Abstract

HomeCirculationVol. 119, No. 10Tumor Necrosis Factor-α and Its Receptors 1 and 2 Free AccessEditorialPDF/EPUBAboutView PDFView EPUBSections ToolsAdd to favoritesDownload citationsTrack citationsPermissions ShareShare onFacebookTwitterLinked InMendeleyReddit Jump toFree AccessEditorialPDF/EPUBTumor Necrosis Factor-α and Its Receptors 1 and 2Yin and Yang in Myocardial Infarction? Rainer Schulz, MD and Gerd Heusch, MD Rainer SchulzRainer Schulz From the Institut für Pathophysiologie, Universitätsklinikum Essen, Essen, Germany. Search for more papers by this author and Gerd HeuschGerd Heusch From the Institut für Pathophysiologie, Universitätsklinikum Essen, Essen, Germany. Search for more papers by this author Originally published2 Mar 2009https://doi.org/10.1161/CIRCULATIONAHA.108.846105Circulation. 2009;119:1355–1357Other version(s) of this articleYou are viewing the most recent version of this article. Previous versions: March 2, 2009: Previous Version 1 In healthy hearts, tumor necrosis factor-α (TNF-α) concentration is low, and TNF-α is mainly located in endothelium and resident mast cells.1 TNF-α receptors (TNFRs) 1 (TNFR1) and 2 (TNFR2) are expressed on most cardiac cells, including cardiomyocytes.2 During myocardial ischemia, preformed TNF-α is released within minutes from resident mast cells and macrophages.3 With persistent ischemia, TNF-α also originates from cardiomyocytes.4 In rats after myocardial infarction (MI), TNFR1 density is increased for 10 days, whereas TNFR2 density remains unchanged.5 In contrast, TNFR1 and TNFR2 are both downregulated in the failing heart, whereas soluble TNFRs are increased6 owing to proteolytic cleavage of cardiomyocyte TNFR and release from circulating exosome-like vesicles.7 Increased soluble TNFRs decrease TNF-α bioactivity while at the same time prolonging its half-life.8Article p 1386TNF-α and Myocardial Ischemia/ Reperfusion InjuryTNF-α contributes to both reversible (contractile dysfunction4,9) and irreversible (MI) injury.1 Preischemic treatment with TNF-α antibodies10 or soluble TNFR1,11 permanent TNF-α knockout,12 or knockout of TNFR1 but not TNFR213 all reduce infarct size. The latter finding highlights the functional difference of TNFR1 and TNFR2 activation for ischemia/reperfusion injury: Only TNFR1 signaling is detrimental (Figure). Download figureDownload PowerPointFigure. Signaling cascade activated (→) or inhibited (⊣) by TNFR1 (red) or TNFR2 (blue) activation. sTNFR1 indicates soluble TNFR1; sTNFR2, soluble TNFR2; S1-P, sphingosine-1-phosphate; NF-κB, nuclear factor-κB; SOCS3, suppressor of cytokine signaling 3; ROS, reactive oxygen species; Akt-P, phosphorylated Akt, a homologue of the transforming v-Akt; NADPH-Ox, NADPH oxidase; STAT3-P, phosphorylated signal transducer and activator of transcription; Jnk-P, phosphorylated c-jun N-terminal kinase; p38-P, phosphorylated mitogen-activated protein kinase; Erk-P, phosphorylated extracellular signal-regulated kinase; PKC, protein kinase C; CamK, calcium/calmodulin-dependent kinase; MMP-2/9, matrix metalloproteinases 2 and 9; and IL-6, interleukin 6. Red indicates proteins involved in apoptosis; purple, proteins involved in growth; green, proteins involved in contractile function; blue, protein involved in inflammation; and yellow, proteins involved in extracellular matrix turnover.TNF-α and CardioprotectionAlthough TNF-α aggravates acute ischemia/reperfusion injury, preconditioning with TNF-α reduces infarct size.1 In addition, accumulation of endogenous TNF-α after coronary microembolization reduces infarct size.9 Finally, the protection of delayed ischemic preconditioning is absent after pretreatment with TNF-α antibodies and in TNF-α–, TNFR1-, and TNFR2-knockout mice13; the latter result reflects the involvement of both TNFRs in cardioprotection.13,14TNF-α and Permanent Coronary OcclusionTransfection with soluble TNFR1 reduces infarct size 3 weeks after MI in rats8 but not in mice.15 Neither genetic ablation of TNF-α16 nor knockout of TNFR117 or TNFR218 alters infarct size when measured 1 day17 or several weeks after coronary ligation.16–18 However, infarct size after 1 day is increased with knockout of both TNFRs and is associated with increased cardiomyocyte apoptosis in the area at risk.19With permanent coronary occlusion, 40% to 70% of mice die within 10 days after MI,15–18,20 and >50% of them die of cardiac rupture.15,16,18 Reduction of TNF-α bioactivity by adenoviral transfection with soluble TNFR1 promotes rather than attenuates cardiac rupture by inhibiting reactive fibrosis and reduces survival within 7 days after MI in male mice.15 In contrast, genetic ablation of TNF-α16 or TNFR1 knockout17,18 increases post-MI survival, without reducing the rate of cardiac rupture.18 In contrast to TNFR knockout, matrix metalloproteinase-2 knockout reduces cardiac rupture and increases survival after MI.20Elevated TNF-α contributes to chronic left ventricular (LV) dysfunction after MI through a local inflammatory response,16 increased matrix metalloproteinase-2 activity, matrix and collagen degradation,16,20 and cardiomyocyte apoptosis.16 TNF-α inhibition with etanercept immediately after MI reduces leukocyte infiltration and extracellular matrix degradation and preserves cardiac function 6 weeks after MI in rats.21 Similarly, TNF-α blockade with soluble TNFR2 attenuates hemodynamic derangements in the first week after MI in rats but does not attenuate LV dilatation 10 weeks after MI.22 In contrast, adenoviral transfection with soluble TNFR1 starting 7 days after MI exacerbates LV dysfunction and remodeling.15TNFR1 knockout attenuates contractile dysfunction after MI,17,18 whereas TNFR2 knockout exaggerates ventricular dilatation and dysfunction. Upregulated transcript levels of inflammatory markers after MI are reduced in TNFR1-knockout mice but further enhanced in TNFR2-knockout mice. Thus, TNF-α worsens LV remodeling via TNFR1 and improves it via TNFR2 after MI.18Hamid and coworkers in their elegant study in the present issue of Circulation now further delineate the ambivalent role of TNFR1 and TNFR2 in post-MI remodeling in male mice.23 Knockout of TNFR1 improved LV ejection fraction; reduced LV dilatation, cardiomyocyte hypertrophy and apoptosis, fibrosis, and inflammation; and ultimately resulted in improved survival after MI. In contrast, knockout of TNFR2 increased LV dilatation, cardiomyocyte hypertrophy and apoptosis, and fibrosis in the remote myocardium but did not affect systolic LV function. However, survival was also increased in TNFR2-knockout mice, and both TNFR-knockout mice had less reactive oxygen species formation and improved diastolic LV pressure. Importantly, Hamid and coworkers23 analyzed the signaling of TNFR1 and TNFR2 activation (Figure) and highlighted the divergent effects of TNFR1 and TNFR2 activation on stress kinase activation, nuclear signaling, and cardiomyocyte death pathways.Some questions remain: Is blockade of TNF-α bioactivity beneficial or deleterious? Increased TNF-α induces contractile dysfunction1 up to heart failure,24,25 and TNFR1 activation is primarily responsible for these detrimental effects.25 However, basal TNF-α improves contractile function via TNFR1 activation,26 and endogenous TNF-α contributes to the protection afforded by preconditioning through both TNFRs13; in addition, elimination of both TNFRs increases cardiomyocyte apoptosis.19 Thus, apart from signaling through TNFR1 or TNFR2, TNF-α can exert both beneficial and deleterious actions, depending on its concentration and duration of exposure.Are time and localization of increased TNF-α important? TNF-α remains elevated for weeks in the infarcted area, whereas it decreases in the peri-infarct area 1 week after MI and remains only slightly above basal values.5 Contractile dysfunction and LV dilatation coincide with the peak increase of TNF-α after MI.22 TNF-α in the infarcted area contributes to cardiomyocyte apoptosis, whereas TNF-α in the peri-infarct area might stimulate fibroblasts,27 thereby stabilize the infarcted area, and attract stem cells28 for cardiac repair and suppression of inflammation.29 Elevated TNF-α in the peri-infarct area depresses contractile function, induces cardiomyocyte apoptosis, and triggers fibrosis; however, when subsequently reduced again, TNF-α in the peri-infarct area might contribute to preservation of contractile function.26What is the contribution of cardiac rupture and arrhythmias to the impact of TNF-α on mortality? Hamid and coworkers23 suggest that less cardiac rupture was responsible for improved survival in TNFR knockouts. However, neither TNFR1 nor TNFR2 knockout affects the rate of cardiac rupture after MI,18 and blockade of TNF-α bioactivity by adenoviral transfer of soluble TNFR1 even increases the rate of cardiac rupture.15 An alternative explanation for the improved survival after MI in TNFR knockouts relates to a reduced incidence of arrhythmias. In post-MI rats, ventricular arrhythmias coincide with the secretion of TNF-α,30 and TNF-α promotes action potential prolongation through formation of reactive oxygen species in rat ventricular myocytes.31 Thus, diminished reactive oxygen species formation in TNFR knockouts, as shown by Hamid and coworkers,23 could restore normal repolarization and reduce arrhythmias. ECG tracings of TNFR1 and TNFR2 knockouts after MI could help identify the underlying cause.Are gender and age important for the actions of TNF-α? The expression of myocardial TNFR is higher in male than in female hearts, and such differential expression may contribute to sex differences in the severity of congestive heart failure and mortality.2,17 Indeed, TNF-α infusion depresses contractile function in male but not female mouse hearts.32 TNF-α–overexpressing mice develop progressive cardiomyocyte hypertrophy and LV dilatation, as well as increased collagen synthesis and deposition, along with marked diastolic dysfunction. Anti-TNF-α treatment prevents changes in extracellular matrix and preserves diastolic function, as did TNFR1 and TNFR2 knockout in the present study; however, this beneficial effect is only observed in young and not in old mice,24 which suggests a sex- and age-dependent signaling cascade after TNFR activation.ConclusionsIndeed, TNF-α and TNFR activation have an ambivalent role in MI. Excessive TNF-α expression and subsequent cardiomyocyte TNFR1 activation are detrimental, whereas a lower TNF-α concentration and subsequent cardiomyocyte TNFR2 activation are protective. TNF-α–induced fibrosis and attraction of stem cells might stabilize the infarcted area, while contributing to structural remodeling and diastolic dysfunction in remote myocardium. Apart from its receptor subtype, the action of TNF-α depends on its concentration, duration of exposure, and localization, as well as on the sex and age of the affected individual. Therefore, it is not surprising that clinical trials in chronic heart failure patients using compounds that antagonize TNF-α, including the TNF-α antibody infliximab and the soluble recombinant TNF-α receptor etanercept, revealed disappointing results, which supports the ambivalent effect of TNF-α.33The opinions expressed in this article are not necessarily those of the editors or of the American Heart Association.DisclosuresNone.FootnotesCorrespondence to Professor Dr Rainer Schulz, Institut für Pathophysiologie, Universitätsklinikum Essen, Hufelandstrasse 55, 45122 Essen, Germany. E-mail [email protected] References 1 Schulz R. TNFα in myocardial ischemia/reperfusion: damage vs. protection. J Mol Cell Cardiol. 2008; 45: 712–714.CrossrefMedlineGoogle Scholar2 Kadokami T, McTiernan CF, Kubota T, Frye CS, Feldman AM. Sex-related survival differences in murine cardiomyopathy are associated with differences in TNF-receptor expression. J Clin Invest. 2000; 106: 589–597.CrossrefMedlineGoogle Scholar3 Gilles S, Zahler S, Welsch U, Sommerhoff CP, Becker BF. Release of TNF-alpha during myocardial reperfusion depends on oxidative stress and is prevented by mast cell stabilizers. Cardiovasc Res. 2003; 60: 608–616.CrossrefMedlineGoogle Scholar4 Dorge H, Schulz R, Belosjorow S, Post H, van de Sand A, Konietzka I, Frede S, Hartung T, Vinten-Johansen J, Youker KA, Entman ML, Erbel R, Heusch G. Coronary microembolization: the role of TNF-alpha in contractile dysfunction. J Mol Cell Cardiol. 2002; 34: 51–62.CrossrefMedlineGoogle Scholar5 Irwin MW, Mak S, Mann DL, Qu R, Penninger JM, Yan A, Dawood F, Wen WH, Shou Z, Liu P. Tissue expression and immunolocalization of tumor necrosis factor-α in postinfarction dysfunctional myocardium. Circulation. 1999; 99: 1492–1498.CrossrefMedlineGoogle Scholar6 Torre-Amione G, Kapadia S, Lee J, Durand JB, Bies RD, Young JB, Mann DL. Tumor necrosis factor-α and tumor necrosis factor receptors in the failing human heart. Circulation. 1996; 93: 704–711.CrossrefMedlineGoogle Scholar7 Zhang J, Hawari FI, Shamburek RD, Adamik B, Kaler M, Islam A, Liao DW, Rouhani FN, Ingham M, Levine SJ. Circulating TNFR1 exosome-like vesicles partition with the LDL fraction of human plasma. Biochem Biophys Res Commun. 2008; 366: 579–584.CrossrefMedlineGoogle Scholar8 Sugano M, Tsuchida K, Hata T, Makino N. In vivo transfer of soluble TNF-alpha receptor 1 gene improves cardiac function and reduces infarct size after myocardial infarction in rats. FASEB J. 2004; 18: 911–913.CrossrefMedlineGoogle Scholar9 Skyschally A, Gres P, Hoffmann S, Haude M, Erbel R, Schulz R, Heusch G. Bidirectional role of tumor necrosis factor-alpha in coronary microembolization: progressive contractile dysfunction versus delayed protection against infarction. Circ Res. 2007; 100: 140–146.LinkGoogle Scholar10 Belosjorow S, Bolle I, Duschin A, Heusch G, Schulz R. TNF-alpha antibodies are as effective as ischemic preconditioning in reducing infarct size in rabbits. Am J Physiol Heart Circ Physiol. 2003; 284: H927–H930.CrossrefMedlineGoogle Scholar11 Sugano M, Hata T, Tsuchida K, Suematsu N, Oyama J, Satoh S, Makino N. Local delivery of soluble TNF-alpha receptor 1 gene reduces infarct size following ischemia/reperfusion injury in rats. Mol Cell Biochem. 2004; 266: 127–132.CrossrefMedlineGoogle Scholar12 Maekawa N, Wada H, Kanda T, Niwa T, Yamada Y, Saito K, Fujiwara H, Sekikawa K, Seishima M. Improved myocardial ischemia/reperfusion injury in mice lacking tumor necrosis factor-alpha. J Am Coll Cardiol. 2002; 39: 1229–1235.CrossrefMedlineGoogle Scholar13 Flaherty MP, Guo Y, Tiwari S, Rezazadeh A, Hunt G, Sanganalmath SK, Tang XL, Bolli R, Dawn B. The role of TNF-alpha receptors p55 and p75 in acute myocardial ischemia/reperfusion injury and late preconditioning. J Mol Cell Cardiol. 2008; 45: 735–741.CrossrefMedlineGoogle Scholar14 Heusch G, Boengler K, Schulz R. Cardioprotection: nitric oxide, protein kinases, and mitochondria. Circulation. 2008; 118: 1915–1919.LinkGoogle Scholar15 Monden Y, Kubota T, Tsutsumi T, Inoue T, Kawano S, Kawamura N, Ide T, Egashira K, Tsutsui H, Sunagawa K. Soluble TNF receptors prevent apoptosis in infiltrating cells and promote ventricular rupture and remodeling after myocardial infarction. Cardiovasc Res. 2007; 73: 794–805.CrossrefMedlineGoogle Scholar16 Sun M, Dawood F, Wen WH, Chen M, Dixon I, Kirshenbaum LA, Liu PP. Excessive tumor necrosis factor activation after infarction contributes to susceptibility of myocardial rupture and left ventricular dysfunction. Circulation. 2004; 110: 3221–3228.LinkGoogle Scholar17 Ramani R, Mathier M, Wang P, Gibson G, Tögel S, Dawson J, Bauer A, Alber S, Watkins SC, McTiernan CF, Feldman AM. Inhibition of tumor necrosis factor receptor-1-mediated pathways has beneficial effects in a murine model of postischemic remodeling. Am J Physiol Heart Circ Physiol. 2004; 287: H1369–H1377.CrossrefMedlineGoogle Scholar18 Monden Y, Kubota T, Inoue T, Tsutsumi T, Kawano S, Ide T, Tsutsui H, Sunagawa K. 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. 2007; 293: H743–H753.CrossrefMedlineGoogle Scholar19 Kurrelmeyer KM, Michael LH, Baumgarten G, Taffet GE, Peschon JJ, Sivasubramanian N, Entman ML, Mann DL. Endogenous tumor necrosis factor protects the adult cardiac myocyte against ischemic-induced apoptosis in a murine model of acute myocardial infarction. Proc Natl Acad Sci U S A. 2000; 97: 5456–5461.CrossrefMedlineGoogle Scholar20 Hayashidani S, Tsutsui H, Ikeuchi M, Shiomi T, Matsusaka H, Kubota T, Imanaka-Yoshida K, Itoh T, Takeshita A. Targeted deletion of MMP-2 attenuates early LV rupture and late remodeling after experimental myocardial infarction. Am J Physiol Heart Circ Physiol. 2003; 285: H1229–H1235.CrossrefMedlineGoogle Scholar21 Berry MF, Woo YJ, Pirolli TJ, Bish LT, Moise MA, Burdick JW, Morine KJ, Jayasankar V, Gardner TJ, Sweeney HL. Administration of a tumor necrosis factor inhibitor at the time of myocardial infarction attenuates subsequent ventricular remodeling. J Heart Lung Transplant. 2004; 23: 1061–1068.CrossrefMedlineGoogle Scholar22 Berthonneche C, Sulpice T, Boucher F, Gouraud L, de Leiris J, O'Connor SE, Herbert JM, Janiak P. New insights into the pathological role of TNF-alpha in early cardiac dysfunction and subsequent heart failure after infarction in rats. Am J Physiol Heart Circ Physiol. 2004; 287: H340–H350.CrossrefMedlineGoogle Scholar23 Hamid T, Gu Y, Ortines RV, Bhattacharya C, Wang G, Xuan Y-T, Prabhu SD. Divergent tumor necrosis factor receptor–related remodeling responses in heart failure: role of nuclear factor-κB and inflammatory activation. Circulation. 2009; 119: 1386–1397.LinkGoogle Scholar24 Li YY, Feng YQ, Kadokami T, McTiernan CF, Draviam R, Watkins SC, Feldman AM. 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 U S A. 2000; 97: 12746–12751.CrossrefMedlineGoogle Scholar25 Higuchi Y, McTiernan CF, Frye CB, McGowan BS, Chan TO, Feldman AM. Tumor necrosis factor receptors 1 and 2 differentially regulate survival, cardiac dysfunction, and remodeling in transgenic mice with tumor necrosis factor-α–induced cardiomyopathy. Circulation. 2004; 109: 1892–1897.LinkGoogle Scholar26 Pinz I, Wax SD, Anderson P, Ingwall JS. Low over-expression of TNFα in the mouse heart increases contractile performance via TNFR1. J Cell Biochem. 2008; 105: 99–107.CrossrefMedlineGoogle Scholar27 Gurantz D, Cowling RT, Varki N, Frikovsky E, Moore CD, Greenberg BH. IL-1β and TNF-α upregulate angiotensin II type 1 (AT1) receptors on cardiac fibroblasts and are associated with increased AT1 density in the post-MI heart. J Mol Cell Cardiol. 2005; 38: 505–515.CrossrefMedlineGoogle Scholar28 Chen Y, Ke Q, Yang Y, Rana JS, Tang J, Morgan JP, Xiao YF. Cardiomyocytes overexpressing TNF-alpha attract migration of embryonic stem cells via activation of p38 and c-Jun amino-terminal kinase. FASEB J. 2003; 17: 2231–2239.CrossrefMedlineGoogle Scholar29 Bao C, Guo J, Lin G, Hu M, Hu Z. TNFR gene-modified mesenchymal stem cells attenuate inflammation and cardiac dysfunction following MI. Scand Cardiovasc J. 2008; 42: 56–62.CrossrefMedlineGoogle Scholar30 Xiao H, Chen Z, Liao Y, Cheng X, Liu K, Wang Y, Wang M, Guo H. Positive correlation of tumor necrosis factor-alpha early expression in myocardium and ventricular arrhythmias in rats with acute myocardial infarction. Arch Med Res. 2008; 39: 285–291.CrossrefMedlineGoogle Scholar31 Fernandez-Velasco M, Ruiz-Hurtado G, Hurtado O, Moro MA, Delgado C. TNF-alpha downregulates transient outward potassium current in rat ventricular myocytes through iNOS overexpression and oxidant species generation. Am J Physiol Heart Circ Physiol. 2007; 293: H238–H245.CrossrefMedlineGoogle Scholar32 Wang M, Tsai BM, Crisostomo PR, Meldrum DR. Tumor necrosis factor receptor 1 signaling resistance in the female myocardium during ischemia. Circulation. 2006; 114 (suppl I): I-282–I-289.LinkGoogle Scholar33 Mousa SA, Goncharuk O, Miller D. Recent advances of TNF-alpha antagonists in rheumatoid arthritis and chronic heart failure. Expert Opin Biol Ther. 2007; 7: 617–625.CrossrefMedlineGoogle Scholar Previous Back to top Next FiguresReferencesRelatedDetailsCited By Liberale L, Ministrini S, Carbone F, Camici G and Montecucco F (2021) Cytokines as therapeutic targets for cardio- and cerebrovascular diseases, Basic Research in Cardiology, 10.1007/s00395-021-00863-x, 116:1, Online publication date: 1-Dec-2021. Gouweleeuw L, Wajant H, Maier O, Eisel U, Blankesteijn W and Schoemaker R (2021) Effects of selective TNFR1 inhibition or TNFR2 stimulation, compared to non-selective TNF inhibition, on (neuro)inflammation and behavior after myocardial infarction in male mice, Brain, Behavior, and Immunity, 10.1016/j.bbi.2021.01.001, 93, (156-171), Online publication date: 1-Mar-2021. Sheng X, Fan T, Jin X and Huang T (2020) Identification of Key Genes Involved in Acute Myocardial Infarction by Comparative Transcriptome Analysis, BioMed Research International, 10.1155/2020/1470867, 2020, (1-10), Online publication date: 6-Oct-2020. Mendieta G, Ben-Aicha S, Gutiérrez M, Casani L, Aržanauskaitė M, Carreras F, Sabate M, Badimon L and Vilahur G (2020) Intravenous Statin Administration During Myocardial Infarction Compared With Oral Post-Infarct Administration, Journal of the American College of Cardiology, 10.1016/j.jacc.2020.01.042, 75:12, (1386-1402), Online publication date: 1-Mar-2020. Ravi S, Willis M and Schisler J (2019) Fibrotic Signaling in Cardiomyopathies Fibrosis in Disease, 10.1007/978-3-319-98143-7_10, (273-317), . Van Taunay J, Albelda M, Frias J and Lipinski M (2018) Biologics and Cardiovascular Disease, Journal of Cardiovascular Pharmacology, 10.1097/FJC.0000000000000595, 72:2, (77-85), Online publication date: 1-Aug-2018. Hadebe N, Cour M and Lecour S (2018) The SAFE pathway for cardioprotection: is this a promising target?, Basic Research in Cardiology, 10.1007/s00395-018-0670-5, 113:2, Online publication date: 1-Mar-2018. Torrey H, Butterworth J, Mera T, Okubo Y, Wang L, Baum D, Defusco A, Plager S, Warden S, Huang D, Vanamee E, Foster R and Faustman D (2017) Targeting TNFR2 with antagonistic antibodies inhibits proliferation of ovarian cancer cells and tumor-associated T regs , Science Signaling, 10.1126/scisignal.aaf8608, 10:462, Online publication date: 17-Jan-2017. Alánová P, Chytilová A, Neckář J, Hrdlička J, Míčová P, Holzerová K, Hlaváčková M, Macháčková K, Papoušek F, Vašinová J, Benák D, Nováková O and Kolář F (2017) Myocardial ischemic tolerance in rats subjected to endurance exercise training during adaptation to chronic hypoxia, Journal of Applied Physiology, 10.1152/japplphysiol.00671.2016, 122:6, (1452-1461), Online publication date: 1-Jun-2017. Butt N, Bache-Mathiesen L, Nordrehaug J, Tuseth V, Munk P, Bonarjee V, Hall T, Jensen S, Halvorsen S, Firat H, Atar D and Larsen A (2017) Administration of the Mitochondrial Permeability Transition Pore Inhibitor, TRO40303, prior to Primary Percutaneous Coronary Intervention, Does Not Affect the Levels of Pro-Inflammatory Cytokines or Acute-Phase Proteins, Cardiology, 10.1159/000475460, 138:2, (122-132), . Sandoval-Pinto E, Padilla-Gutiérrez J, Valdés-Alvarado E, García-González I, Valdez-Haro A, Muñoz-Valle J, Flores-Salinas H, Brennan-Bourdon L and Valle Y (2016) Association of the −1031T>C polymorphism and soluble TNF-α levels with Acute Coronary Syndrome, Cytokine, 10.1016/j.cyto.2015.11.014, 78, (37-43), Online publication date: 1-Feb-2016. Zheng D, Zhao J, Yang J, Wang M and Zhang X (2016) Enhanced ADAM17 expression is associated with cardiac remodeling in rats with acute myocardial infarction, Life Sciences, 10.1016/j.lfs.2016.02.097, 151, (61-69), Online publication date: 1-Apr-2016. Jin H, Chen L, Zhang Z, Xu Y, Song B, Xu R, Oudit G, Gao P, Zhu D and Zhong J (2015) Deletion of angiotensin-converting enzyme 2 exacerbates renal inflammation and injury in apolipoprotein E-deficient mice through modulation of the nephrin and TNF-alpha-TNFRSF1A signaling, Journal of Translational Medicine, 10.1186/s12967-015-0616-8, 13:1, Online publication date: 1-Dec-2015. Chytilová A, Borchert G, Mandíková-Alánová P, Hlaváčková M, Kopkan L, Khan M, Imig J, Kolář F and Neckář J (2015) Tumour necrosis factor- α contributes to improved cardiac ischaemic tolerance in rats adapted to chronic continuous hypoxia , Acta Physiologica, 10.1111/apha.12489, 214:1, (97-108), Online publication date: 1-May-2015. Liu Q, Wang T, Yu H, Liu B and Jia R (2014) Interaction Between Interleukin-1 Beta and Angiotensin II Receptor 1 in Hypothalamic Paraventricular Nucleus Contributes to Progression of Heart Failure, Journal of Interferon & Cytokine Research, 10.1089/jir.2013.0159, 34:11, (870-875), Online publication date: 1-Nov-2014. Schönberger T, Jürgens T, Müller J, Armbruster N, Niermann C, Gorressen S, Sommer J, Tian H, di Paolo G, Scheller J, Fischer J, Gawaz M and Elvers M (2014) Pivotal Role of Phospholipase D1 in Tumor Necrosis Factor-α–Mediated Inflammation and Scar Formation after Myocardial Ischemia and Reperfusion in Mice, The American Journal of Pathology, 10.1016/j.ajpath.2014.06.005, 184:9, (2450-2464), Online publication date: 1-Sep-2014. Duerrschmid C, Crawford J, Reineke E, Taffet G, Trial J, Entman M and Haudek S (2013) TNF receptor 1 signaling is critically involved in mediating angiotensin-II-induced cardiac fibrosis, Journal of Molecular and Cellular Cardiology, 10.1016/j.yjmcc.2013.01.006, 57, (59-67), Online publication date: 1-Apr-2013. Liu H, Luiten P, Eisel U, Dejongste M and Schoemaker R (2013) Depression after myocardial infarction: TNF-α-induced alterations of the blood–brain barrier and its putative therapeutic implications, Neuroscience & Biobehavioral Reviews, 10.1016/j.neubiorev.2013.02.004, 37:4, (561-572), Online publication date: 1-May-2013. Diamantopoulos A, Larsen A and Omdal R (2013) Is it safe to use TNF-α blockers for systemic inflammatory disease in patients with heart failure? Importance of dosage and receptor specificity, International Journal of Cardiology, 10.1016/j.ijcard.2012.11.112, 167:5, (1719-1723), Online publication date: 1-Sep-2013. Syriga M and Mavroidis M (2013) Complement System Activation in Cardiac and Skeletal Muscle Pathology: Friend or Foe? Complement Therapeutics, 10.1007/978-1-4614-4118-2_14, (207-218), . Ljung L, Simard J, Jacobsson L, Rantapää-Dahlqvist S and Askling J (2011) Treatment with tumor necrosis factor inhibitors and the risk of acute coronary syndromes in early rheumatoid arthritis, Arthritis & Rheumatism, 10.1002/art.30654, 64:1, (42-52), Online publication date: 1-Jan-2012. Khanna A, Xu J and Mehra M (2011) Antioxidant N-acetyl cysteine reverses cigarette smoke-induced myocardial infarction by inhibiting inflammation and oxidative stress in a rat model, Laboratory Investigation, 10.1038/labinvest.2011.146, 92:2, (224-235), Online publication date: 1-Feb-2012. Lacerda L, Opie L and Lecour S (2012) Influence of Tumour Necrosis Factor Alpha on the Outcome of Ischaemic Postconditioning in the Presence of Obesity and Diabetes, Experimental Diabetes Research, 10.1155/2012/502654, 2012, (1-10), . Villarreal-Calderon R, Reed W, Palacios-Moreno J, Keefe S, Herritt L, Brooks D, Torres-Jardón R and Calderón-Garcidueñas L (2012) Urban air pollution produces up-regulation of myocardial inflammatory genes and dark chocolate provides cardioprotection, Experimental and Toxicologic Pathology, 10.1016/j.etp.2010.09.002, 64:4, (297-306), Online publication date: 1-May-2012. Villarreal-Calderon R, Dale G, Delgado-Chávez R, Torres-Jardón R, Zhu H, Herritt L, Gónzalez-Maciel A, Reynoso-Robles R, Yuan Y, Wang J, Solorio-López E, Medina-Cortina H and Calderón-Garcidueñas L (2012) Intra-city Differences in Cardiac Expression of Inflammatory Genes and Inflammasomes in Young Urbanites: A Pilot Study, Journal of Toxicologic Pathology, 10.1293/tox.25.163, 25:2, (163-173), . Kuzmin V, Abramochkin D, Mitrochin V, Tian B, Makarenko E, Kovalchuk L, Khoreva M, Nikonova A, Kalugin L, Lysenko N, Lozinsky I, Rozanov A, Arutyunov G, Kiseleva I and Kamkin A (2012) The Role of Proinflammatory Cytokines in Regulation of Cardiac Bioelectrical Activity: Link to Mechanoelectrical Feedback Mechanical Stretch and Cytokines, 10.1007/978-94-007-2004-6_5, (107-153), . Parajuli N, Yuan Y, Zheng X, Bedja D and Cai Z (2012) Phosphatase PTEN is critically involved in post-myocardial infarction remodeling through the Akt/interleukin-10 signaling pathway, Basic Research in Cardiology, 10.1007/s00395-012-0248-6, 107:2, Online publication date: 1-Mar-2012. Martin M and Blaxall B (2012) Cardiac Intercellular Communication: Are Myocytes and Fibroblasts Fair-Weather Friends?, Journal of Cardiovascular Translational Research, 10.1007/s12265-012-9404-5, 5:6, (768-782), Online publication date: 1-Dec-2012. Kang Y, Gao F, Li H, Cardinale J, Elks C, Zang W, Yu X, Xu Y, Qi J, Yang Q and Francis J (2011) NF-κB in the paraventricular nucleus modulates neurotransmitters and contributes to sympathoe

Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call