado-GuerreroR, BrossonD, LiuZ, EijsinkVG, et al. (2009) Structural and functional characterization of a putative polysaccharide deacetylase of the human parasite Encephalitozoon cuniculi. Protein Sci 18: 1197–1209.

58. LoukasA, MaizelsRM (2000) Helminth C-type lectins and host-parasite interactions. Parasitol Today 16: 333–339.

59. PetriWAJr, HaqueR, MannBJ (2002) The bittersweet interface of parasite and host: lectin-carbohydrate interactions during human invasion by the parasite Entamoeba histolytica. Annu Rev Microbiol 56: 39–64.

60. KamperJ, KahmannR, BolkerM, MaLJ, BrefortT, et al. (2006) Insights from the genome of the biotrophic fungal plant pathogen Ustilago maydis. Nature 444: 97–101.

61. SchornackS, HuitemaE, CanoLM, BozkurtTO, OlivaR, et al. (2009) Ten things to know about oomycete effectors. Mol Plant Pathol 10: 795–803.

62. SargeantTJ, MartiM, CalerE, CarltonJM, SimpsonK, et al. (2006) Lineage-specific expansion of proteins exported to erythrocytes in malaria parasites. Genome Biol 7: R12.

63. NoelCJ, DiazN, Sicheritz-PontenT, SafarikovaL, TachezyJ, et al. (2010) Trichomonas vaginalis vast BspA-like gene family: evidence for functional diversity from structural organisation and transcriptomics. BMC Genomics 11: 99.

64. AbramyanJ, StajichJE (2012) Species-specific chitin-binding module 18 expansion in the amphibian pathogen Batrachochytrium dendrobatidis. MBio 3: e00150–00112.

65. SpanuPD, AbbottJC, AmselemJ, BurgisTA, SoanesDM, et al. (2010) Genome expansion and gene loss in powdery mildew fungi reveal tradeoffs in extreme parasitism. Science 330: 1543–1546.

66. DiaN, LavieL, MetenierG, ToguebayeBS, VivaresCP, et al. (2007) InterB multigenic family, a gene repertoire associated with subterminal chromosome regions of Encephalitozoon cuniculi and conserved in several human-infecting microsporidian species. Curr Genet 51: 171–186.

67. FankhauserN, Nguyen-HaTM, AdlerJ, MaserP (2007) Surface antigens and potential virulence factors from parasites detected by comparative genomics of perfect amino acid repeats. Proteome Sci 5: 20.

68. SchornackS, van DammeM, BozkurtTO, CanoLM, SmokerM, et al. (2010) Ancient class of translocated oomycete effectors targets the host nucleus. Proc Natl Acad Sci U S A 107: 17421–17426.

69. Sambrook J, Fritsch EF, Maniatis T (1989) Molecular cloning : a laboratory manual. Cold Spring Harbor, N.Y.: Cold Spring Harbor Laboratory.

70. SchattnerP, BrooksAN, LoweTM (2005) The tRNAscan-SE, snoscan and snoGPS web servers for the detection of tRNAs and snoRNAs. Nucleic Acids Res 33: W686–689.

71. Aronesty E (2011) Command-line tools for processing biological sequencing data. ea-utils http://code.google.com/p/ea-utils: Expression Analysis, Durham, NC

72. LangmeadB, TrapnellC, PopM, SalzbergSL (2009) Ultrafast and memory-efficient alignment of short DNA sequences to the human genome. Genome Biol 10: R25.

Štítky
Genetika Reprodukčná medicína

Článok vyšiel v časopise

PLOS Genetics


2013 Číslo 8
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#