#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

Dual-Site Phosphorylation of the Control of Virulence Regulator Impacts Group A Streptococcal Global Gene Expression and Pathogenesis


Group A Streptococcus (GAS) causes a variety of human diseases ranging from mild throat infections to deadly invasive infections. The capacity of GAS to cause infections at such diverse locations is dependent on its ability to precisely control the production of a broad variety of virulence factors. The control of virulence regulator (CovR) is a master regulator of GAS genes encoding virulence factors. It is known that CovR can be phosphorylated on aspartate-53 in vitro and that such phosphorylation increases its regulatory activity, but what additional factors influence CovR-mediated gene expression have not been established. Herein we show for the first time that CovR is phosphorylated in vivo and that phosphorylation of CovR on threonine-65 by the threonine/serine kinase Stk prevents aspartate-53 phosphorylation, thereby decreasing CovR regulatory activity. Further, while CovR-mediated gene repression is highly dependent on aspartate-53 phosphorylation, CovR-mediated gene activation proceeds via a phosphorylation-independent mechanism. Modifications in CovR phosphorylation sites significantly affected the expression of GAS virulence factors during infection and markedly altered the ability of GAS to cause disease in mice. These data establish that multiple inter-related pathways converge to influence CovR phosphorylation, thereby providing new insight into the complex regulatory network used by GAS during infection.


Vyšlo v časopise: Dual-Site Phosphorylation of the Control of Virulence Regulator Impacts Group A Streptococcal Global Gene Expression and Pathogenesis. PLoS Pathog 10(5): e32767. doi:10.1371/journal.ppat.1004088
Kategorie: Research Article
prolekare.web.journal.doi_sk: https://doi.org/10.1371/journal.ppat.1004088

Souhrn

Group A Streptococcus (GAS) causes a variety of human diseases ranging from mild throat infections to deadly invasive infections. The capacity of GAS to cause infections at such diverse locations is dependent on its ability to precisely control the production of a broad variety of virulence factors. The control of virulence regulator (CovR) is a master regulator of GAS genes encoding virulence factors. It is known that CovR can be phosphorylated on aspartate-53 in vitro and that such phosphorylation increases its regulatory activity, but what additional factors influence CovR-mediated gene expression have not been established. Herein we show for the first time that CovR is phosphorylated in vivo and that phosphorylation of CovR on threonine-65 by the threonine/serine kinase Stk prevents aspartate-53 phosphorylation, thereby decreasing CovR regulatory activity. Further, while CovR-mediated gene repression is highly dependent on aspartate-53 phosphorylation, CovR-mediated gene activation proceeds via a phosphorylation-independent mechanism. Modifications in CovR phosphorylation sites significantly affected the expression of GAS virulence factors during infection and markedly altered the ability of GAS to cause disease in mice. These data establish that multiple inter-related pathways converge to influence CovR phosphorylation, thereby providing new insight into the complex regulatory network used by GAS during infection.


Zdroje

1. RecinosDA, SekedatMD, HernandezA, CohenTS, SakhtahH, et al. (2012) Redundant phenazine operons in Pseudomonas aeruginosa exhibit environment-dependent expression and differential roles in pathogenicity. Proc Natl Acad Sci U S A 109: 19420–19425.

2. BohmeK, SteinmannR, KortmannJ, SeekircherS, HerovenAK, et al. (2012) Concerted actions of a thermo-labile regulator and a unique intergenic RNA thermosensor control Yersinia virulence. PLoS Pathog 8: e1002518.

3. MandlikA, LivnyJ, RobinsWP, RitchieJM, MekalanosJJ, et al. (2011) RNA-Seq-based monitoring of infection-linked changes in Vibrio cholerae gene expression. Cell Host Microbe 10: 165–174.

4. BalasubramanianD, SchneperL, KumariH, MatheeK (2013) A dynamic and intricate regulatory network determines Pseudomonas aeruginosa virulence. Nucleic Acids Res 41: 1–20.

5. CaiW, WannemuehlerY, Dell'annaG, NicholsonB, BarbieriNL, et al. (2013) A novel two-component signaling system facilitates uropathogenic Escherichia coli's ability to exploit abundant host metabolites. PLoS Pathog 9: e1003428.

6. LyonGJ, MayvilleP, MuirTW, NovickRP (2000) Rational design of a global inhibitor of the virulence response in Staphylococcus aureus, based in part on localization of the site of inhibition to the receptor-histidine kinase, AgrC. Proc Natl Acad Sci U S A 97: 13330–13335.

7. ZahrtTC, DereticV (2001) Mycobacterium tuberculosis signal transduction system required for persistent infections. Proc Natl Acad Sci U S A 98: 12706–12711.

8. GaoR, StockAM (2009) Biological insights from structures of two-component proteins. Annu Rev Microbiol 63: 133–154.

9. StockAM, RobinsonVL, GoudreauPN (2000) Two-component signal transduction. Annu Rev Biochem 69: 183–215.

10. KobirA, ShiL, BoskovicA, GrangeasseC, FranjevicD, et al. (2011) Protein phosphorylation in bacterial signal transduction. Biochim Biophys Acta 1810: 989–994.

11. RaskoDA, MoreiraCG, Li deR, ReadingNC, RitchieJM, et al. (2008) Targeting QseC signaling and virulence for antibiotic development. Science 321: 1078–1080.

12. GotohY, DoiA, FurutaE, DubracS, IshizakiY, et al. (2010) Novel antibacterial compounds specifically targeting the essential WalR response regulator. J Antibiot (Tokyo) 63: 127–134.

13. GotohY, EguchiY, WatanabeT, OkamotoS, DoiA, et al. (2010) Two-component signal transduction as potential drug targets in pathogenic bacteria. Curr Opin Microbiol 13: 232–239.

14. TangYT, GaoR, HavranekJJ, GroismanEA, StockAM, et al. (2012) Inhibition of bacterial virulence: drug-like molecules targeting the Salmonella enterica PhoP response regulator. Chem Biol Drug Des 79: 1007–1017.

15. LevinJC, WesselsMR (1998) Identification of csrR/csrS, a genetic locus that regulates hyaluronic acid capsule synthesis in group A Streptococcus. Mol Microbiol 30: 209–219.

16. ChurchwardG (2007) The two faces of Janus: virulence gene regulation by CovR/S in group A streptococci. Mol Microbiol 64: 34–41.

17. FederleMJ, McIverKS, ScottJR (1999) A response regulator that represses transcription of several virulence operons in the group A Streptococcus. J Bacteriol 181: 3649–3657.

18. CarapetisJR, SteerAC, MulhollandEK, WeberM (2005) The global burden of group A streptococcal diseases. Lancet Infect Dis 5: 685–694.

19. EnglebergNC, HeathA, MillerA, RiveraC, DiRitaVJ (2001) Spontaneous mutations in the CsrRS two-component regulatory system of Streptococcus pyogenes result in enhanced virulence in a murine model of skin and soft tissue infection. J Infect Dis 183: 1043–1054.

20. GrahamMR, SmootLM, MigliaccioCA, VirtanevaK, SturdevantDE, et al. (2002) Virulence control in group A Streptococcus by a two-component gene regulatory system: global expression profiling and in vivo infection modeling. Proc Natl Acad Sci USA 99: 13855–13860.

21. DaltonTL, ScottJR (2004) CovS inactivates CovR and is required for growth under conditions of general stress in Streptococcus pyogenes. J Bacteriol 186: 3928–3937.

22. GusaAA, GaoJ, StringerV, ChurchwardG, ScottJR (2006) Phosphorylation of the group A streptococcal CovR response regulator causes dimerization and promoter-specific recruitment by RNA polymerase. J Bacteriol 188: 4620–4626.

23. BarbieriCM, WuT, StockAM (2013) Comprehensive analysis of OmpR phosphorylation, dimerization, and DNA binding supports a canonical model for activation. J Mol Biol 425: 1612–1626.

24. DaltonTL, HobbRI, ScottJR (2006) Analysis of the role of CovR and CovS in the dissemination of Streptococcus pyogenes in invasive skin disease. Microb Pathog 40: 221–227.

25. TrevinoJ, PerezN, Ramirez-PenaE, LiuZ, ShelburneSA3rd, et al. (2009) CovS simultaneously activates and inhibits the CovR-mediated repression of distinct subsets of group A Streptococcus virulence factor-encoding genes. Infect Immun 77: 3141–3149.

26. AgarwalS, PancholiP, PancholiV (2011) Role of serine/threonine phosphatase (SP-STP) in Streptococcus pyogenes physiology and virulence. J Biol Chem 286: 41368–41380.

27. RajagopalL, ClancyA, RubensCE (2003) A eukaryotic type serine/threonine kinase and phosphatase in Streptococcus agalactiae reversibly phosphorylate an inorganic pyrophosphatase and affect growth, cell segregation, and virulence. J Biol Chem 278: 14429–14441.

28. RajagopalL, VoA, SilvestroniA, RubensCE (2006) Regulation of cytotoxin expression by converging eukaryotic-type and two-component signalling mechanisms in Streptococcus agalactiae. Mol Microbiol 62: 941–957.

29. LinWJ, WalthersD, ConnellyJE, BurnsideK, JewellKA, et al. (2009) Threonine phosphorylation prevents promoter DNA binding of the Group B Streptococcus response regulator CovR. Mol Microbiol 71: 1477–1495.

30. LemboA, GurneyMA, BurnsideK, BanerjeeA, de los ReyesM, et al. (2010) Regulation of CovR expression in Group B Streptococcus impacts blood-brain barrier penetration. Mol Microbiol 77: 431–443.

31. WhidbeyC, HarrellMI, BurnsideK, NgoL, BecraftAK, et al. (2013) A hemolytic pigment of Group B Streptococcus allows bacterial penetration of human placenta. J Exp Med 210: 1265–1281.

32. MillerAA, EnglebergNC, DiRitaVJ (2001) Repression of virulence genes by phosphorylation-dependent oligomerization of CsrR at target promoters in Streptococcus pyogenes. Mol Microbiol 40: 976–990.

33. FederleMJ, ScottJR (2002) Identification of binding sites for the group A streptococcal global regulator CovR. Mol Microbiol 43: 1161–1172.

34. JiangSM, IshmaelN, HotoppJD, PulitiM, TissiL, et al. (2008) Variation in the group B Streptococcus CsrRS regulon and effects on pathogenicity. J Bacteriol 190: 1956–1965.

35. BarbieriCM, StockAM (2008) Universally applicable methods for monitoring response regulator aspartate phosphorylation both in vitro and in vivo using Phos-tag-based reagents. Anal Biochem 376: 73–82.

36. WayneKJ, LiS, KazmierczakKM, TsuiHC, WinklerME (2012) Involvement of WalK (VicK) phosphatase activity in setting WalR (VicR) response regulator phosphorylation level and limiting cross-talk in Streptococcus pneumoniae D39 cells. Mol Microbiol 86: 645–660.

37. BoulangerA, ChenQ, HintonDM, StibitzS (2013) In vivo phosphorylation dynamics of the Bordetella pertussis virulence-controlling response regulator BvgA. Mol Microbiol 88: 156–172.

38. KinoshitaE, Kinoshita-KikutaE (2011) Improved Phos-tag SDS-PAGE under neutral pH conditions for advanced protein phosphorylation profiling. Proteomics 11: 319–323.

39. ShelburneSA, OlsenRJ, SuberB, SahasrabhojaneP, SumbyP, et al. (2010) A combination of independent transcriptional regulators shapes bacterial virulence gene expression during infection. PLoS Pathog 6: e1000817.

40. BeresSB, CarrollRK, SheaPR, SitkiewiczI, Martinez-GutierrezJC, et al. (2010) Molecular complexity of successive bacterial epidemics deconvoluted by comparative pathogenomics. Proc Natl Acad Sci U S A 107: 4371–4376.

41. GryllosI, GrifantiniR, ColapricoA, JiangS, DeforceE, et al. (2007) Mg(2+) signalling defines the group A streptococcal CsrRS (CovRS) regulon. Mol Microbiol 65: 671–683.

42. JinH, PancholiV (2006) Identification and biochemical characterization of a eukaryotic-type serine/threonine kinase and its cognate phosphatase in Streptococcus pyogenes: their biological functions and substrate identification. J Mol Biol 357: 1351–1372.

43. HorstmannN, SahasrabhojaneP, SuberB, KumaraswamiM, OlsenRJ, et al. (2011) Distinct single amino acid replacements in the control of virulence regulator protein differentially impact streptococcal pathogenesis. PLoS Pathog 7: e1002311.

44. KansalRG, DattaV, AzizRK, AbdeltawabNF, RoweS, et al. (2010) Dissection of the molecular basis for hypervirulence of an in vivo-selected phenotype of the widely disseminated M1T1 strain of group A Streptococcus bacteria. J Infect Dis 201: 855–865.

45. AshbaughCD, AlbertiS, WesselsMR (1998) Molecular analysis of the capsule gene region of group A Streptococcus: the hasAB genes are sufficient for capsule expression. J Bacteriol 180: 4955–4959.

46. SumbyP, ZhangS, WhitneyAR, FalugiF, GrandiG, et al. (2008) A chemokine-degrading extracellular protease made by group A Streptococcus alters pathogenesis by enhancing evasion of the innate immune response. Infect Immun 76: 978–985.

47. RasmussenM, MullerHP, BjorckL (1999) Protein GRAB of Streptococcus pyogenes regulates proteolysis at the bacterial surface by binding alpha2-macroglobulin. J Biol Chem 274: 15336–15344.

48. GusaAA, FroehlichBJ, DesaiD, StringerV, ScottJR (2007) CovR activation of the dipeptide permease promoter (PdppA) in Group A Streptococcus. J Bacteriol 189: 1407–1416.

49. LiuM, ZhuH, ZhangJ, LeiB (2007) Active and passive immunizations with the streptococcal esterase Sse protect mice against subcutaneous infection with group A streptococci. Infect Immun 75: 3651–3657.

50. KapurV, KanjilalS, HamrickMR, LiLL, WhittamTS, et al. (1995) Molecular population genetic analysis of the streptokinase gene of Streptococcus pyogenes: mosaic alleles generated by recombination. Mol Microbiol 16: 509–519.

51. von Pawel-RammingenU, JohanssonBP, BjorckL (2002) IdeS, a novel streptococcal cysteine proteinase with unique specificity for immunoglobulin G. EMBO J 21: 1607–1615.

52. NizetV, BeallB, BastDJ, DattaV, KilburnL, et al. (2000) Genetic locus for streptolysin S production by group A Streptococcus. Infect Immun 68: 4245–4254.

53. SriskandanS, UnnikrishnanM, KrauszT, CohenJ (2000) Mitogenic factor (MF) is the major DNase of serotype M89 Streptococcus pyogenes. Microbiology 146(Pt 11): 2785–2792.

54. LyonWR, GibsonCM, CaparonMG (1998) A role for trigger factor and an Rgg-like regulator in the transcription, secretion and processing of the cysteine proteinase of Streptococcus pyogenes. EMBO J 17: 6263–6275.

55. Tran-WinklerHJ, LoveJF, GryllosI, WesselsMR (2011) Signal transduction through CsrRS confers an invasive phenotype in group A Streptococcus. PLoS Pathog 7: e1002361.

56. GaoJ, GusaAA, ScottJR, ChurchwardG (2005) Binding of the global response regulator protein CovR to the sag promoter of Streptococcus pyogenes reveals a new mode of CovR-DNA interaction. J Biol Chem 280: 38948–38956.

57. GusaAA, ScottJR (2005) The CovR response regulator of group A Streptococcus (GAS) acts directly to repress its own promoter. Mol Microbiol 56: 1195–1207.

58. ChaoJD, PapavinasasundaramKG, ZhengX, Chavez-SteenbockA, WangX, et al. (2010) Convergence of Ser/Thr and two-component signaling to coordinate expression of the dormancy regulon in Mycobacterium tuberculosis. J Biol Chem 285: 29239–29246.

59. XuH, CaimanoMJ, LinT, HeM, RadolfJD, et al. (2010) Role of acetyl-phosphate in activation of the Rrp2-RpoN-RpoS pathway in Borrelia burgdorferi. PLoS Pathog 6: e1001104.

60. Chiang-NiC, TsouCC, LinYS, ChuangWJ, LinMT, et al. (2008) The transcriptional terminator sequences downstream of the covR gene terminate covR/S operon transcription to generate covR monocistronic transcripts in Streptococcus pyogenes. Gene 427: 99–103.

61. CousinC, DerouicheA, ShiL, PagotY, PoncetS, et al. (2013) Protein-serine/threonine/tyrosine kinases in bacterial signaling and regulation. FEMS Microbiol Lett 346: 11–19.

62. PancholiV, BoelG, JinH (2010) Streptococcus pyogenes Ser/Thr kinase-regulated cell wall hydrolase is a cell division plane-recognizing and chain-forming virulence factor. J Biol Chem 285: 30861–30874.

63. BeilharzK, NovakovaL, FaddaD, BrannyP, MassiddaO, et al. (2012) Control of cell division in Streptococcus pneumoniae by the conserved Ser/Thr protein kinase StkP. Proc Natl Acad Sci U S A 109: E905–913.

64. BugryshevaJ, FroehlichBJ, FreibergJA, ScottJR (2011) Serine/threonine protein kinase Stk is required for virulence, stress response, and penicillin tolerance in Streptococcus pyogenes. Infect Immun 79: 4201–4209.

65. LiuM, ZhuH, LiJ, GarciaCC, FengW, et al. (2012) Group A Streptococcus secreted esterase hydrolyzes platelet-activating factor to impede neutrophil recruitment and facilitate innate immune evasion. PLoS Pathog 8: e1002624.

66. IkebeT, AtoM, MatsumuraT, HasegawaH, SataT, et al. (2010) Highly frequent mutations in negative regulators of multiple virulence genes in group A streptococcal toxic shock syndrome isolates. PLoS Pathog 6: e1000832.

67. DeutscherJ, FranckeC, PostmaPW (2006) How phosphotransferase system-related protein phosphorylation regulates carbohydrate metabolism in bacteria. Microbiol Mol Biol Rev 70: 939–1031.

68. SchumacherMA, AllenGS, DielM, SeidelG, HillenW, et al. (2004) Structural basis for allosteric control of the transcription regulator CcpA by the phosphoprotein HPr-Ser46-P. Cell 118: 731–741.

69. GaoR, MackTR, StockAM (2007) Bacterial response regulators: versatile regulatory strategies from common domains. Trends Biochem Sci 32: 225–234.

70. AmesSK, FrankemaN, KenneyLJ (1999) C-terminal DNA binding stimulates N-terminal phosphorylation of the outer membrane protein regulator OmpR from Escherichia coli. Proc Natl Acad Sci U S A 96: 11792–11797.

71. BachhawatP, StockAM (2007) Crystal structures of the receiver domain of the response regulator PhoP from Escherichia coli in the absence and presence of the phosphoryl analog beryllofluoride. J Bacteriol 189: 5987–5995.

72. GryllosI, LevinJC, WesselsMR (2003) The CsrR/CsrS two-component system of group A Streptococcus responds to environmental Mg2+. Proc Natl Acad Sci USA 100: 4227–4232.

73. OlsenRJ, LauciricaDR, WatkinsME, FeskeML, Garcia-BustillosJR, et al. (2012) Polymorphisms in regulator of protease B (RopB) alter disease phenotype and strain virulence of serotype M3 group A Streptococcus. J Infect Dis 205: 1719–1729.

74. CarrollRK, IiiSA, OlsenRJ, SuberB, SahasrabhojaneP, et al. (2011) Naturally occurring single amino acid replacements in a regulatory protein alter streptococcal gene expression and virulence in mice. J Clin Invest 121: 1956–68.

75. TannerS, ShuH, FrankA, WangLC, ZandiE, et al. (2005) InsPecT: identification of posttranslationally modified peptides from tandem mass spectra. Anal Chem 77: 4626–4639.

76. LukomskiS, SreevatsanS, AmbergC, ReichardtW, WoischnikM, et al. (1997) Inactivation of Streptococcus pyogenes extracellular cysteine protease significantly decreases mouse lethality of serotype M3 and M49 strains. J Clin Invest 99: 2574–2580.

77. VirtanevaK, PorcellaSF, GrahamMR, IrelandRM, JohnsonCA, et al. (2005) Longitudinal analysis of the group A Streptococcus transcriptome in experimental pharyngitis in cynomolgus macaques. Proc Natl Acad Sci USA 102: 9014–9019.

78. AndersS, HuberW (2010) Differential expression analysis for sequence count data. Genome Biol 11: R106.

79. ReinerA, YekutieliD, BenjaminiY (2003) Identifying differentially expressed genes using false discovery rate controlling procedures. Bioinformatics 19: 368–375.

80. LiJ, KasperDL, AusubelFM, RosnerB, MichelJL (1997) Inactivation of the alpha C protein antigen gene, bca, by a novel shuttle/suicide vector results in attenuation of virulence and immunity in group B Streptococcus. Proc Natl Acad Sci U S A 94: 13251–13256.

Štítky
Hygiena a epidemiológia Infekčné lekárstvo Laboratórium

Článok vyšiel v časopise

PLOS Pathogens


2014 Číslo 5
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#