#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

A Rho GDP Dissociation Inhibitor Produced by Apoptotic T-Cells Inhibits Growth of


Most people who are infected with Mycobacterium tuberculosis (M. tb) have latent tuberculosis infection (LTBI) with protective immunity. Patients with active tuberculosis have severe disease and ineffective immunity. Understanding how LTBI individuals control infection without developing disease provides important insight into the mechanisms of protective immunity against tuberculosis, and this information is essential for development of an effective vaccine. It is known that a lymphocyte population called T-cells contributes significantly to protective immunity against tuberculosis infection. In the current study, using human and murine models of M. tb infection, we found that a soluble factor, Rho GDP dissociation inhibitor (D4GDI), produced by a subpopulation of T-cells (CD4+CD25+Foxp3+) inhibits M. tb growth. We also found that D4GDI induces M. tb genes that are expressed during the non-replicative state. Our results suggest that D4GDI has a previously undescribed positive effect on immunity by enhancing host antimicrobial activity. These findings also may aid in understanding the factors that induce LTBI. Further, this information will facilitate development of improved vaccines and immunotherapeutic strategies to prevent and treat tuberculosis, respectively.


Vyšlo v časopise: A Rho GDP Dissociation Inhibitor Produced by Apoptotic T-Cells Inhibits Growth of. PLoS Pathog 11(2): e32767. doi:10.1371/journal.ppat.1004617
Kategorie: Research Article
prolekare.web.journal.doi_sk: https://doi.org/10.1371/journal.ppat.1004617

Souhrn

Most people who are infected with Mycobacterium tuberculosis (M. tb) have latent tuberculosis infection (LTBI) with protective immunity. Patients with active tuberculosis have severe disease and ineffective immunity. Understanding how LTBI individuals control infection without developing disease provides important insight into the mechanisms of protective immunity against tuberculosis, and this information is essential for development of an effective vaccine. It is known that a lymphocyte population called T-cells contributes significantly to protective immunity against tuberculosis infection. In the current study, using human and murine models of M. tb infection, we found that a soluble factor, Rho GDP dissociation inhibitor (D4GDI), produced by a subpopulation of T-cells (CD4+CD25+Foxp3+) inhibits M. tb growth. We also found that D4GDI induces M. tb genes that are expressed during the non-replicative state. Our results suggest that D4GDI has a previously undescribed positive effect on immunity by enhancing host antimicrobial activity. These findings also may aid in understanding the factors that induce LTBI. Further, this information will facilitate development of improved vaccines and immunotherapeutic strategies to prevent and treat tuberculosis, respectively.


Zdroje

1. Flynn JL, Chan J (2001) Immunology of tuberculosis. Annu Rev Immunol 19: 93–129. doi: 10.1146/annurev.immunol.19.1.93 11244032

2. Cooper AM, Dalton DK, Stewart TA, Griffin JP, Russell DG, et al. (1993) Disseminated tuberculosis in interferon gamma gene-disrupted mice. J Exp Med 178: 2243–2247. doi: 10.1084/jem.178.6.2243 8245795

3. Hori S, Nomura T, Sakaguchi S (2003) Control of regulatory T cell development by the transcription factor Foxp3. Science 299: 1057–1061. doi: 10.1126/science.1079490 12522256

4. Scott-Browne JP, Shafiani S, Tucker-Heard G, Ishida-Tsubota K, Fontenot JD, et al. (2007) Expansion and function of Foxp3-expressing T regulatory cells during tuberculosis. J Exp Med 204: 2159–2169. doi: 10.1084/jem.20062105 17709423

5. Kursar M, Koch M, Mittrucker HW, Nouailles G, Bonhagen K, et al. (2007) Cutting Edge: Regulatory T cells prevent efficient clearance of Mycobacterium tuberculosis. J Immunol 178: 2661–2665. doi: 10.4049/jimmunol.178.5.2661 17312107

6. Garg A, Barnes PF, Roy S, Quiroga MF, Wu S, et al. (2008) Mannose-capped lipoarabinomannan- and prostaglandin E2-dependent expansion of regulatory T cells in human Mycobacterium tuberculosis infection. Eur J Immunol 38: 459–469. doi: 10.1002/eji.200737268 18203140

7. Periasamy S, Dhiman R, Barnes PF, Paidipally P, Tvinnereim A, et al. (2011) Programmed death 1 and cytokine inducible SH2-containing protein dependent expansion of regulatory T cells upon stimulation with Mycobacterium tuberculosis. J Infect Dis 203: 1256–1263. doi: 10.1093/infdis/jir011 21383382

8. Hirsch CS, Toossi Z, Othieno C, Johnson JL, Schwander SK, et al. (1999) Depressed T-cell interferon-gamma responses in pulmonary tuberculosis: analysis of underlying mechanisms and modulation with therapy. J Infect Dis 180: 2069–2073. doi: 10.1086/315114 10558973

9. Mahan CS, Thomas JJ, Boom WH, Rojas RE (2009) CD4+CD25(high) Foxp3+ regulatory T cells downregulate human Vdelta2+ T-lymphocyte function triggered by anti-CD3 or phosphoantigen. Immunology 127: 398–407. doi: 10.1111/j.1365-2567.2008.02982.x 19019089

10. Morgan ME, van Bilsen JH, Bakker AM, Heemskerk B, Schilham MW, et al. (2005) Expression of FOXP3 mRNA is not confined to CD4+CD25+ T regulatory cells in humans. Hum Immunol 66: 13–20. doi: 10.1016/j.humimm.2004.05.016 15620457

11. Wang J, Ioan-Facsinay A, van der Voort EI, Huizinga TW, Toes RE (2007) Transient expression of FOXP3 in human activated nonregulatory CD4+ T cells. Eur J Immunol 37: 129–138. doi: 10.1002/eji.200636435 17154262

12. Roncarolo MG, Gregori S (2008) Is FOXP3 a bona fide marker for human regulatory T cells?. Eur J Immunol 38: 925–927. doi: 10.1002/eji.200838168 18395862

13. Canaday DH, Wilkinson RJ, Li Q, Harding CV, Silver RF, et al. (2001) CD4(+) and CD8(+) T cells kill intracellular Mycobacterium tuberculosis by a perforin and Fas/Fas ligand-independent mechanism. J Immunol 167: 2734–2742. doi: 10.4049/jimmunol.167.5.2734 11509617

14. Silver RF, Li Q, Boom WH, Ellner JJ (1998) Lymphocyte-dependent inhibition of growth of virulent Mycobacterium tuberculosis H37Rv within human monocytes: requirement for CD4+ T cells in purified protein derivative-positive, but not in purified protein derivative-negative subjects. J Immunol 160: 2408–2417. 9498784

15. Dovas A, Couchman JR (2005) RhoGDI: multiple functions in the regulation of Rho family GTPase activities. Biochem J 390: 1–9. doi: 10.1042/BJ20050104 16083425

16. Guillemot JC, Kruskal BA, Adra CN, Zhu S, Ko JL, et al. (1996) Targeted disruption of guanosine diphosphate-dissociation inhibitor for Rho-related proteins, GDID4: normal hematopoietic differentiation but subtle defect in superoxide production by macrophages derived from in vitro embryonal stem cell differentiation. Blood 88: 2722–2731. 8839868

17. Honaker RW, Leistikow RL, Bartek IL, Voskuil MI (2009) Unique roles of DosT and DosS in DosR regulon induction and Mycobacterium tuberculosis dormancy. Infect Immun 77: 3258–3263. doi: 10.1128/IAI.01449-08 19487478

18. Rustad TR, Sherrid AM, Minch KJ, Sherman DR (2009) Hypoxia: a window into Mycobacterium tuberculosis latency. Cell Microbiol 11: 1151–1159. doi: 10.1111/j.1462-5822.2009.01325.x 19388905

19. Converse PJ, Karakousis PC, Klinkenberg LG, Kesavan AK, Ly LH, et al. (2009) Role of the dosR-dosS two-component regulatory system in Mycobacterium tuberculosis virulence in three animal models. Infect Immun 77: 1230–1237. doi: 10.1128/IAI.01117-08 19103767

20. Leistikow RL, Morton RA, Bartek IL, Frimpong I, Wagner K, et al. (2010) The Mycobacterium tuberculosis DosR regulon assists in metabolic homeostasis and enables rapid recovery from nonrespiring dormancy. J Bacteriol 192: 1662–1670. doi: 10.1128/JB.00926-09 20023019

21. Kendall SL, Movahedzadeh F, Rison SC, Wernisch L, Parish T, et al. (2004) The Mycobacterium tuberculosis dosRS two-component system is induced by multiple stresses. Tuberculosis (Edinb) 84: 247–255. doi: 10.1016/j.tube.2003.12.007

22. Barnes PF, Mistry SD, Cooper CL, Pirmez C, Rea TH, et al. (1989) Compartmentalization of a CD4+ T lymphocyte subpopulation in tuberculous pleuritis. J Immunol 142: 1114–1119. 2464638

23. Barnes PF, Fong SJ, Brennan PJ, Twomey PE, Mazumder A, et al. (1990) Local production of tumor necrosis factor and IFN-gamma in tuberculous pleuritis. J Immunol 145: 149–154. 2113553

24. Belkaid Y, Piccirillo CA, Mendez S, Shevach EM, Sacks DL (2002) CD4+CD25+ regulatory T cells control Leishmania major persistence and immunity. Nature 420: 502–507. doi: 10.1038/nature01152 12466842

25. Aseffa A, Gumy A, Launois P, MacDonald HR, Louis JA, et al. (2002) The early IL-4 response to Leishmania major and the resulting Th2 cell maturation steering progressive disease in BALB/c mice are subject to the control of regulatory CD4+CD25+ T cells. J Immunol 169: 3232–3241. doi: 10.4049/jimmunol.169.6.3232 12218142

26. Walther M, Tongren JE, Andrews L, Korbel D, King E, et al. (2005) Upregulation of TGF-beta, FOXP3, and CD4+CD25+ regulatory T cells correlates with more rapid parasite growth in human malaria infection. Immunity 23: 287–296. doi: 10.1016/j.immuni.2005.08.006 16169501

27. Hisaeda H, Maekawa Y, Iwakawa D, Okada H, Himeno K, et al. (2004) Escape of malaria parasites from host immunity requires CD4+CD25+ regulatory T cells. Nat Med 10: 29–30. doi: 10.1038/nm975 14702631

28. Lundgren A, Suri-Payer E, Enarsson K, Svennerholm AM, Lundin BS (2003) Helicobacter pylori-specific CD4+ CD25high regulatory T cells suppress memory T-cell responses to H. pylori in infected individuals. Infect Immun 71: 1755–1762. doi: 10.1128/IAI.71.4.1755-1762.2003

29. Harris PR, Wright SW, Serrano C, Riera F, Duarte I, et al. (2008) Helicobacter pylori gastritis in children is associated with a regulatory T-cell response. Gastroenterology 134: 491–499. doi: 10.1053/j.gastro.2007.11.006 18242215

30. Kinter AL, Hennessey M, Bell A, Kern S, Lin Y, et al. (2004) CD25(+)CD4(+) regulatory T cells from the peripheral blood of asymptomatic HIV-infected individuals regulate CD4(+) and CD8(+) HIV-specific T cell immune responses in vitro and are associated with favorable clinical markers of disease status. J Exp Med 200: 331–343. doi: 10.1084/jem.20032069 15280419

31. Aandahl EM, Michaelsson J, Moretto WJ, Hecht FM, Nixon DF (2004) Human CD4+ CD25+ regulatory T cells control T-cell responses to human immunodeficiency virus and cytomegalovirus antigens. J Virol 78: 2454–2459. doi: 10.1128/JVI.78.5.2454-2459.2004 14963140

32. Baumforth KR, Birgersdotter A, Reynolds GM, Wei W, Kapatai G, et al. (2008) Expression of the Epstein-Barr virus-encoded Epstein-Barr virus nuclear antigen 1 in Hodgkin’s lymphoma cells mediates Up-regulation of CCL20 and the migration of regulatory T cells. Am J Pathol 173: 195–204. doi: 10.2353/ajpath.2008.070845 18502823

33. Cabrera R, Tu Z, Xu Y, Firpi RJ, Rosen HR, et al. (2004) An immunomodulatory role for CD4(+)CD25(+) regulatory T lymphocytes in hepatitis C virus infection. Hepatology 40: 1062–1071. doi: 10.1002/hep.20454 15486925

34. Stoop JN, van der Molen RG, Baan CC, van der Laan LJ, Kuipers EJ, et al. (2005) Regulatory T cells contribute to the impaired immune response in patients with chronic hepatitis B virus infection. Hepatology 41: 771–778. doi: 10.1002/hep.20649 15791617

35. Peng G, Li S, Wu W, Sun Z, Chen Y, et al. (2008) Circulating CD4+ CD25+ regulatory T cells correlate with chronic hepatitis B infection. Immunology 123: 57–65. doi: 10.1111/j.1365-2567.2007.02691.x 17764450

36. Pandiyan P, Conti HR, Zheng L, Peterson AC, Mathern DR, et al. (2011) CD4(+)CD25(+)Foxp3(+) regulatory T cells promote Th17 cells in vitro and enhance host resistance in mouse Candida albicans Th17 cell infection model. Immunity 34: 422–434. doi: 10.1016/j.immuni.2011.03.002 21435589

37. Garcia-Mata R, Boulter E, Burridge K (2011) The ‘invisible hand’: regulation of RHO GTPases by RHOGDIs. Nat Rev Mol Cell Biol 12: 493–504. doi: 10.1038/nrm3153 21779026

38. Etienne-Manneville S, Hall A (2002) Rho GTPases in cell biology. Nature 420: 629–635. doi: 10.1038/nature01148 12478284

39. Watanabe T, Urano E, Miyauchi K, Ichikawa R, Hamatake M, et al. (2012) The Hematopoietic Cell-Specific Rho GTPase Inhibitor ARHGDIB/D4GDI Limits HIV Type 1 Replication. AIDS Res Hum Retroviruses 28: 912–922. doi: 10.1089/aid.2011.0180

40. Juffermans NP, Florquin S, Camoglio L, Verbon A, Kolk AH, et al. (2000) Interleukin-1 signaling is essential for host defense during murine pulmonary tuberculosis. J Infect Dis 182: 902–908. doi: 10.1086/315771 10950787

41. Mayer-Barber KD, Barber DL, Shenderov K, White SD, Wilson MS, et al. (2010) Caspase-1 independent IL-1beta production is critical for host resistance to Mycobacterium tuberculosis and does not require TLR signaling in vivo. J Immunol 184: 3326–3330. doi: 10.4049/jimmunol.0904189 20200276

42. Jayaraman P, Sada-Ovalle I, Nishimura T, Anderson AC, Kuchroo VK, et al. (2013) IL-1beta promotes antimicrobial immunity in macrophages by regulating TNFR signaling and caspase-3 activation. J Immunol 190: 4196–4204. doi: 10.4049/jimmunol.1202688 23487424

43. Kleinnijenhuis J, Joosten LA, van de Veerdonk FL, Savage N, van Crevel R, et al. (2009) Transcriptional and inflammasome-mediated pathways for the induction of IL-1beta production by Mycobacterium tuberculosis. Eur J Immunol 39: 1914–1922. doi: 10.1002/eji.200839115 19544485

44. Liu PT, Schenk M, Walker VP, Dempsey PW, Kanchanapoomi M, et al. (2009) Convergence of IL-1beta and VDR activation pathways in human TLR2/1-induced antimicrobial responses. PLoS ONE 4: e5810. doi: 10.1371/journal.pone.0005810 19503839

45. Verway M, Bouttier M, Wang TT, Carrier M, Calderon M, et al. (2013) Vitamin D induces interleukin-1beta expression: paracrine macrophage epithelial signaling controls M. tuberculosis infection. PLoS Pathog 9: e1003407. doi: 10.1371/journal.ppat.1003407

46. Flynn JL, Goldstein MM, Chan J, Triebold KJ, Pfeffer K, et al. (1995) Tumor necrosis factor-alpha is required in the protective immune response against Mycobacterium tuberculosis in mice. Immunity 2: 561–572. doi: 10.1016/1074-7613(95)90001-2 7540941

47. Rook GA, Taverne J, Leveton C, Steele J (1987) The role of gamma-interferon, vitamin D3 metabolites and tumour necrosis factor in the pathogenesis of tuberculosis. Immunology 62: 229–234. 3119471

48. Hirsch CS, Ellner JJ, Russell DG, Rich EA (1994) Complement receptor-mediated uptake and tumor necrosis factor-alpha-mediated growth inhibition of Mycobacterium tuberculosis by human alveolar macrophages. J Immunol 152: 743–753. 8283049

49. Fang FC (2004) Antimicrobial reactive oxygen and nitrogen species: concepts and controversies. Nat Rev Microbiol 2: 820–832. doi: 10.1038/nrmicro1004 15378046

50. Freeman AF, Holland SM (2007) Persistent bacterial infections and primary immune disorders. Curr Opin Microbiol 10: 70–75. doi: 10.1016/j.mib.2006.11.005 17208513

51. Babior BM (1999) NADPH oxidase: an update. Blood 93: 1464–1476. 10029572

52. Casanova JL, Abel L (2002) Genetic dissection of immunity to mycobacteria: the human model. Annu Rev Immunol 20: 581–620. doi: 10.1146/annurev.immunol.20.081501.125851 11861613

53. Wu W, Hsu YM, Bi L, Songyang Z, Lin X (2009) CARD9 facilitates microbe-elicited production of reactive oxygen species by regulating the LyGDI-Rac1 complex. Nat Immunol 10: 1208–1214. doi: 10.1038/ni.1788 19767757

54. Miller JL, Velmurugan K, Cowan MJ, Briken V (2010) The type I NADH dehydrogenase of Mycobacterium tuberculosis counters phagosomal NOX2 activity to inhibit TNF-alpha-mediated host cell apoptosis. PLoS Pathog 6: e1000864. doi: 10.1371/journal.ppat.1000864 20421951

55. Yang CS, Shin DM, Kim KH, Lee ZW, Lee CH, et al. (2009) NADPH oxidase 2 interaction with TLR2 is required for efficient innate immune responses to mycobacteria via cathelicidin expression. J Immunol 182: 3696–3705. doi: 10.4049/jimmunol.0802217 19265148

56. Deretic V (2012) Autophagy as an innate immunity paradigm: expanding the scope and repertoire of pattern recognition receptors. Curr Opin Immunol 24: 21–31. doi: 10.1016/j.coi.2011.10.006 22118953

57. Brightbill HD, Libraty DH, Krutzik SR, Yang RB, Belisle JT, et al. (1999) Host defense mechanisms triggered by microbial lipoproteins through toll-like receptors. Science 285: 732–736. doi: 10.1126/science.285.5428.732 10426995

58. Behar SM, Martin CJ, Nunes-Alves C, Divangahi M, Remold HG (2011) Lipids, apoptosis, and cross-presentation: links in the chain of host defense against Mycobacterium tuberculosis. Microbes Infect 13: 749–756. doi: 10.1016/j.micinf.2011.03.002 21458584

59. Keane J, Remold HG, Kornfeld H (2000) Virulent Mycobacterium tuberculosis strains evade apoptosis of infected alveolar macrophages. J Immunol 164: 2016–2020. doi: 10.4049/jimmunol.164.4.2016 10657653

60. Sly LM, Hingley-Wilson SM, Reiner NE, McMaster WR (2003) Survival of Mycobacterium tuberculosis in host macrophages involves resistance to apoptosis dependent upon induction of antiapoptotic Bcl-2 family member Mcl-1. J Immunol 170: 430–437. doi: 10.4049/jimmunol.170.1.430 12496428

61. Velmurugan K, Chen B, Miller JL, Azogue S, Gurses S, et al. (2007) Mycobacterium tuberculosis nuoG is a virulence gene that inhibits apoptosis of infected host cells. PLoS Pathog 3: e110. doi: 10.1371/journal.ppat.0030110 17658950

62. Kundu M, Pathak SK, Kumawat K, Basu S, Chatterjee G, et al. (2009) A TNF- and c-Cbl-dependent FLIP(S)-degradation pathway and its function in Mycobacterium tuberculosis-induced macrophage apoptosis. Nat Immunol 10: 918–926. doi: 10.1038/ni.1754 19597496

63. Shafiani S, Tucker-Heard G, Kariyone A, Takatsu K, Urdahl KB (2010) Pathogen-specific regulatory T cells delay the arrival of effector T cells in the lung during early tuberculosis. J Exp Med 207: 1409–1420. doi: 10.1084/jem.20091885 20547826

64. Green AM, Mattila JT, Bigbee CL, Bongers KS, Lin PL, et al. (2010) CD4(+) regulatory T cells in a cynomolgus macaque model of Mycobacterium tuberculosis infection. J Infect Dis 202: 533–541. doi: 10.1086/654896 20617900

65. Guyot-Revol V, Innes JA, Hackforth S, Hinks T, Lalvani A (2006) Regulatory T cells are expanded in blood and disease sites in patients with tuberculosis. Am J Respir Crit Care Med 173: 803–810. doi: 10.1164/rccm.200508-1294OC 16339919

66. Rahman S, Gudetta B, Fink J, Granath A, Ashenafi S, et al. (2009) Compartmentalization of immune responses in human tuberculosis: few CD8+ effector T cells but elevated levels of FoxP3+ regulatory t cells in the granulomatous lesions. Am J Pathol 174: 2211–2224. doi: 10.2353/ajpath.2009.080941 19435796

67. Sharma PK, Saha PK, Singh A, Sharma SK, Ghosh B, et al. (2009) FoxP3+ regulatory T cells suppress effector T-cell function at pathologic site in miliary tuberculosis. Am J Respir Crit Care Med 179: 1061–1070. doi: 10.1164/rccm.200804-529OC 19246720

68. Park HD, Guinn KM, Harrell MI, Liao R, Voskuil MI, et al. (2003) Rv3133c/dosR is a transcription factor that mediates the hypoxic response of Mycobacterium tuberculosis. Mol Microbiol 48: 833–843. doi: 10.1046/j.1365-2958.2003.03474.x 12694625

69. Karakousis PC, Yoshimatsu T, Lamichhane G, Woolwine SC, Nuermberger EL, et al. (2004) Dormancy phenotype displayed by extracellular Mycobacterium tuberculosis within artificial granulomas in mice. J Exp Med 200: 647–657. doi: 10.1084/jem.20040646 15353557

70. Shiloh MU, Manzanillo P, Cox JS (2008) Mycobacterium tuberculosis senses host-derived carbon monoxide during macrophage infection. Cell Host Microbe 3: 323–330. doi: 10.1016/j.chom.2008.03.007 18474359

71. Tan T, Lee WL, Alexander DC, Grinstein S, Liu J (2006) The ESAT-6/CFP-10 secretion system of Mycobacterium tuberculosis modulates phagosome maturation. Cell Microbiol 8: 1417–1429. doi: 10.1111/j.1462-5822.2006.00721.x 16922861

72. Dhiman R, Periasamy S, Barnes PF, Jaiswal AG, Paidipally P, et al. (2012) NK1.1+ Cells and IL-22 Regulate Vaccine-Induced Protective Immunity against Challenge with Mycobacterium tuberculosis. J Immunol 189: 897–905. doi: 10.4049/jimmunol.1102833 22711885

73. Junqueira-Kipnis AP, Kipnis A, Jamieson A, Juarrero MG, Diefenbach A, et al. (2003) NK cells respond to pulmonary infection with Mycobacterium tuberculosis, but play a minimal role in protection. J Immunol 171: 6039–6045. doi: 10.4049/jimmunol.171.11.6039 14634116

Štítky
Hygiena a epidemiológia Infekčné lekárstvo Laboratórium

Článok vyšiel v časopise

PLOS Pathogens


2015 Číslo 2
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#