Skip to main content

Advertisement

Log in

Proinflammatory cytokines in heart failure: double-edged swords

  • Published:
Heart Failure Reviews Aims and scope Submit manuscript

Abstract

Increased circulating and intracardiac levels of proinflammatory cytokines have been associated with chronic heart failure. Following an initial insult, the increased production of proinflammatory cytokines, including TNF-α, IL-6, IL-1, and IL-18, jeopardizes the surrounding tissue through propagation of the inflammatory response and direct effects on the cardiac myocyte structure and function. Cardiac myocyte hypertrophy, contractile dysfunction, cardiac myocyte apoptosis, and extracellular matrix remodeling contribute enormously to the development and progression of chronic heart failure. Despite the identification of efficacious pharmacological regimens and introduction of mechanical interventions, chronic heart failure remains among the leading causes of mortality worldwide. To introduce novel therapeutic strategies that modulate the inflammatory response in the context of the failing heart, it is of prime importance to determine the contributions of TNF-α, IL-6, IL-1, and IL-18 in mediating cardiac adaptive and maladaptive responses, as well as delineating their downstream intracellular signaling pathways and their potential therapeutic implications.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3
Fig. 4

Similar content being viewed by others

References

  1. Oppenheim JJ (2001) Cytokines: past, present, and future. Int J Hematol 74:3–8

    Article  PubMed  CAS  Google Scholar 

  2. Mann DL (2002) Inflammatory mediators and the failing heart: past, present, and the foreseeable future. Circ Res 91:988–998

    Article  PubMed  CAS  Google Scholar 

  3. El-Menyar AA (2008) Cytokines and myocardial dysfunction: state of the art. J Card Fail 14:61–74

    Article  PubMed  CAS  Google Scholar 

  4. Petersen JW, Felker GM (2006) Inflammatory biomarkers in heart failure. Congest Heart Fail 12:324–328

    Article  PubMed  CAS  Google Scholar 

  5. Yndestad A, Damas JK, Oie E, Ueland T, Gullestad L, Aukrust P (2006) Systemic inflammation in heart failure–the whys and wherefores. Heart Fail Rev 11:83–92

    Article  PubMed  CAS  Google Scholar 

  6. Fairweather D, Rose NR (2005) Inflammatory heart disease: a role for cytokines. Lupus 14:646–651

    Article  PubMed  CAS  Google Scholar 

  7. Cihakova D, Rose NR (2008) Pathogenesis of myocarditis and dilated cardiomyopathy. Adv Immunol 99:95–114

    Article  PubMed  CAS  Google Scholar 

  8. Lane JR, Neumann DA, Lafond-Walker A, Herskowitz A, Rose NR (1993) Role of IL-1 and tumor necrosis factor in coxsackie virus-induced autoimmune myocarditis. J Immunol 151:1682–1690

    PubMed  CAS  Google Scholar 

  9. Fairweather D, Frisancho-Kiss S, Gatewood S, Njoku D, Steele R, Barrett M, Rose NR (2004) Mast cells and innate cytokines are associated with susceptibility to autoimmune heart disease following coxsackievirus B3 infection. Autoimmunity 37:131–145

    Article  PubMed  CAS  Google Scholar 

  10. Fairweather D, Yusung S, Frisancho S, Barrett M, Gatewood S, Steele R, Rose NR (2003) IL-12 receptor beta 1 and Toll-like receptor 4 increase IL-1 beta- and IL-18-associated myocarditis and coxsackievirus replication. J Immunol 170:4731–4737

    PubMed  CAS  Google Scholar 

  11. Hansson GK, Robertson AK, Soderberg-Naucler C (2006) Inflammation and atherosclerosis. Annu Rev Pathol 1:297–329

    Article  PubMed  CAS  Google Scholar 

  12. Robertson AK, Hansson GK (2006) T cells in atherogenesis: for better or for worse? Arterioscler Thromb Vasc Biol 26:2421–2432

    Article  PubMed  CAS  Google Scholar 

  13. Kleemann R, Zadelaar S, Kooistra T (2008) Cytokines and atherosclerosis: a comprehensive review of studies in mice. Cardiovasc Res 79:360–376

    Article  PubMed  CAS  Google Scholar 

  14. Kan H, Finkel MS (2001) Interactions between cytokines and neurohormonal systems in the failing heart. Heart Fail Rev 6:119–127

    Article  PubMed  CAS  Google Scholar 

  15. Henriksen PA, Newby DE (2003) Therapeutic inhibition of tumour necrosis factor alpha in patients with heart failure: cooling an inflamed heart. Heart 89:14–18

    Article  PubMed  CAS  Google Scholar 

  16. Roncon-Albuquerque R Jr, Vasconcelos M, Lourenco AP, Brandao-Nogueira A, Teles A, Henriques-Coelho T, Leite-Moreira AF (2006) Acute changes of biventricular gene expression in volume and right ventricular pressure overload. Life Sci 78:2633–2642

    Article  PubMed  CAS  Google Scholar 

  17. Baumgarten G, Knuefermann P, Kalra D, Gao F, Taffet GE, Michael L, Blackshear PJ, Carballo E, Sivasubramanian N, Mann DL (2002) Load-dependent and -independent regulation of proinflammatory cytokine and cytokine receptor gene expression in the adult mammalian heart. Circulation 105:2192–2197

    Article  PubMed  CAS  Google Scholar 

  18. Gurevitch J, Frolkis I, Yuhas Y, Paz Y, Matsa M, Mohr R, Yakirevich V (1996) Tumor necrosis factor-alpha is released from the isolated heart undergoing ischemia and reperfusion. J Am Coll Cardiol 28:247–252

    Article  PubMed  CAS  Google Scholar 

  19. Meldrum DR, Cleveland JC Jr, Cain BS, Meng X, Harken AH (1998) Increased myocardial tumor necrosis factor-alpha in a crystalloid-perfused model of cardiac ischemia-reperfusion injury. Ann Thorac Surg 65:439–443

    Article  PubMed  CAS  Google Scholar 

  20. Kapadia S, Lee J, Torre-Amione G, Birdsall HH, Ma TS, Mann DL (1995) Tumor necrosis factor-alpha gene and protein expression in adult feline myocardium after endotoxin administration. J Clin Invest 96:1042–1052

    Article  PubMed  CAS  Google Scholar 

  21. Giroir BP, Johnson JH, Brown T, Allen GL, Beutler B (1992) The tissue distribution of tumor necrosis factor biosynthesis during endotoxemia. J Clin Invest 90:693–698

    Article  PubMed  CAS  Google Scholar 

  22. Nakano M, Knowlton AA, Dibbs Z, Mann DL (1998) Tumor necrosis factor-alpha confers resistance to hypoxic injury in the adult mammalian cardiac myocyte. Circulation 97:1392–1400

    PubMed  CAS  Google Scholar 

  23. Yokoyama T, Nakano M, Bednarczyk JL, McIntyre BW, Entman M, Mann DL (1997) Tumor necrosis factor-alpha provokes a hypertrophic growth response in adult cardiac myocytes. Circulation 95:1247–1252

    PubMed  CAS  Google Scholar 

  24. Engel D, Peshock R, Armstong RC, Sivasubramanian N, Mann DL (2004) Cardiac myocyte apoptosis provokes adverse cardiac remodeling in transgenic mice with targeted TNF overexpression. Am J Physiol Heart Circ Physiol 287:H1303–H1311

    Article  PubMed  CAS  Google Scholar 

  25. Tovey MG (1989) Expression of the genes of interferons and other cytokines in normal and diseased tissues of man. Experientia 45:526–535

    Article  PubMed  CAS  Google Scholar 

  26. Tovey MG, Content J, Gresser I, Gugenheim J, Blanchard B, Guymarho J, Poupart P, Gigou M, Shaw A, Fiers W (1988) Genes for IFN-beta-2 (IL-6), tumor necrosis factor, and IL-1 are expressed at high levels in the organs of normal individuals. J Immunol 141:3106–3110

    PubMed  CAS  Google Scholar 

  27. Hunt JS, Chen HL, Hu XL, Chen TY, Morrison DC (1992) Tumor necrosis factor-alpha gene expression in the tissues of normal mice. Cytokine 4:340–346

    Article  PubMed  CAS  Google Scholar 

  28. Sharma R, Anker SD (2002) Cytokines, apoptosis and cachexia: the potential for TNF antagonism. Int J Cardiol 85:161–171

    Article  PubMed  Google Scholar 

  29. Ura H, Hirata K, Yamaguchi K, Katsuramaki T, Denno R (1998) Mechanism of the development of organ failure. Nippon Geka Gakkai Zasshi 99:485–489

    PubMed  CAS  Google Scholar 

  30. Esmon CT (1999) Possible involvement of cytokines in diffuse intravascular coagulation and thrombosis. Baillieres Best Pract Res Clin Haematol 12:343–359

    PubMed  CAS  Google Scholar 

  31. Caille V, Bossi P, Grimaldi D, Vieillard-Baro A (2004) Physiopathology of severe sepsis. Presse Med 33:256–261 (discussion 269)

    Article  PubMed  Google Scholar 

  32. von Haehling S, Jankowska EA, Anker SD (2004) Tumour necrosis factor-alpha and the failing heart–pathophysiology and therapeutic implications. Basic Res Cardiol 99:18–28

    Article  CAS  Google Scholar 

  33. Hamid T, Gu Y, Ortines RV, Bhattacharya C, Wang G, Xuan YT, Prabhu SD (2009) Divergent tumor necrosis factor receptor-related remodeling responses in heart failure: role of nuclear factor-kappaB and inflammatory activation. Circulation 119:1386–1397

    Article  PubMed  CAS  Google Scholar 

  34. Higuchi Y, McTiernan CF, Frye CB, McGowan BS, Chan TO, Feldman AM (2004) Tumor necrosis factor receptors 1 and 2 differentially regulate survival, cardiac dysfunction, and remodeling in transgenic mice with tumor necrosis factor-alpha-induced cardiomyopathy. Circulation 109:1892–1897

    Article  PubMed  CAS  Google Scholar 

  35. Monden Y, Kubota T, Inoue T, Tsutsumi T, Kawano S, Ide T, Tsutsui H, Sunagawa K (2007) Tumor necrosis factor-alpha is toxic via receptor 1 and protective via receptor 2 in a murine model of myocardial infarction. Am J Physiol Heart Circ Physiol 293:H743–H753

    Article  PubMed  CAS  Google Scholar 

  36. Nozaki N, Yamaguchi S, Yamaoka M, Okuyama M, Nakamura H, Tomoike H (1998) Enhanced expression and shedding of tumor necrosis factor (TNF) receptors from mononuclear leukocytes in human heart failure. J Mol Cell Cardiol 30:2003–2012

    Article  PubMed  CAS  Google Scholar 

  37. Balakumar P, Singh M (2006) Anti-tumour necrosis factor-alpha therapy in heart failure: future directions. Basic Clin Pharmacol Toxicol 99:391–397

    Article  PubMed  CAS  Google Scholar 

  38. Bozkurt B (2000) Activation of cytokines as a mechanism of disease progression in heart failure. Ann Rheum Dis 59(Suppl 1):i90–i93

    Article  PubMed  CAS  Google Scholar 

  39. Eddy LJ, Goeddel DV, Wong GH (1992) Tumor necrosis factor-alpha pretreatment is protective in a rat model of myocardial ischemia-reperfusion injury. Biochem Biophys Res Commun 184:1056–1059

    Article  PubMed  CAS  Google Scholar 

  40. Chen Z, Siu B, Ho YS, Vincent R, Chua CC, Hamdy RC, Chua BH (1998) Overexpression of MnSOD protects against myocardial ischemia/reperfusion injury in transgenic mice. J Mol Cell Cardiol 30:2281–2289

    Article  PubMed  CAS  Google Scholar 

  41. Wong GH, Goeddel DV (1988) Induction of manganous superoxide dismutase by tumor necrosis factor: possible protective mechanism. Science 242:941–944

    Article  PubMed  CAS  Google Scholar 

  42. Nakano M, Knowlton AA, Yokoyama T, Lesslauer W, Mann DL (1996) Tumor necrosis factor-alpha-induced expression of heat shock protein 72 in adult feline cardiac myocytes. Am J Physiol 270:H1231–H1239

    PubMed  CAS  Google Scholar 

  43. Sharma HS, Stahl J, Weisensee D, Low-Friedrich I (1996) Cytoprotective mechanisms in cultured cardiomyocytes. Mol Cell Biochem 160–161:217–224

    Article  PubMed  Google Scholar 

  44. Low-Friedrich I, Weisensee D, Mitrou P, Schoeppe W (1992) Cytokines induce stress protein formation in cultured cardiac myocytes. Basic Res Cardiol 87:12–18

    Article  PubMed  CAS  Google Scholar 

  45. Meng X, Harken AH (2002) The interaction between Hsp70 and TNF-alpha expression: a novel mechanism for protection of the myocardium against post-injury depression. Shock 17:345–353

    Article  PubMed  Google Scholar 

  46. Grunenfelder J, Zund G, Stucki V, Hoerstrup SP, Kadner A, Schoeberlein A, Turina M (2001) Heat shock protein upregulation lowers cytokine levels after ischemia and reperfusion. Eur Surg Res 33:383–387

    Article  PubMed  CAS  Google Scholar 

  47. Meng X, Banerjee A, Ao L, Meldrum DR, Cain BS, Shames BD, Harken AH (1999) Inhibition of myocardial TNF-alpha production by heat shock. A potential mechanism of stress-induced cardioprotection against postischemic dysfunction. Ann N Y Acad Sci 874:69–82

    Article  PubMed  CAS  Google Scholar 

  48. Sun M, Chen M, Dawood F, Zurawska U, Li JY, Parker T, Kassiri Z, Kirshenbaum LA, Arnold M, Khokha R, Liu PP (2007) Tumor necrosis factor-alpha mediates cardiac remodeling and ventricular dysfunction after pressure overload state. Circulation 115:1398–1407

    Article  PubMed  CAS  Google Scholar 

  49. Janczewski AM, Kadokami T, Lemster B, Frye CS, McTiernan CF, Feldman AM (2003) Morphological and functional changes in cardiac myocytes isolated from mice overexpressing TNF-alpha. Am J Physiol Heart Circ Physiol 284:H960–H969

    PubMed  CAS  Google Scholar 

  50. Dibbs ZI, Diwan A, Nemoto S, DeFreitas G, Abdellatif M, Carabello BA, Spinale FG, Feuerstein G, Sivasubramanian N, Mann DL (2003) Targeted overexpression of transmembrane tumor necrosis factor provokes a concentric cardiac hypertrophic phenotype. Circulation 108:1002–1008

    Article  PubMed  CAS  Google Scholar 

  51. Turner NA, Mughal RS, Warburton P, O’Regan DJ, Ball SG, Porter KE (2007) Mechanism of TNFalpha-induced IL-1alpha, IL-1beta and IL-6 expression in human cardiac fibroblasts: effects of statins and thiazolidinediones. Cardiovasc Res 76:81–90

    Article  PubMed  CAS  Google Scholar 

  52. Isoda K, Kamezawa Y, Tada N, Sato M, Ohsuzu F (2001) Myocardial hypertrophy in transgenic mice overexpressing human interleukin 1 alpha. J Card Fail 7:355–364

    Article  PubMed  CAS  Google Scholar 

  53. Hirota H, Yoshida K, Kishimoto T, Taga T (1995) Continuous activation of gp130, a signal-transducing receptor component for interleukin 6-related cytokines, causes myocardial hypertrophy in mice. Proc Natl Acad Sci USA 92:4862–4866

    Article  PubMed  CAS  Google Scholar 

  54. Flesch M, Hoper A, Dell’Italia L, Evans K, Bond R, Peshock R, Diwan A, Brinsa TA, Wei CC, Sivasubramanian N, Spinale FG, Mann DL (2003) Activation and functional significance of the renin-angiotensin system in mice with cardiac restricted overexpression of tumor necrosis factor. Circulation 108:598–604

    Article  PubMed  CAS  Google Scholar 

  55. Nakamura K, Fushimi K, Kouchi H, Mihara K, Miyazaki M, Ohe T, Namba M (1998) Inhibitory effects of antioxidants on neonatal rat cardiac myocyte hypertrophy induced by tumor necrosis factor-alpha and angiotensin II. Circulation 98:794–799

    PubMed  CAS  Google Scholar 

  56. Kumar A, Brar R, Wang P, Dee L, Skorupa G, Khadour F, Schulz R, Parrillo JE (1999) Role of nitric oxide and cGMP in human septic serum-induced depression of cardiac myocyte contractility. Am J Physiol 276:R265–R276

    PubMed  CAS  Google Scholar 

  57. Oral H, Dorn GW 2nd, Mann DL (1997) Sphingosine mediates the immediate negative inotropic effects of tumor necrosis factor-alpha in the adult mammalian cardiac myocyte. J Biol Chem 272:4836–4842

    Article  PubMed  CAS  Google Scholar 

  58. Kumar A, Paladugu B, Mensing J, Parrillo JE (2007) Nitric oxide-dependent and -independent mechanisms are involved in TNF-alpha -induced depression of cardiac myocyte contractility. Am J Physiol Regul Integr Comp Physiol 292:R1900–R1906

    PubMed  CAS  Google Scholar 

  59. Kojda G, Kottenberg K (1999) Regulation of basal myocardial function by NO. Cardiovasc Res 41:514–523

    Article  PubMed  CAS  Google Scholar 

  60. Stein B, Frank P, Schmitz W, Scholz H, Thoenes M (1996) Endotoxin and cytokines induce direct cardiodepressive effects in mammalian cardiomyocytes via induction of nitric oxide synthase. J Mol Cell Cardiol 28:1631–1639

    Article  PubMed  CAS  Google Scholar 

  61. Schulz R, Panas DL, Catena R, Moncada S, Olley PM, Lopaschuk GD (1995) The role of nitric oxide in cardiac depression induced by interleukin-1 beta and tumour necrosis factor-alpha. Br J Pharmacol 114:27–34

    PubMed  CAS  Google Scholar 

  62. Ungureanu-Longrois D, Balligand JL, Simmons WW, Okada I, Kobzik L, Lowenstein CJ, Kunkel SL, Michel T, Kelly RA, Smith TW (1995) Induction of nitric oxide synthase activity by cytokines in ventricular myocytes is necessary but not sufficient to decrease contractile responsiveness to beta-adrenergic agonists. Circ Res 77:494–502

    PubMed  CAS  Google Scholar 

  63. Ungureanu-Longrois D, Balligand JL, Okada I, Simmons WW, Kobzik L, Lowenstein CJ, Kunkel SL, Michel T, Kelly RA, Smith TW (1995) Contractile responsiveness of ventricular myocytes to isoproterenol is regulated by induction of nitric oxide synthase activity in cardiac microvascular endothelial cells in heterotypic primary culture. Circ Res 77:486–493

    PubMed  CAS  Google Scholar 

  64. Kelm M, Schrader J (1990) Control of coronary vascular tone by nitric oxide. Circ Res 66:1561–1575

    PubMed  CAS  Google Scholar 

  65. Radomski MW, Palmer RM, Moncada S (1987) The anti-aggregating properties of vascular endothelium: interactions between prostacyclin and nitric oxide. Br J Pharmacol 92:639–646

    PubMed  CAS  Google Scholar 

  66. Elahi M, Asopa S, Matata B (2007) NO-cGMP and TNF-alpha counter regulatory system in blood: understanding the mechanisms leading to myocardial dysfunction and failure. Biochim Biophys Acta 1772:5–14

    PubMed  CAS  Google Scholar 

  67. Dettbarn CA, Betto R, Salviati G, Palade P, Jenkins GM, Sabbadini RA (1994) Modulation of cardiac sarcoplasmic reticulum ryanodine receptor by sphingosine. J Mol Cell Cardiol 26:229–242

    Article  PubMed  CAS  Google Scholar 

  68. Goldhaber JI, Kim KH, Natterson PD, Lawrence T, Yang P, Weiss JN (1996) Effects of TNF-alpha on [Ca2+]i and contractility in isolated adult rabbit ventricular myocytes. Am J Physiol 271:H1449–H1455

    PubMed  CAS  Google Scholar 

  69. Haudek SB, Taffet GE, Schneider MD, Mann DL (2007) TNF provokes cardiomyocyte apoptosis and cardiac remodeling through activation of multiple cell death pathways. J Clin Invest 117:2692–2701

    Article  PubMed  CAS  Google Scholar 

  70. Bozkurt B, Kribbs SB, Clubb FJ Jr, Michael LH, Didenko VV, Hornsby PJ, Seta Y, Oral H, Spinale FG, Mann DL (1998) Pathophysiologically relevant concentrations of tumor necrosis factor-alpha promote progressive left ventricular dysfunction and remodeling in rats. Circulation 97:1382–1391

    PubMed  CAS  Google Scholar 

  71. Kubota T, McTiernan CF, Frye CS, Slawson SE, Lemster BH, Koretsky AP, Demetris AJ, Feldman AM (1997) Dilated cardiomyopathy in transgenic mice with cardiac-specific overexpression of tumor necrosis factor-alpha. Circ Res 81:627–635

    PubMed  CAS  Google Scholar 

  72. O’Brien NW, Gellings NM, Guo M, Barlow SB, Glembotski CC, Sabbadini RA (2003) Factor associated with neutral sphingomyelinase activation and its role in cardiac cell death. Circ Res 92:589–591

    Article  PubMed  CAS  Google Scholar 

  73. Caulfield JB, Borg TK (1979) The collagen network of the heart. Lab Invest 40:364–372

    PubMed  CAS  Google Scholar 

  74. Fedak PW, Verma S, Weisel RD, Li RK (2005) Cardiac remodeling and failure from molecules to man (part II). Cardiovasc Pathol 14:49–60

    Article  PubMed  CAS  Google Scholar 

  75. Ju H, Dixon IM (1996) Extracellular matrix and cardiovascular diseases. Can J Cardiol 12:1259–1267

    PubMed  CAS  Google Scholar 

  76. Eghbali M, Blumenfeld OO, Seifter S, Buttrick PM, Leinwand LA, Robinson TF, Zern MA, Giambrone MA (1989) Localization of types I, III and IV collagen mRNAs in rat heart cells by in situ hybridization. J Mol Cell Cardiol 21:103–113

    Article  PubMed  CAS  Google Scholar 

  77. Eghbali M, Czaja MJ, Zeydel M, Weiner FR, Zern MA, Seifter S, Blumenfeld OO (1988) Collagen chain mRNAs in isolated heart cells from young and adult rats. J Mol Cell Cardiol 20:267–276

    Article  PubMed  CAS  Google Scholar 

  78. Siwik DA, Chang DL, Colucci WS (2000) Interleukin-1beta and tumor necrosis factor-alpha decrease collagen synthesis and increase matrix metalloproteinase activity in cardiac fibroblasts in vitro. Circ Res 86:1259–1265

    PubMed  CAS  Google Scholar 

  79. Siwik DA, Colucci WS (2004) Regulation of matrix metalloproteinases by cytokines and reactive oxygen/nitrogen species in the myocardium. Heart Fail Rev 9:43–51

    Article  PubMed  CAS  Google Scholar 

  80. Sivasubramanian N, Coker ML, Kurrelmeyer KM, MacLellan WR, DeMayo FJ, Spinale FG, Mann DL (2001) Left ventricular remodeling in transgenic mice with cardiac restricted overexpression of tumor necrosis factor. Circulation 104:826–831

    Article  PubMed  CAS  Google Scholar 

  81. Li YY, Feng YQ, Kadokami T, McTiernan CF, Draviam R, Watkins SC, Feldman AM (2000) Myocardial extracellular matrix remodeling in transgenic mice overexpressing tumor necrosis factor alpha can be modulated by anti-tumor necrosis factor alpha therapy. Proc Natl Acad Sci USA 97:12746–12751

    Article  PubMed  CAS  Google Scholar 

  82. Gurantz D, Cowling RT, Villarreal FJ, Greenberg BH (1999) Tumor necrosis factor-alpha upregulates angiotensin II type 1 receptors on cardiac fibroblasts. Circ Res 85:272–279

    PubMed  CAS  Google Scholar 

  83. Peng J, Gurantz D, Tran V, Cowling RT, Greenberg BH (2002) Tumor necrosis factor-alpha-induced AT1 receptor upregulation enhances angiotensin II-mediated cardiac fibroblast responses that favor fibrosis. Circ Res 91:1119–1126

    Article  PubMed  CAS  Google Scholar 

  84. Bradham WS, Moe G, Wendt KA, Scott AA, Konig A, Romanova M, Naik G, Spinale FG (2002) TNF-alpha and myocardial matrix metalloproteinases in heart failure: relationship to LV remodeling. Am J Physiol Heart Circ Physiol 282:H1288–H1295

    PubMed  CAS  Google Scholar 

  85. Li YY, Kadokami T, Wang P, McTiernan CF, Feldman AM (2002) MMP inhibition modulates TNF-alpha transgenic mouse phenotype early in the development of heart failure. Am J Physiol Heart Circ Physiol 282:H983–H989

    PubMed  CAS  Google Scholar 

  86. Deschamps AM, Spinale FG (2006) Pathways of matrix metalloproteinase induction in heart failure: bioactive molecules and transcriptional regulation. Cardiovasc Res 69:666–676

    Article  PubMed  CAS  Google Scholar 

  87. Liacini A, Sylvester J, Li WQ, Huang W, Dehnade F, Ahmad M, Zafarullah M (2003) Induction of matrix metalloproteinase-13 gene expression by TNF-alpha is mediated by MAP kinases, AP-1, and NF-kappaB transcription factors in articular chondrocytes. Exp Cell Res 288:208–217

    Article  PubMed  CAS  Google Scholar 

  88. Yeh CH, Lin YM, Wu YC, Lin PJ (2005) Inhibition of NF-kappa B activation can attenuate ischemia/reperfusion-induced contractility impairment via decreasing cardiomyocytic proinflammatory gene up-regulation and matrix metalloproteinase expression. J Cardiovasc Pharmacol 45:301–309

    Article  PubMed  CAS  Google Scholar 

  89. Fischer P, Hilfiker-Kleiner D (2008) Role of gp130-mediated signalling pathways in the heart and its impact on potential therapeutic aspects. Br J Pharmacol 153(Suppl 1):S414–S427

    PubMed  CAS  Google Scholar 

  90. Heinrich PC, Behrmann I, Muller-Newen G, Schaper F, Graeve L (1998) Interleukin-6-type cytokine signalling through the gp130/Jak/STAT pathway. Biochem J 334(Pt 2):297–314

    PubMed  CAS  Google Scholar 

  91. Fischer P, Hilfiker-Kleiner D (2007) Survival pathways in hypertrophy and heart failure: the gp130-STAT3 axis. Basic Res Cardiol 102:279–297

    Article  PubMed  CAS  Google Scholar 

  92. Naka T, Nishimoto N, Kishimoto T (2002) The paradigm of IL-6: from basic science to medicine. Arthritis Res 4(Suppl 3):S233–242

    Article  PubMed  Google Scholar 

  93. Hilfiker-Kleiner D, Hilfiker A, Drexler H (2005) Many good reasons to have STAT3 in the heart. Pharmacol Ther 107:131–137

    Article  PubMed  CAS  Google Scholar 

  94. Yamauchi-Takihara K, Kishimoto T (2000) Cytokines and their receptors in cardiovascular diseases–role of gp130 signalling pathway in cardiac myocyte growth and maintenance. Int J Exp Pathol 81:1–16

    Article  PubMed  CAS  Google Scholar 

  95. Smart N, Mojet MH, Latchman DS, Marber MS, Duchen MR, Heads RJ (2006) IL-6 induces PI 3-kinase and nitric oxide-dependent protection and preserves mitochondrial function in cardiomyocytes. Cardiovasc Res 69:164–177

    Article  PubMed  CAS  Google Scholar 

  96. Liao Z, Brar BK, Cai Q, Stephanou A, O’Leary RM, Pennica D, Yellon DM, Latchman DS (2002) Cardiotrophin-1 (CT-1) can protect the adult heart from injury when added both prior to ischaemia and at reperfusion. Cardiovasc Res 53:902–910

    Article  PubMed  CAS  Google Scholar 

  97. Negoro S, Kunisada K, Tone E, Funamoto M, Oh H, Kishimoto T, Yamauchi-Takihara K (2000) Activation of JAK/STAT pathway transduces cytoprotective signal in rat acute myocardial infarction. Cardiovasc Res 47:797–805

    Article  PubMed  CAS  Google Scholar 

  98. Hattori R, Maulik N, Otani H, Zhu L, Cordis G, Engelman RM, Siddiqui MA, Das DK (2001) Role of STAT3 in ischemic preconditioning. J Mol Cell Cardiol 33:1929–1936

    Article  PubMed  CAS  Google Scholar 

  99. Hirota H, Chen J, Betz UA, Rajewsky K, Gu Y, Ross J Jr, Muller W, Chien KR (1999) Loss of a gp130 cardiac muscle cell survival pathway is a critical event in the onset of heart failure during biomechanical stress. Cell 97:189–198

    Article  PubMed  CAS  Google Scholar 

  100. Lopez N, Varo N, Diez J, Fortuno MA (2007) Loss of myocardial LIF receptor in experimental heart failure reduces cardiotrophin-1 cytoprotection. A role for neurohumoral agonists? Cardiovasc Res 75:536–545

    Article  PubMed  CAS  Google Scholar 

  101. Negoro S, Oh H, Tone E, Kunisada K, Fujio Y, Walsh K, Kishimoto T, Yamauchi-Takihara K (2001) Glycoprotein 130 regulates cardiac myocyte survival in doxorubicin-induced apoptosis through phosphatidylinositol 3-kinase/Akt phosphorylation and Bcl-xL/caspase-3 interaction. Circulation 103:555–561

    PubMed  CAS  Google Scholar 

  102. Ito H, Miller SC, Billingham ME, Akimoto H, Torti SV, Wade R, Gahlmann R, Lyons G, Kedes L, Torti FM (1990) Doxorubicin selectively inhibits muscle gene expression in cardiac muscle cells in vivo and in vitro. Proc Natl Acad Sci USA 87:4275–4279

    Article  PubMed  CAS  Google Scholar 

  103. Jeyaseelan R, Poizat C, Wu HY, Kedes L (1997) Molecular mechanisms of doxorubicin-induced cardiomyopathy. Selective suppression of Reiske iron-sulfur protein, ADP/ATP translocase, and phosphofructokinase genes is associated with ATP depletion in rat cardiomyocytes. J Biol Chem 272:5828–5832

    Article  PubMed  CAS  Google Scholar 

  104. Jacoby JJ, Kalinowski A, Liu MG, Zhang SS, Gao Q, Chai GX, Ji L, Iwamoto Y, Li E, Schneider M, Russell KS, Fu XY (2003) Cardiomyocyte-restricted knockout of STAT3 results in higher sensitivity to inflammation, cardiac fibrosis, and heart failure with advanced age. Proc Natl Acad Sci USA 100:12929–12934

    Article  PubMed  CAS  Google Scholar 

  105. Yajima T, Yasukawa H, Jeon ES, Xiong D, Dorner A, Iwatate M, Nara M, Zhou H, Summers-Torres D, Hoshijima M, Chien KR, Yoshimura A, Knowlton KU (2006) Innate defense mechanism against virus infection within the cardiac myocyte requiring gp130-STAT3 signaling. Circulation 114:2364–2373

    Article  PubMed  CAS  Google Scholar 

  106. Saito M, Yoshida K, Hibi M, Taga T, Kishimoto T (1992) Molecular cloning of a murine IL-6 receptor-associated signal transducer, gp130, and its regulated expression in vivo. J Immunol 148:4066–4071

    PubMed  CAS  Google Scholar 

  107. Matsui H, Fujio Y, Kunisada K, Hirota H, Yamauchi-Takihara K (1996) Leukemia inhibitory factor induces a hypertrophic response mediated by gp130 in murine cardiac myocytes. Res Commun Mol Pathol Pharmacol 93:149–162

    PubMed  CAS  Google Scholar 

  108. Pennica D, Shaw KJ, Swanson TA, Moore MW, Shelton DL, Zioncheck KA, Rosenthal A, Taga T, Paoni NF, Wood WI (1995) Cardiotrophin-1. Biological activities and binding to the leukemia inhibitory factor receptor/gp130 signaling complex. J Biol Chem 270:10915–10922

    Article  PubMed  CAS  Google Scholar 

  109. Wollert KC, Taga T, Saito M, Narazaki M, Kishimoto T, Glembotski CC, Vernallis AB, Heath JK, Pennica D, Wood WI, Chien KR (1996) Cardiotrophin-1 activates a distinct form of cardiac muscle cell hypertrophy. Assembly of sarcomeric units in series VIA gp130/leukemia inhibitory factor receptor-dependent pathways. J Biol Chem 271:9535–9545

    Article  PubMed  CAS  Google Scholar 

  110. Kunisada K, Negoro S, Tone E, Funamoto M, Osugi T, Yamada S, Okabe M, Kishimoto T, Yamauchi-Takihara K (2000) Signal transducer and activator of transcription 3 in the heart transduces not only a hypertrophic signal but a protective signal against doxorubicin-induced cardiomyopathy. Proc Natl Acad Sci USA 97:315–319

    Article  PubMed  CAS  Google Scholar 

  111. Kunisada K, Hirota H, Fujio Y, Matsui H, Tani Y, Yamauchi-Takihara K, Kishimoto T (1996) Activation of JAK-STAT and MAP kinases by leukemia inhibitory factor through gp130 in cardiac myocytes. Circulation 94:2626–2632

    PubMed  CAS  Google Scholar 

  112. Oh H, Fujio Y, Kunisada K, Hirota H, Matsui H, Kishimoto T, Yamauchi-Takihara K (1998) Activation of phosphatidylinositol 3-kinase through glycoprotein 130 induces protein kinase B and p70 S6 kinase phosphorylation in cardiac myocytes. J Biol Chem 273:9703–9710

    Article  PubMed  CAS  Google Scholar 

  113. Li YJ, Cui W, Tian ZJ, Hao YM, Du J, Liu F, Zhang H, Zu XG, Liu SY, Xie RQ, Yang XH, Wu YZ, Chen L, An W (2004) Crosstalk between ERK1/2 and STAT3 in the modulation of cardiomyocyte hypertrophy induced by cardiotrophin-1. Chin Med J (Engl) 117:1135–1142

    CAS  Google Scholar 

  114. Kodama H, Fukuda K, Pan J, Sano M, Takahashi T, Kato T, Makino S, Manabe T, Murata M, Ogawa S (2000) Significance of ERK cascade compared with JAK/STAT and PI3-K pathway in gp130-mediated cardiac hypertrophy. Am J Physiol Heart Circ Physiol 279:H1635–H1644

    PubMed  CAS  Google Scholar 

  115. Kunisada K, Tone E, Fujio Y, Matsui H, Yamauchi-Takihara K, Kishimoto T (1998) Activation of gp130 transduces hypertrophic signals via STAT3 in cardiac myocytes. Circulation 98:346–352

    PubMed  CAS  Google Scholar 

  116. Zhang X, Blenis J, Li HC, Schindler C, Chen-Kiang S (1995) Requirement of serine phosphorylation for formation of STAT-promoter complexes. Science 267:1990–1994

    Article  PubMed  CAS  Google Scholar 

  117. Wen Z, Zhong Z, Darnell JE Jr (1995) Maximal activation of transcription by Stat1 and Stat3 requires both tyrosine and serine phosphorylation. Cell 82:241–250

    Article  PubMed  CAS  Google Scholar 

  118. Tian ZJ, Cui W, Li YJ, Hao YM, Du J, Liu F, Zhang H, Zu XG, Liu SY, Chen L, An W (2004) Different contributions of STAT3, ERK1/2, and PI3-K signaling to cardiomyocyte hypertrophy by cardiotrophin-1. Acta Pharmacol Sin 25:1157–1164

    PubMed  CAS  Google Scholar 

  119. Miyamoto T, Takeishi Y, Takahashi H, Shishido T, Arimoto T, Tomoike H, Kubota I (2004) Activation of distinct signal transduction pathways in hypertrophied hearts by pressure and volume overload. Basic Res Cardiol 99:328–337

    Article  PubMed  CAS  Google Scholar 

  120. Maass DL, White J, Horton JW (2002) IL-1beta and IL-6 act synergistically with TNF-alpha to alter cardiac contractile function after burn trauma. Shock 18:360–366

    Article  PubMed  Google Scholar 

  121. Kinugawa K, Takahashi T, Kohmoto O, Yao A, Aoyagi T, Momomura S, Hirata Y, Serizawa T (1994) Nitric oxide-mediated effects of interleukin-6 on [Ca2+]i and cell contraction in cultured chick ventricular myocytes. Circ Res 75:285–295

    PubMed  CAS  Google Scholar 

  122. Yu X, Kennedy RH, Liu SJ (2003) JAK2/STAT3, not ERK1/2, mediates interleukin-6-induced activation of inducible nitric-oxide synthase and decrease in contractility of adult ventricular myocytes. J Biol Chem 278:16304–16309

    Article  PubMed  CAS  Google Scholar 

  123. Tsuruda T, Jougasaki M, Boerrigter G, Huntley BK, Chen HH, D’Assoro AB, Lee SC, Larsen AM, Cataliotti A, Burnett JC Jr (2002) Cardiotrophin-1 stimulation of cardiac fibroblast growth: roles for glycoprotein 130/leukemia inhibitory factor receptor and the endothelin type A receptor. Circ Res 90:128–134

    Article  PubMed  CAS  Google Scholar 

  124. Wang F, Trial J, Diwan A, Gao F, Birdsall H, Entman M, Hornsby P, Sivasubramaniam N, Mann D (2002) Regulation of cardiac fibroblast cellular function by leukemia inhibitory factor. J Mol Cell Cardiol 34:1309–1316

    Article  PubMed  CAS  Google Scholar 

  125. Gallagher G, Menzie S, Huang Y, Jackson C, Hunyor SN (2007) Regional cardiac dysfunction is associated with specific alterations in inflammatory cytokines and matrix metalloproteinases after acute myocardial infarction in sheep. Basic Res Cardiol 102:63–72

    Article  PubMed  CAS  Google Scholar 

  126. Weiss TW, Kvakan H, Kaun C, Zorn G, Speidl WS, Pfaffenberger S, Maurer G, Huber K, Wojta J (2005) The gp130 ligand oncostatin M regulates tissue inhibitor of metalloproteinases-1 through ERK1/2 and p38 in human adult cardiac myocytes and in human adult cardiac fibroblasts: a possible role for the gp130/gp130 ligand system in the modulation of extracellular matrix degradation in the human heart. J Mol Cell Cardiol 39:545–551

    Article  PubMed  CAS  Google Scholar 

  127. Dinarello CA (2009) Immunological and inflammatory functions of the interleukin-1 family. Annu Rev Immunol 27:519–550

    Article  PubMed  CAS  Google Scholar 

  128. Dinarello CA (1998) Interleukin-1, interleukin-1 receptors and interleukin-1 receptor antagonist. Int Rev Immunol 16:457–499

    Article  PubMed  CAS  Google Scholar 

  129. Yue P, Massie BM, Simpson PC, Long CS (1998) Cytokine expression increases in nonmyocytes from rats with postinfarction heart failure. Am J Physiol 275:H250–H258

    PubMed  CAS  Google Scholar 

  130. Shioi T, Matsumori A, Kihara Y, Inoko M, Ono K, Iwanaga Y, Yamada T, Iwasaki A, Matsushima K, Sasayama S (1997) Increased expression of interleukin-1 beta and monocyte chemotactic and activating factor/monocyte chemoattractant protein-1 in the hypertrophied and failing heart with pressure overload. Circ Res 81:664–671

    PubMed  CAS  Google Scholar 

  131. Freeman GL, Colston JT, Zabalgoitia M, Chandrasekar B (1998) Contractile depression and expression of proinflammatory cytokines and iNOS in viral myocarditis. Am J Physiol 274:H249–H258

    PubMed  CAS  Google Scholar 

  132. Ono K, Matsumori A, Shioi T, Furukawa Y, Sasayama S (1998) Cytokine gene expression after myocardial infarction in rat hearts: possible implication in left ventricular remodeling. Circulation 98:149–156

    PubMed  CAS  Google Scholar 

  133. Brown JM, White CW, Terada LS, Grosso MA, Shanley PF, Mulvin DW, Banerjee A, Whitman GJ, Harken AH, Repine JE (1990) Interleukin 1 pretreatment decreases ischemia/reperfusion injury. Proc Natl Acad Sci USA 87:5026–5030

    Article  PubMed  CAS  Google Scholar 

  134. Maulik N, Engelman RM, Wei Z, Lu D, Rousou JA, Das DK (1993) Interleukin-1 alpha preconditioning reduces myocardial ischemia reperfusion injury. Circulation 88:387–394

    CAS  Google Scholar 

  135. Nogae C, Makino N, Hata T, Nogae I, Takahashi S, Suzuki K, Taniguchi N, Yanaga T (1995) Interleukin 1 alpha-induced expression of manganous superoxide dismutase reduces myocardial reperfusion injury in the rat. J Mol Cell Cardiol 27:2091–2099

    Article  PubMed  CAS  Google Scholar 

  136. Zhang ML, Li ZP, Xiao H (2007) Different expressions of inflammatory cytokines in two types of cardiac hypertrophy in rats. Beijing Da Xue Xue Bao 39:570–575

    PubMed  CAS  Google Scholar 

  137. Dai RP, Dheen ST, He BP, Tay SS (2004) Differential expression of cytokines in the rat heart in response to sustained volume overload. Eur J Heart Fail 6:693–703

    Article  PubMed  CAS  Google Scholar 

  138. Palmer JN, Hartogensis WE, Patten M, Fortuin FD, Long CS (1995) Interleukin-1 beta induces cardiac myocyte growth but inhibits cardiac fibroblast proliferation in culture. J Clin Invest 95:2555–2564

    Article  PubMed  CAS  Google Scholar 

  139. Thaik CM, Calderone A, Takahashi N, Colucci WS (1995) Interleukin-1 beta modulates the growth and phenotype of neonatal rat cardiac myocytes. J Clin Invest 96:1093–1099

    Article  PubMed  CAS  Google Scholar 

  140. Harada E, Nakagawa O, Yoshimura M, Harada M, Nakagawa M, Mizuno Y, Shimasaki Y, Nakayama M, Yasue H, Kuwahara K, Saito Y, Nakao K (1999) Effect of interleukin-1 beta on cardiac hypertrophy and production of natriuretic peptides in rat cardiocyte culture. J Mol Cell Cardiol 31:1997–2006

    Article  PubMed  CAS  Google Scholar 

  141. Nishikawa K, Yoshida M, Kusuhara M, Ishigami N, Isoda K, Miyazaki K, Ohsuzu F (2006) Left ventricular hypertrophy in mice with a cardiac-specific overexpression of interleukin-1. Am J Physiol Heart Circ Physiol 291:H176–H183

    Article  PubMed  CAS  Google Scholar 

  142. Kumar A, Thota V, Dee L, Olson J, Uretz E, Parrillo JE (1996) Tumor necrosis factor alpha and interleukin 1beta are responsible for in vitro myocardial cell depression induced by human septic shock serum. J Exp Med 183:949–958

    Article  PubMed  CAS  Google Scholar 

  143. Prabhu SD (2004) Cytokine-induced modulation of cardiac function. Circ Res 95:1140–1153

    Article  PubMed  CAS  Google Scholar 

  144. Balligand JL, Ungureanu-Longrois D, Simmons WW, Pimental D, Malinski TA, Kapturczak M, Taha Z, Lowenstein CJ, Davidoff AJ, Kelly RA et al (1994) Cytokine-inducible nitric oxide synthase (iNOS) expression in cardiac myocytes. Characterization and regulation of iNOS expression and detection of iNOS activity in single cardiac myocytes in vitro. J Biol Chem 269:27580–27588

    PubMed  CAS  Google Scholar 

  145. Tsujino M, Hirata Y, Imai T, Kanno K, Eguchi S, Ito H, Marumo F (1994) Induction of nitric oxide synthase gene by interleukin-1 beta in cultured rat cardiocytes. Circulation 90:375–383

    PubMed  CAS  Google Scholar 

  146. Oddis CV, Finkel MS (1995) Cytokine-stimulated nitric oxide production inhibits mitochondrial activity in cardiac myocytes. Biochem Biophys Res Commun 213:1002–1009

    Article  PubMed  CAS  Google Scholar 

  147. Tatsumi T, Matoba S, Kawahara A, Keira N, Shiraishi J, Akashi K, Kobara M, Tanaka T, Katamura M, Nakagawa C, Ohta B, Shirayama T, Takeda K, Asayama J, Fliss H, Nakagawa M (2000) Cytokine-induced nitric oxide production inhibits mitochondrial energy production and impairs contractile function in rat cardiac myocytes. J Am Coll Cardiol 35:1338–1346

    Article  PubMed  CAS  Google Scholar 

  148. Panas D, Khadour FH, Szabo C, Schulz R (1998) Proinflammatory cytokines depress cardiac efficiency by a nitric oxide-dependent mechanism. Am J Physiol 275:H1016–H1023

    PubMed  CAS  Google Scholar 

  149. Wang D, McMillin JB, Bick R, Buja LM (1996) Response of the neonatal rat cardiomyocyte in culture to energy depletion: effects of cytokines, nitric oxide, and heat shock proteins. Lab Invest 75:809–818

    PubMed  CAS  Google Scholar 

  150. Combes A, Frye CS, Lemster BH, Brooks SS, Watkins SC, Feldman AM, McTiernan CF (2002) Chronic exposure to interleukin 1beta induces a delayed and reversible alteration in excitation-contraction coupling of cultured cardiomyocytes. Pflugers Arch 445:246–256

    Article  PubMed  CAS  Google Scholar 

  151. McTiernan CF, Lemster BH, Frye C, Brooks S, Combes A, Feldman AM (1997) Interleukin-1 beta inhibits phospholamban gene expression in cultured cardiomyocytes. Circ Res 81:493–503

    PubMed  CAS  Google Scholar 

  152. Frank KF, Bolck B, Erdmann E, Schwinger RH (2003) Sarcoplasmic reticulum Ca2+-ATPase modulates cardiac contraction and relaxation. Cardiovasc Res 57:20–27

    Article  PubMed  CAS  Google Scholar 

  153. Haghighi K, Gregory KN, Kranias EG (2004) Sarcoplasmic reticulum Ca-ATPase-phospholamban interactions and dilated cardiomyopathy. Biochem Biophys Res Commun 322:1214–1222

    Article  PubMed  CAS  Google Scholar 

  154. Miyamoto MI, del Monte F, Schmidt U, DiSalvo TS, Kang ZB, Matsui T, Guerrero JL, Gwathmey JK, Rosenzweig A, Hajjar RJ (2000) Adenoviral gene transfer of SERCA2a improves left-ventricular function in aortic-banded rats in transition to heart failure. Proc Natl Acad Sci USA 97:793–798

    Article  PubMed  CAS  Google Scholar 

  155. del Monte F, Williams E, Lebeche D, Schmidt U, Rosenzweig A, Gwathmey JK, Lewandowski ED, Hajjar RJ (2001) Improvement in survival and cardiac metabolism after gene transfer of sarcoplasmic reticulum Ca(2+)-ATPase in a rat model of heart failure. Circulation 104:1424–1429

    Article  PubMed  CAS  Google Scholar 

  156. Ing DJ, Zang J, Dzau VJ, Webster KA, Bishopric NH (1999) Modulation of cytokine-induced cardiac myocyte apoptosis by nitric oxide, Bak, and Bcl-x. Circ Res 84:21–33

    PubMed  CAS  Google Scholar 

  157. Pinsky DJ, Cai B, Yang X, Rodriguez C, Sciacca RR, Cannon PJ (1995) The lethal effects of cytokine-induced nitric oxide on cardiac myocytes are blocked by nitric oxide synthase antagonism or transforming growth factor beta. J Clin Invest 95:677–685

    Article  PubMed  CAS  Google Scholar 

  158. Suzuki K, Murtuza B, Smolenski RT, Sammut IA, Suzuki N, Kaneda Y, Yacoub MH (2001) Overexpression of interleukin-1 receptor antagonist provides cardioprotection against ischemia-reperfusion injury associated with reduction in apoptosis. Circulation 104:I308–I313

    Article  PubMed  CAS  Google Scholar 

  159. Li YJ, Ding WH, Gao W, Huo Y, Hong T, Zhu RY, Ma DL (2004) The protective effect of interleukin-1 receptor antagonist on postischemic reperfused myocardium and its possible mechanism. Zhonghua Yi Xue Za Zhi 84:548–553

    PubMed  CAS  Google Scholar 

  160. Abbate A, Salloum FN, Vecile E, Das A, Hoke NN, Straino S, Biondi-Zoccai GG, Houser JE, Qureshi IZ, Ownby ED, Gustini E, Biasucci LM, Severino A, Capogrossi MC, Vetrovec GW, Crea F, Baldi A, Kukreja RC, Dobrina A (2008) Anakinra, a recombinant human interleukin-1 receptor antagonist, inhibits apoptosis in experimental acute myocardial infarction. Circulation 117:2670–2683

    Article  PubMed  CAS  Google Scholar 

  161. Brown RD, Jones GM, Laird RE, Hudson P, Long CS (2007) Cytokines regulate matrix metalloproteinases and migration in cardiac fibroblasts. Biochem Biophys Res Commun 362:200–205

    Article  PubMed  CAS  Google Scholar 

  162. Guo XG, Uzui H, Mizuguchi T, Ueda T, Chen JZ, Lee JD (2008) Imidaprilat inhibits matrix metalloproteinase-2 activity in human cardiac fibroblasts induced by interleukin-1beta via NO-dependent pathway. Int J Cardiol 126:414–420

    Article  PubMed  Google Scholar 

  163. Xie Z, Singh M, Singh K (2004) Differential regulation of matrix metalloproteinase-2 and -9 expression and activity in adult rat cardiac fibroblasts in response to interleukin-1beta. J Biol Chem 279:39513–39519

    Article  PubMed  CAS  Google Scholar 

  164. Kida Y, Kobayashi M, Suzuki T, Takeshita A, Okamatsu Y, Hanazawa S, Yasui T, Hasegawa K (2005) Interleukin-1 stimulates cytokines, prostaglandin E2 and matrix metalloproteinase-1 production via activation of MAPK/AP-1 and NF-kappaB in human gingival fibroblasts. Cytokine 29:159–168

    Article  PubMed  CAS  Google Scholar 

  165. Akira S (2000) The role of IL-18 in innate immunity. Curr Opin Immunol 12:59–63

    Article  PubMed  CAS  Google Scholar 

  166. Nakanishi K, Yoshimoto T, Tsutsui H, Okamura H (2001) Interleukin-18 regulates both Th1 and Th2 responses. Annu Rev Immunol 19:423–474

    Article  PubMed  CAS  Google Scholar 

  167. Woldbaek PR, Tonnessen T, Henriksen UL, Florholmen G, Lunde PK, Lyberg T, Christensen G (2003) Increased cardiac IL-18 mRNA, pro-IL-18 and plasma IL-18 after myocardial infarction in the mouse: a potential role in cardiac dysfunction. Cardiovasc Res 59:122–131

    Article  PubMed  CAS  Google Scholar 

  168. Raeburn CD, Dinarello CA, Zimmerman MA, Calkins CM, Pomerantz BJ, McIntyre RC Jr, Harken AH, Meng X (2002) Neutralization of IL-18 attenuates lipopolysaccharide-induced myocardial dysfunction. Am J Physiol Heart Circ Physiol 283:H650–H657

    PubMed  CAS  Google Scholar 

  169. Chandrasekar B, Mummidi S, Claycomb WC, Mestril R, Nemer M (2005) Interleukin-18 is a pro-hypertrophic cytokine that acts through a phosphatidylinositol 3-kinase-phosphoinositide-dependent kinase-1-Akt-GATA4 signaling pathway in cardiomyocytes. J Biol Chem 280:4553–4567

    Article  PubMed  CAS  Google Scholar 

  170. Colston JT, Boylston WH, Feldman MD, Jenkinson CP, de la Rosa SD, Barton A, Trevino RJ, Freeman GL, Chandrasekar B (2007) Interleukin-18 knockout mice display maladaptive cardiac hypertrophy in response to pressure overload. Biochem Biophys Res Commun 354:552–558

    Article  PubMed  CAS  Google Scholar 

  171. Shioi T, Kang PM, Douglas PS, Hampe J, Yballe CM, Lawitts J, Cantley LC, Izumo S (2000) The conserved phosphoinositide 3-kinase pathway determines heart size in mice. EMBO J 19:2537–2548

    Article  PubMed  CAS  Google Scholar 

  172. Condorelli G, Drusco A, Stassi G, Bellacosa A, Roncarati R, Iaccarino G, Russo MA, Gu Y, Dalton N, Chung C, Latronico MV, Napoli C, Sadoshima J, Croce CM, Ross J Jr (2002) Akt induces enhanced myocardial contractility and cell size in vivo in transgenic mice. Proc Natl Acad Sci USA 99:12333–12338

    Article  PubMed  CAS  Google Scholar 

  173. Purcell NH, Tang G, Yu C, Mercurio F, DiDonato JA, Lin A (2001) Activation of NF-kappa B is required for hypertrophic growth of primary rat neonatal ventricular cardiomyocytes. Proc Natl Acad Sci USA 98:6668–6673

    Article  PubMed  CAS  Google Scholar 

  174. Nemoto S, Sheng Z, Lin A (1998) Opposing effects of Jun kinase and p38 mitogen-activated protein kinases on cardiomyocyte hypertrophy. Mol Cell Biol 18:3518–3526

    PubMed  CAS  Google Scholar 

  175. Bueno OF, De Windt LJ, Tymitz KM, Witt SA, Kimball TR, Klevitsky R, Hewett TE, Jones SP, Lefer DJ, Peng CF, Kitsis RN, Molkentin JD (2000) The MEK1-ERK1/2 signaling pathway promotes compensated cardiac hypertrophy in transgenic mice. EMBO J 19:6341–6350

    Article  PubMed  CAS  Google Scholar 

  176. Zechner D, Thuerauf DJ, Hanford DS, McDonough PM, Glembotski CC (1997) A role for the p38 mitogen-activated protein kinase pathway in myocardial cell growth, sarcomeric organization, and cardiac-specific gene expression. J Cell Biol 139:115–127

    Article  PubMed  CAS  Google Scholar 

  177. Woldbaek PR, Sande JB, Stromme TA, Lunde PK, Djurovic S, Lyberg T, Christensen G, Tonnessen T (2005) Daily administration of interleukin-18 causes myocardial dysfunction in healthy mice. Am J Physiol Heart Circ Physiol 289:H708–H714

    Article  PubMed  CAS  Google Scholar 

  178. Platis A, Yu Q, Moore D, Khojeini E, Tsau P, Larson D (2008) The effect of daily administration of IL-18 on cardiac structure and function. Perfusion 23:237–242

    Article  PubMed  CAS  Google Scholar 

  179. Netea MG, Kullberg BJ, Verschueren I, Van Der Meer JW (2000) Interleukin-18 induces production of proinflammatory cytokines in mice: no intermediate role for the cytokines of the tumor necrosis factor family and interleukin-1beta. Eur J Immunol 30:3057–3060

    Article  PubMed  CAS  Google Scholar 

  180. Puren AJ, Fantuzzi G, Gu Y, Su MS, Dinarello CA (1998) Interleukin-18 (IFNgamma-inducing factor) induces IL-8 and IL-1beta via TNFalpha production from non-CD14+ human blood mononuclear cells. J Clin Invest 101:711–721

    Article  PubMed  CAS  Google Scholar 

  181. Olee T, Hashimoto S, Quach J, Lotz M (1999) IL-18 is produced by articular chondrocytes and induces proinflammatory and catabolic responses. J Immunol 162:1096–1100

    PubMed  CAS  Google Scholar 

  182. Okamura H, Tsutsi H, Komatsu T, Yutsudo M, Hakura A, Tanimoto T, Torigoe K, Okura T, Nukada Y, Hattori K et al (1995) Cloning of a new cytokine that induces IFN-gamma production by T cells. Nature 378:88–91

    Article  PubMed  CAS  Google Scholar 

  183. Ueno N, Kashiwamura S, Ueda H, Okamura H, Tsuji NM, Hosohara K, Kotani J, Marukawa S (2005) Role of interleukin 18 in nitric oxide production and pancreatic damage during acute pancreatitis. Shock 24:564–570

    Article  PubMed  CAS  Google Scholar 

  184. Morel JC, Park CC, Woods JM, Koch AE (2001) A novel role for interleukin-18 in adhesion molecule induction through NF kappa B and phosphatidylinositol (PI) 3-kinase-dependent signal transduction pathways. J Biol Chem 276:37069–37075

    Article  PubMed  CAS  Google Scholar 

  185. Morel JC, Park CC, Zhu K, Kumar P, Ruth JH, Koch AE (2002) Signal transduction pathways involved in rheumatoid arthritis synovial fibroblast interleukin-18-induced vascular cell adhesion molecule-1 expression. J Biol Chem 277:34679–34691

    Article  PubMed  CAS  Google Scholar 

  186. Leung BP, Culshaw S, Gracie JA, Hunter D, Canetti CA, Campbell C, Cunha F, Liew FY, McInnes IB (2001) A role for IL-18 in neutrophil activation. J Immunol 167:2879–2886

    PubMed  CAS  Google Scholar 

  187. Dao T, Ohashi K, Kayano T, Kurimoto M, Okamura H (1996) Interferon-gamma-inducing factor, a novel cytokine, enhances Fas ligand-mediated cytotoxicity of murine T helper 1 cells. Cell Immunol 173:230–235

    Article  PubMed  CAS  Google Scholar 

  188. Dao T, Mehal WZ, Crispe IN (1998) IL-18 augments perforin-dependent cytotoxicity of liver NK-T cells. J Immunol 161:2217–2222

    PubMed  CAS  Google Scholar 

  189. Keira N, Tatsumi T, Matoba S, Shiraishi J, Yamanaka S, Akashi K, Kobara M, Asayama J, Fushiki S, Fliss H, Nakagawa M (2002) Lethal effect of cytokine-induced nitric oxide and peroxynitrite on cultured rat cardiac myocytes. J Mol Cell Cardiol 34:583–596

    Article  PubMed  CAS  Google Scholar 

  190. Chandrasekar B, Vemula K, Surabhi RM, Li-Weber M, Owen-Schaub LB, Jensen LE, Mummidi S (2004) Activation of intrinsic and extrinsic proapoptotic signaling pathways in interleukin-18-mediated human cardiac endothelial cell death. J Biol Chem 279:20221–20233

    Article  PubMed  CAS  Google Scholar 

  191. Marino E, Cardier JE (2003) Differential effect of IL-18 on endothelial cell apoptosis mediated by TNF-alpha and Fas (CD95). Cytokine 22:142–148

    Article  PubMed  CAS  Google Scholar 

  192. Tsutsui H, Nakanishi K, Matsui K, Higashino K, Okamura H, Miyazawa Y, Kaneda K (1996) IFN-gamma-inducing factor up-regulates Fas ligand-mediated cytotoxic activity of murine natural killer cell clones. J Immunol 157:3967–3973

    PubMed  CAS  Google Scholar 

  193. Chandrasekar B, Valente AJ, Freeman GL, Mahimainathan L, Mummidi S (2006) Interleukin-18 induces human cardiac endothelial cell death via a novel signaling pathway involving NF-kappaB-dependent PTEN activation. Biochem Biophys Res Commun 339:956–963

    Article  PubMed  CAS  Google Scholar 

  194. Stambolic V, Suzuki A, de la Pompa JL, Brothers GM, Mirtsos C, Sasaki T, Ruland J, Penninger JM, Siderovski DP, Mak TW (1998) Negative regulation of PKB/Akt-dependent cell survival by the tumor suppressor PTEN. Cell 95:29–39

    Article  PubMed  CAS  Google Scholar 

  195. Datta SR, Brunet A, Greenberg ME (1999) Cellular survival: a play in three Akts. Genes Dev 13:2905–2927

    Article  PubMed  CAS  Google Scholar 

  196. Zha J, Harada H, Yang E, Jockel J, Korsmeyer SJ (1996) Serine phosphorylation of death agonist BAD in response to survival factor results in binding to 14–3-3 not BCL-X(L). Cell 87:619–628

    Article  PubMed  CAS  Google Scholar 

  197. Yang E, Zha J, Jockel J, Boise LH, Thompson CB, Korsmeyer SJ (1995) Bad, a heterodimeric partner for Bcl-XL and Bcl-2, displaces Bax and promotes cell death. Cell 80:285–291

    Article  PubMed  CAS  Google Scholar 

  198. Reddy VS, Harskamp RE, van Ginkel MW, Calhoon J, Baisden CE, Kim IS, Valente AJ, Chandrasekar B (2008) Interleukin-18 stimulates fibronectin expression in primary human cardiac fibroblasts via PI3K-Akt-dependent NF-kappaB activation. J Cell Physiol 215:697–707

    Article  PubMed  CAS  Google Scholar 

  199. Yu Q, Vazquez R, Khojeini EV, Patel C, Venkataramani R, Larson DF (2009) IL-18 induction of osteopontin mediates cardiac fibrosis and diastolic dysfunction in mice. Am J Physiol Heart Circ Physiol 297:H76–H85

    Article  PubMed  CAS  Google Scholar 

  200. Redfield MM, Jacobsen SJ, Burnett JC Jr, Mahoney DW, Bailey KR, Rodeheffer RJ (2003) Burden of systolic and diastolic ventricular dysfunction in the community: appreciating the scope of the heart failure epidemic. JAMA 289:194–202

    Article  PubMed  Google Scholar 

  201. Lloyd-Jones DM, Larson MG, Leip EP, Beiser A, D’Agostino RB, Kannel WB, Murabito JM, Vasan RS, Benjamin EJ, Levy D (2002) Lifetime risk for developing congestive heart failure: the Framingham Heart Study. Circulation 106:3068–3072

    Article  PubMed  Google Scholar 

  202. Schocken DD, Benjamin EJ, Fonarow GC, Krumholz HM, Levy D, Mensah GA, Narula J, Shor ES, Shor ES, Young JB, Hong Y (2008) Prevention of heart failure: a scientific statement from the American Heart Association Councils on Epidemiology and Prevention, Clinical Cardiology, Cardiovascular Nursing, and High Blood Pressure Research; Quality of Care and Outcomes Research Interdisciplinary Working Group; and Functional Genomics and Translational Biology Interdisciplinary Working Group. Circulation 117:2544–2565

    Article  PubMed  Google Scholar 

  203. Chung ES, Packer M, Lo KH, Fasanmade AA, Willerson JT (2003) Randomized, double-blind, placebo-controlled, pilot trial of infliximab, a chimeric monoclonal antibody to tumor necrosis factor-alpha, in patients with moderate-to-severe heart failure: results of the anti-TNF Therapy Against Congestive Heart Failure (ATTACH) trial. Circulation 107:3133–3140

    Article  PubMed  CAS  Google Scholar 

  204. Mann DL, McMurray JJ, Packer M, Swedberg K, Borer JS, Colucci WS, Djian J, Drexler H, Feldman A, Kober L, Krum H, Liu P, Nieminen M, Tavazzi L, van Veldhuisen DJ, Waldenstrom A, Warren M, Westheim A, Zannad F, Fleming T (2004) Targeted anticytokine therapy in patients with chronic heart failure: results of the Randomized Etanercept Worldwide Evaluation (RENEWAL). Circulation 109:1594–1602

    Article  PubMed  CAS  Google Scholar 

  205. Heymans S, Hirsch E, Anker SD, Aukrust P, Balligand JL, Cohen-Tervaert JW, Drexler H, Filippatos G, Felix SB, Gullestad L, Hilfiker-Kleiner D, Janssens S, Latini R, Neubauer G, Paulus WJ, Pieske B, Ponikowski P, Schroen B, Schultheiss HP, Tschope C, Van Bilsen M, Zannad F, McMurray J, Shah AM (2009) Inflammation as a therapeutic target in heart failure? A scientific statement from the Translational Research Committee of the Heart Failure Association of the European Society of Cardiology. Eur J Heart Fail 11:119–129

    Article  PubMed  CAS  Google Scholar 

  206. Scallon BJ, Moore MA, Trinh H, Knight DM, Ghrayeb J (1995) Chimeric anti-TNF-alpha monoclonal antibody cA2 binds recombinant transmembrane TNF-alpha and activates immune effector functions. Cytokine 7:251–259

    Article  PubMed  CAS  Google Scholar 

  207. Lugering A, Schmidt M, Lugering N, Pauels HG, Domschke W, Kucharzik T (2001) Infliximab induces apoptosis in monocytes from patients with chronic active Crohn’s disease by using a caspase-dependent pathway. Gastroenterology 121:1145–1157

    Article  PubMed  CAS  Google Scholar 

  208. Torre-Amione G (2005) Immune activation in chronic heart failure. Am J Cardiol 95:3C–8C (discussion 38C–40C)

    Article  PubMed  CAS  Google Scholar 

  209. Celis R, Torre-Martinez G, Torre-Amione G (2008) Evidence for activation of immune system in heart failure: is there a role for anti-inflammatory therapy? Curr Opin Cardiol 23:254–260

    Article  PubMed  Google Scholar 

  210. Torre-Amione G, Anker SD, Bourge RC, Colucci WS, Greenberg BH, Hildebrandt P, Keren A, Motro M, Moye LA, Otterstad JE, Pratt CM, Ponikowski P, Rouleau JL, Sestier F, Winkelmann BR, Young JB (2008) Results of a non-specific immunomodulation therapy in chronic heart failure (ACCLAIM trial): a placebo-controlled randomised trial. Lancet 371:228–236

    Article  PubMed  CAS  Google Scholar 

  211. Gullestad L, Aass H, Fjeld JG, Wikeby L, Andreassen AK, Ihlen H, Simonsen S, Kjekshus J, Nitter-Hauge S, Ueland T, Lien E, Froland SS, Aukrust P (2001) Immunomodulating therapy with intravenous immunoglobulin in patients with chronic heart failure. Circulation 103:220–225

    PubMed  CAS  Google Scholar 

  212. Sliwa K, Woodiwiss A, Candy G, Badenhorst D, Libhaber C, Norton G, Skudicky D, Sareli P (2002) Effects of pentoxifylline on cytokine profiles and left ventricular performance in patients with decompensated congestive heart failure secondary to idiopathic dilated cardiomyopathy. Am J Cardiol 90:1118–1122

    Article  PubMed  CAS  Google Scholar 

  213. Sliwa K, Woodiwiss A, Kone VN, Candy G, Badenhorst D, Norton G, Zambakides C, Peters F, Essop R (2004) Therapy of ischemic cardiomyopathy with the immunomodulating agent pentoxifylline: results of a randomized study. Circulation 109:750–755

    Article  PubMed  CAS  Google Scholar 

  214. Staudt A, Hummel A, Ruppert J, Dorr M, Trimpert C, Birkenmeier K, Krieg T, Staudt Y, Felix SB (2006) Immunoadsorption in dilated cardiomyopathy: 6-month results from a randomized study. Am Heart J 152:712–716

    Article  PubMed  Google Scholar 

  215. Fairweather D, Afanasyeva M, Rose NR (2004) Cellular immunity: a role for cytokines. In: Doria A, Pauletto P (eds) Handbook of systemic autoimmune diseases: the heart in systemic autoimmune diseases. Elsevier, Amsterdam, pp 3–7

    Google Scholar 

  216. Frangogiannis NG (2006) Targeting the inflammatory response in healing myocardial infarcts. Curr Med Chem 13:1877–1893

    Article  PubMed  CAS  Google Scholar 

  217. Jugdutt BI (2008) Aging and remodeling during healing of the wounded heart: current therapies and novel drug targets. Curr Drug Targets 9:325–344

    Article  PubMed  CAS  Google Scholar 

Download references

Acknowledgments

Noel R. Rose was supported by PHS GRANT R01HL067290. We are grateful to thank Samira Hatami for her valuable assistance in preparing the figures.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Nima Rezaei.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Hedayat, M., Mahmoudi, M.J., Rose, N.R. et al. Proinflammatory cytokines in heart failure: double-edged swords. Heart Fail Rev 15, 543–562 (2010). https://doi.org/10.1007/s10741-010-9168-4

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s10741-010-9168-4

Keywords

Navigation