#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

IRF-5-Mediated Inflammation Limits CD8 T Cell Expansion by Inducing HIF-1α and Impairing Dendritic Cell Functions during Infection


Inflammation is essential for inducing, sustaining, and regulating CD8+ T cell responses. The transcription factor IRF-5 is mainly responsible for initiating the inflammatory response following experimental Leishmani donovani infection. IRF-5 activates several genes encoding key pro-inflammatory cytokines, such as IL-6 and TNF. In this study, we investigate the role of IRF-5-mediated inflammation in regulating antigen-specific CD8+ T cell responses during L. donovani infection. Our data demonstrate that the inflammatory response induced by IRF-5 limits the expansion CD8+ T cell. This negative effect is mediated by the induction of HIF-1α in dendritic cells. Indeed, we observed a significant increase in CD8+ T cell expansion in mice lacking HIF-1α expression in dendritic cells. Moreover, these mice had a significantly lower parasite burden in the spleen, suggesting that induction of HIF-1α may represent an immune evasive mechanism adopted by Leishmania parasites to establish persistent infections.


Vyšlo v časopise: IRF-5-Mediated Inflammation Limits CD8 T Cell Expansion by Inducing HIF-1α and Impairing Dendritic Cell Functions during Infection. PLoS Pathog 11(6): e32767. doi:10.1371/journal.ppat.1004938
Kategorie: Research Article
prolekare.web.journal.doi_sk: https://doi.org/10.1371/journal.ppat.1004938

Souhrn

Inflammation is essential for inducing, sustaining, and regulating CD8+ T cell responses. The transcription factor IRF-5 is mainly responsible for initiating the inflammatory response following experimental Leishmani donovani infection. IRF-5 activates several genes encoding key pro-inflammatory cytokines, such as IL-6 and TNF. In this study, we investigate the role of IRF-5-mediated inflammation in regulating antigen-specific CD8+ T cell responses during L. donovani infection. Our data demonstrate that the inflammatory response induced by IRF-5 limits the expansion CD8+ T cell. This negative effect is mediated by the induction of HIF-1α in dendritic cells. Indeed, we observed a significant increase in CD8+ T cell expansion in mice lacking HIF-1α expression in dendritic cells. Moreover, these mice had a significantly lower parasite burden in the spleen, suggesting that induction of HIF-1α may represent an immune evasive mechanism adopted by Leishmania parasites to establish persistent infections.


Zdroje

1. Omer FM, de Souza JB, Riley EM (2003) Differential induction of TGF-beta regulates proinflammatory cytokine production and determines the outcome of lethal and nonlethal Plasmodium yoelii infections. J Immunol 171: 5430–5436. 14607947

2. Wu Y, Wang QH, Zheng L, Feng H, Liu J, et al. (2007) Plasmodium yoelii: distinct CD4(+)CD25(+) regulatory T cell responses during the early stages of infection in susceptible and resistant mice. Exp Parasitol 115: 301–304. 17084842

3. 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. 12466842

4. Anderson CF, Mendez S, Sacks DL (2005) Nonhealing infection despite Th1 polarization produced by a strain of Leishmania major in C57BL/6 mice. J Immunol 174: 2934–2941. 15728505

5. Brooks DG, Trifilo MJ, Edelmann KH, Teyton L, McGavern DB, et al. (2006) Interleukin-10 determines viral clearance or persistence in vivo. Nat Med 12: 1301–1309. 17041596

6. Ejrnaes M, Filippi CM, Martinic MM, Ling EM, Togher LM, et al. (2006) Resolution of a chronic viral infection after interleukin-10 receptor blockade. J Exp Med 203: 2461–2472. 17030951

7. Hernandez-Pando R, Aguilar D, Hernandez ML, Orozco H, Rook G (2004) Pulmonary tuberculosis in BALB/c mice with non-functional IL-4 genes: changes in the inflammatory effects of TNF-alpha and in the regulation of fibrosis. Eur J Immunol 34: 174–183. 14971043

8. Beil WJ, Meinardus-Hager G, Neugebauer DC, Sorg C (1992) Differences in the onset of the inflammatory response to cutaneous leishmaniasis in resistant and susceptible mice. J Leukoc Biol 52: 135–142. 1506767

9. Bogdan C, Donhauser N, Doring R, Rollinghoff M, Diefenbach A, et al. (2000) Fibroblasts as host cells in latent leishmaniosis. J Exp Med 191: 2121–2130. 10859337

10. Gorak PM, Engwerda CR, Kaye PM (1998) Dendritic cells, but not macrophages, produce IL-12 immediately following Leishmania donovani infection. Eur J Immunol 28: 687–695. 9521079

11. Iezzi G, Frohlich A, Ernst B, Ampenberger F, Saeland S, et al. (2006) Lymph node resident rather than skin-derived dendritic cells initiate specific T cell responses after Leishmania major infection. J Immunol 177: 1250–1256. 16818784

12. Peters NC, Egen JG, Secundino N, Debrabant A, Kimblin N, et al. (2008) In vivo imaging reveals an essential role for neutrophils in leishmaniasis transmitted by sand flies. Science 321: 970–974. doi: 10.1126/science.1159194 18703742

13. Kaye PM, Svensson M, Ato M, Maroof A, Polley R, et al. (2004) The immunopathology of experimental visceral leishmaniasis. Immunol Rev 201: 239–253. 15361245

14. Ato M, Stager S, Engwerda CR, Kaye PM (2002) Defective CCR7 expression on dendritic cells contributes to the development of visceral leishmaniasis. Nat Immunol 3: 1185–1191. 12436111

15. Smelt SC, Engwerda CR, McCrossen M, Kaye PM (1997) Destruction of follicular dendritic cells during chronic visceral leishmaniasis. J Immunol 158: 3813–3821. 9103448

16. Engwerda CR, Ato M, Cotterell SE, Mynott TL, Tschannerl A, et al. (2002) A role for tumor necrosis factor-alpha in remodeling the splenic marginal zone during Leishmania donovani infection. Am J Pathol 161: 429–437. 12163368

17. Engwerda CR, Murphy ML, Cotterell SE, Smelt SC, Kaye PM (1998) Neutralization of IL-12 demonstrates the existence of discrete organ-specific phases in the control of Leishmania donovani. Eur J Immunol 28: 669–680. 9521077

18. Engwerda CR, Smelt SC, Kaye PM (1996) An in vivo analysis of cytokine production during Leishmania donovani infection in scid mice. Exp Parasitol 84: 195–202. 8932769

19. Paun A, Bankoti R, Joshi T, Pitha PM, Stager S (2011) Critical role of IRF-5 in the development of T helper 1 responses to Leishmania donovani infection. PLoS Pathog 7: e1001246. doi: 10.1371/journal.ppat.1001246 21253574

20. Schoenemeyer A, Barnes BJ, Mancl ME, Latz E, Goutagny N, et al. (2005) The interferon regulatory factor, IRF5, is a central mediator of toll-like receptor 7 signaling. J Biol Chem 280: 17005–17012. 15695821

21. Barnes BJ, Richards J, Mancl M, Hanash S, Beretta L, et al. (2004) Global and distinct targets of IRF-5 and IRF-7 during innate response to viral infection. J Biol Chem 279: 45194–45207. 15308637

22. Takaoka A, Yanai H, Kondo S, Duncan G, Negishi H, et al. (2005) Integral role of IRF-5 in the gene induction programme activated by Toll-like receptors. Nature 434: 243–249. 15665823

23. Honda K, Takaoka A, Taniguchi T (2006) Type I interferon [corrected] gene induction by the interferon regulatory factor family of transcription factors. Immunity 25: 349–360. 16979567

24. Honda K, Taniguchi T (2006) IRFs: master regulators of signalling by Toll-like receptors and cytosolic pattern-recognition receptors. Nat Rev Immunol 6: 644–658. 16932750

25. Joshi T, Rodriguez S, Perovic V, Cockburn IA, Stager S (2009) B7-H1 blockade increases survival of dysfunctional CD8(+) T cells and confers protection against Leishmania donovani infections. PLoS Pathog 5: e1000431. doi: 10.1371/journal.ppat.1000431 19436710

26. Pham NL, Badovinac VP, Harty JT (2011) Differential role of "Signal 3" inflammatory cytokines in regulating CD8 T cell expansion and differentiation in vivo. Front Immunol 2: 4. doi: 10.3389/fimmu.2011.00004 22566795

27. Curtsinger JM, Gerner MY, Lins DC, Mescher MF (2007) Signal 3 availability limits the CD8 T cell response to a solid tumor. J Immunol 178: 6752–6760. 17513722

28. Curtsinger JM, Mescher MF (2010) Inflammatory cytokines as a third signal for T cell activation. Curr Opin Immunol 22: 333–340. doi: 10.1016/j.coi.2010.02.013 20363604

29. Haring JS, Badovinac VP, Harty JT (2006) Inflaming the CD8+ T cell response. Immunity 25: 19–29. 16860754

30. Joshi NS, Cui W, Chandele A, Lee HK, Urso DR, et al. (2007) Inflammation directs memory precursor and short-lived effector CD8(+) T cell fates via the graded expression of T-bet transcription factor. Immunity 27: 281–295. 17723218

31. Takemoto N, Intlekofer AM, Northrup JT, Wherry EJ, Reiner SL (2006) Cutting Edge: IL-12 inversely regulates T-bet and eomesodermin expression during pathogen-induced CD8+ T cell differentiation. J Immunol 177: 7515–7519. 17114419

32. Curtsinger JM, Lins DC, Johnson CM, Mescher MF (2005) Signal 3 tolerant CD8 T cells degranulate in response to antigen but lack granzyme B to mediate cytolysis. J Immunol 175: 4392–4399. 16177080

33. Badovinac VP, Tvinnereim AR, Harty JT (2000) Regulation of antigen-specific CD8+ T cell homeostasis by perforin and interferon-gamma. Science 290: 1354–1358. 11082062

34. Curtsinger JM, Valenzuela JO, Agarwal P, Lins D, Mescher MF (2005) Type I IFNs provide a third signal to CD8 T cells to stimulate clonal expansion and differentiation. J Immunol 174: 4465–4469. 15814665

35. Stager S, Alexander J, Kirby AC, Botto M, Rooijen NV, et al. (2003) Natural antibodies and complement are endogenous adjuvants for vaccine-induced CD8+ T-cell responses. Nat Med 9: 1287–1292. 14502281

36. Kolumam GA, Thomas S, Thompson LJ, Sprent J, Murali-Krishna K (2005) Type I interferons act directly on CD8 T cells to allow clonal expansion and memory formation in response to viral infection. J Exp Med 202: 637–650. 16129706

37. Aichele P, Unsoeld H, Koschella M, Schweier O, Kalinke U, et al. (2006) CD8 T cells specific for lymphocytic choriomeningitis virus require type I IFN receptor for clonal expansion. J Immunol 176: 4525–4529. 16585541

38. Whitmire JK, Tan JT, Whitton JL (2005) Interferon-gamma acts directly on CD8+ T cells to increase their abundance during virus infection. J Exp Med 201: 1053–1059. 15809350

39. Richer MJ, Nolz JC, Harty JT (2013) Pathogen-specific inflammatory milieux tune the antigen sensitivity of CD8(+) T cells by enhancing T cell receptor signaling. Immunity 38: 140–152. doi: 10.1016/j.immuni.2012.09.017 23260194

40. Stelekati E, Shin H, Doering TA, Dolfi DV, Ziegler CG, et al. (2014) Bystander chronic infection negatively impacts development of CD8(+) T cell memory. Immunity 40: 801–813. doi: 10.1016/j.immuni.2014.04.010 24837104

41. Semenza GL (2009) Regulation of oxygen homeostasis by hypoxia-inducible factor 1. Physiology (Bethesda) 24: 97–106. doi: 10.1152/physiol.00045.2008 19364912

42. Semenza GL (2009) Regulation of cancer cell metabolism by hypoxia-inducible factor 1. Semin Cancer Biol 19: 12–16. doi: 10.1016/j.semcancer.2008.11.009 19114105

43. Semenza GL, Jiang BH, Leung SW, Passantino R, Concordet JP, et al. (1996) Hypoxia response elements in the aldolase A, enolase 1, and lactate dehydrogenase A gene promoters contain essential binding sites for hypoxia-inducible factor 1. J Biol Chem 271: 32529–32537. 8955077

44. Semenza GL (2007) Hypoxia and cancer. Cancer Metastasis Rev 26: 223–224. 17404692

45. Jung Y, Isaacs JS, Lee S, Trepel J, Liu ZG, et al. (2003) Hypoxia-inducible factor induction by tumour necrosis factor in normoxic cells requires receptor-interacting protein-dependent nuclear factor kappa B activation. Biochem J 370: 1011–1017. 12479793

46. Zhou J, Schmid T, Brune B (2003) Tumor necrosis factor-alpha causes accumulation of a ubiquitinated form of hypoxia inducible factor-1alpha through a nuclear factor-kappaB-dependent pathway. Mol Biol Cell 14: 2216–2225. 12808024

47. Spirig R, Djafarzadeh S, Regueira T, Shaw SG, von Garnier C, et al. (2010) Effects of TLR agonists on the hypoxia-regulated transcription factor HIF-1alpha and dendritic cell maturation under normoxic conditions. PLoS One 5: e0010983. doi: 10.1371/journal.pone.0010983 20539755

48. Rius J, Guma M, Schachtrup C, Akassoglou K, Zinkernagel AS, et al. (2008) NF-kappaB links innate immunity to the hypoxic response through transcriptional regulation of HIF-1alpha. Nature 453: 807–811. doi: 10.1038/nature06905 18432192

49. Tacchini L, Gammella E, De Ponti C, Recalcati S, Cairo G (2008) Role of HIF-1 and NF-kappaB transcription factors in the modulation of transferrin receptor by inflammatory and anti-inflammatory signals. J Biol Chem 283: 20674–20686. doi: 10.1074/jbc.M800365200 18519569

50. Degrossoli A, Arrais-Silva WW, Colhone MC, Gadelha FR, Joazeiro PP, et al. (2011) The influence of low oxygen on macrophage response to Leishmania infection. Scand J Immunol 74: 165–175. doi: 10.1111/j.1365-3083.2011.02566.x 21517930

51. Degrossoli A, Bosetto MC, Lima CB, Giorgio S (2007) Expression of hypoxia-inducible factor 1alpha in mononuclear phagocytes infected with Leishmania amazonensis. Immunol Lett 114: 119–125. 17983667

52. Singh AK, Mukhopadhyay C, Biswas S, Singh VK, Mukhopadhyay CK (2012) Intracellular pathogen Leishmania donovani activates hypoxia inducible factor-1 by dual mechanism for survival advantage within macrophage. PLoS One 7: e38489. doi: 10.1371/journal.pone.0038489 22701652

53. Mancino A, Schioppa T, Larghi P, Pasqualini F, Nebuloni M, et al. (2008) Divergent effects of hypoxia on dendritic cell functions. Blood 112: 3723–3734. doi: 10.1182/blood-2008-02-142091 18694997

54. Maroof A, Beattie L, Zubairi S, Svensson M, Stager S, et al. (2008) Posttranscriptional regulation of II10 gene expression allows natural killer cells to express immunoregulatory function. Immunity 29: 295–305. doi: 10.1016/j.immuni.2008.06.012 18701085

55. Ohteki T, Fukao T, Suzue K, Maki C, Ito M, et al. (1999) Interleukin 12-dependent interferon gamma production by CD8alpha+ lymphoid dendritic cells. J Exp Med 189: 1981–1986. 10377194

56. Xiao Z, Casey KA, Jameson SC, Curtsinger JM, Mescher MF (2009) Programming for CD8 T cell memory development requires IL-12 or type I IFN. J Immunol 182: 2786–2794. doi: 10.4049/jimmunol.0803484 19234173

57. Wilson DC, Grotenbreg GM, Liu K, Zhao Y, Frickel EM, et al. (2010) Differential regulation of effector- and central-memory responses to Toxoplasma gondii Infection by IL-12 revealed by tracking of Tgd057-specific CD8+ T cells. PLoS Pathog 6: e1000815. doi: 10.1371/journal.ppat.1000815 20333242

58. Rao RR, Li Q, Odunsi K, Shrikant PA (2010) The mTOR kinase determines effector versus memory CD8+ T cell fate by regulating the expression of transcription factors T-bet and Eomesodermin. Immunity 32: 67–78. doi: 10.1016/j.immuni.2009.10.010 20060330

59. Barnes BJ, Moore PA, Pitha PM (2001) Virus-specific activation of a novel interferon regulatory factor, IRF-5, results in the induction of distinct interferon alpha genes. J Biol Chem 276: 23382–23390. 11303025

60. Arango Duque G, Fukuda M, Turco SJ, Stager S, Descoteaux A (2014) Leishmania Promastigotes Induce Cytokine Secretion in Macrophages through the Degradation of Synaptotagmin XI. J Immunol 193: 2363–2372. doi: 10.4049/jimmunol.1303043 25063865

61. Ohta A, Sitkovsky M (2001) Role of G-protein-coupled adenosine receptors in downregulation of inflammation and protection from tissue damage. Nature 414: 916–920. 11780065

62. Sitkovsky M, Lukashev D (2005) Regulation of immune cells by local-tissue oxygen tension: HIF1 alpha and adenosine receptors. Nat Rev Immunol 5: 712–721. 16110315

63. Hasko G, Linden J, Cronstein B, Pacher P (2008) Adenosine receptors: therapeutic aspects for inflammatory and immune diseases. Nat Rev Drug Discov 7: 759–770. doi: 10.1038/nrd2638 18758473

64. Csoka B, Himer L, Selmeczy Z, Vizi ES, Pacher P, et al. (2008) Adenosine A2A receptor activation inhibits T helper 1 and T helper 2 cell development and effector function. Faseb J 22: 3491–3499. doi: 10.1096/fj.08-107458 18625677

65. Lukashev D, Klebanov B, Kojima H, Grinberg A, Ohta A, et al. (2006) Cutting edge: hypoxia-inducible factor 1alpha and its activation-inducible short isoform I.1 negatively regulate functions of CD4+ and CD8+ T lymphocytes. J Immunol 177: 4962–4965. 17015677

66. Thiel M, Caldwell CC, Kreth S, Kuboki S, Chen P, et al. (2007) Targeted deletion of HIF-1alpha gene in T cells prevents their inhibition in hypoxic inflamed tissues and improves septic mice survival. PLoS One 2: e853. 17786224

67. Ben-Shoshan J, Maysel-Auslender S, Mor A, Keren G, George J (2008) Hypoxia controls CD4+CD25+ regulatory T-cell homeostasis via hypoxia-inducible factor-1alpha. Eur J Immunol 38: 2412–2418. doi: 10.1002/eji.200838318 18792019

68. Panther E, Corinti S, Idzko M, Herouy Y, Napp M, et al. (2003) Adenosine affects expression of membrane molecules, cytokine and chemokine release, and the T-cell stimulatory capacity of human dendritic cells. Blood 101: 3985–3990. 12446452

69. Brenner S, Prosch S, Schenke-Layland K, Riese U, Gausmann U, et al. (2003) cAMP-induced Interleukin-10 promoter activation depends on CCAAT/enhancer-binding protein expression and monocytic differentiation. J Biol Chem 278: 5597–5604. 12493739

70. Doedens AL, Stockmann C, Rubinstein MP, Liao D, Zhang N, et al. (2010) Macrophage expression of hypoxia-inducible factor-1 alpha suppresses T-cell function and promotes tumor progression. Cancer Res 70: 7465–7475. doi: 10.1158/0008-5472.CAN-10-1439 20841473

71. Colegio OR, Chu NQ, Szabo AL, Chu T, Rhebergen AM, et al. (2014) Functional polarization of tumour-associated macrophages by tumour-derived lactic acid. Nature 513: 559–563. doi: 10.1038/nature13490 25043024

72. Corzo CA, Condamine T, Lu L, Cotter MJ, Youn JI, et al. (2010) HIF-1alpha regulates function and differentiation of myeloid-derived suppressor cells in the tumor microenvironment. J Exp Med 207: 2439–2453. doi: 10.1084/jem.20100587 20876310

73. Noman MZ, Desantis G, Janji B, Hasmim M, Karray S, et al. (2014) PD-L1 is a novel direct target of HIF-1alpha, and its blockade under hypoxia enhanced MDSC-mediated T cell activation. J Exp Med 211: 781–790. doi: 10.1084/jem.20131916 24778419

74. Kilani MM, Mohammed KA, Nasreen N, Tepper RS, Antony VB (2004) RSV causes HIF-1alpha stabilization via NO release in primary bronchial epithelial cells. Inflammation 28: 245–251. 16133997

75. Nizet V, Johnson RS (2009) Interdependence of hypoxic and innate immune responses. Nat Rev Immunol 9: 609–617. doi: 10.1038/nri2607 19704417

76. Peyssonnaux C, Datta V, Cramer T, Doedens A, Theodorakis EA, et al. (2005) HIF-1alpha expression regulates the bactericidal capacity of phagocytes. J Clin Invest 115: 1806–1815. 16007254

77. Spear W, Chan D, Coppens I, Johnson RS, Giaccia A, et al. (2006) The host cell transcription factor hypoxia-inducible factor 1 is required for Toxoplasma gondii growth and survival at physiological oxygen levels. Cell Microbiol 8: 339–352. 16441443

78. Brown KM, Suvorova E, Farrell A, McLain A, Dittmar A, et al. (2014) Forward genetic screening identifies a small molecule that blocks Toxoplasma gondii growth by inhibiting both host- and parasite-encoded kinases. PLoS Pathog 10: e1004180. doi: 10.1371/journal.ppat.1004180 24945800

79. Cramer T, Yamanishi Y, Clausen BE, Forster I, Pawlinski R, et al. (2003) HIF-1alpha is essential for myeloid cell-mediated inflammation. Cell 112: 645–657. 12628185

80. Anand RJ, Gribar SC, Li J, Kohler JW, Branca MF, et al. (2007) Hypoxia causes an increase in phagocytosis by macrophages in a HIF-1alpha-dependent manner. J Leukoc Biol 82: 1257–1265. 17675562

81. Hwang II, Watson IR, Der SD, Ohh M (2006) Loss of VHL confers hypoxia-inducible factor (HIF)-dependent resistance to vesicular stomatitis virus: role of HIF in antiviral response. J Virol 80: 10712–10723. 16928739

82. Vassilaki N, Kalliampakou KI, Kotta-Loizou I, Befani C, Liakos P, et al. (2013) Low oxygen tension enhances hepatitis C virus replication. J Virol 87: 2935–2948. doi: 10.1128/JVI.02534-12 23269812

83. Veeranna RP, Haque M, Davis DA, Yang M, Yarchoan R (2012) Kaposi's sarcoma-associated herpesvirus latency-associated nuclear antigen induction by hypoxia and hypoxia-inducible factors. J Virol 86: 1097–1108. doi: 10.1128/JVI.05167-11 22090111

84. Tomaskova J, Oveckova I, Labudova M, Lukacikova L, Laposova K, et al. (2011) Hypoxia induces the gene expression and extracellular transmission of persistent lymphocytic choriomeningitis virus. J Virol 85: 13069–13076. doi: 10.1128/JVI.00829-11 21957293

85. Kohler T, Reizis B, Johnson RS, Weighardt H, Forster I (2012) Influence of hypoxia-inducible factor 1alpha on dendritic cell differentiation and migration. Eur J Immunol 42: 1226–1236. doi: 10.1002/eji.201142053 22539295

86. Weigert A, Weichand B, Sekar D, Sha W, Hahn C, et al. (2012) HIF-1alpha is a negative regulator of plasmacytoid DC development in vitro and in vivo. Blood 120: 3001–3006. doi: 10.1182/blood-2012-03-417022 22936665

87. Bankoti R, Gupta K, Levchenko A, Stager S (2012) Marginal zone B cells regulate antigen-specific T cell responses during infection. J Immunol 188: 3961–3971. doi: 10.4049/jimmunol.1102880 22412197

88. Stager S, Maroof A, Zubairi S, Sanos SL, Kopf M, et al. (2006) Distinct roles for IL-6 and IL-12p40 in mediating protection against Leishmania donovani and the expansion of IL-10+ CD4+ T cells. Eur J Immunol 36: 1764–1771. 16791879

89. Patel TH, Kimura H, Weiss CR, Semenza GL, Hofmann LV (2005) Constitutively active HIF-1alpha improves perfusion and arterial remodeling in an endovascular model of limb ischemia. Cardiovasc Res 68: 144–154. 15921668

90. Sarkar K, Cai Z, Gupta R, Parajuli N, Fox-Talbot K, et al. (2012) Hypoxia-inducible factor 1 transcriptional activity in endothelial cells is required for acute phase cardioprotection induced by ischemic preconditioning. Proc Natl Acad Sci U S A 109: 10504–10509. doi: 10.1073/pnas.1208314109 22699503

91. Ranatunga D, Hedrich CM, Wang F, McVicar DW, Nowak N, et al. (2009) A human IL10 BAC transgene reveals tissue-specific control of IL-10 expression and alters disease outcome. Proc Natl Acad Sci U S A 106: 17123–17128. doi: 10.1073/pnas.0904955106 19805095

Štítky
Hygiena a epidemiológia Infekčné lekárstvo Laboratórium

Článok vyšiel v časopise

PLOS Pathogens


2015 Číslo 6
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#