C. Janeway, Approaching the Asymptote? Evolution and Revolution in Immunology, Cold Spring Harbor Symposia on Quantitative Biology, vol.54, issue.0, pp.1-13, 1989.
DOI : 10.1101/SQB.1989.054.01.003

R. Medzhitov, Toll-like receptors and innate immunity, Nature Reviews Immunology, vol.274, issue.2, pp.135-145, 2001.
DOI : 10.1074/jbc.274.19.12955

K. Anderson, J. "-urgens, G. , and N. , Establishment of dorsal-ventral polarity in the Drosophila embryo: Genetic studies on the role of the Toll gene product, Cell, vol.42, issue.3, p.779, 1985.
DOI : 10.1016/0092-8674(85)90274-0

B. Lemaitre, E. Nicolas, L. Michaut, J. Reichhart, and J. Hoffmann, The Dorsoventral Regulatory Gene Cassette sp??tzle/Toll/cactus Controls the Potent Antifungal Response in Drosophila Adults, Cell, vol.86, issue.6, pp.973-983, 1996.
DOI : 10.1016/S0092-8674(00)80172-5

R. Medzhitov, P. Preston-hurlburt, and C. Janeway, A human homologue of the Drosophila Toll protein signals activation of adaptive immunity, Nature, vol.254, issue.6640, pp.394-397, 1997.
DOI : 10.1126/science.1925582

H. Kono and K. Rock, How dying cells alert the immune system to danger, Nature Reviews Immunology, vol.171, issue.4, pp.279-289, 2008.
DOI : 10.4049/jimmunol.171.1.398

T. Arumugam, E. Okun, S. Tang, J. Thundyil, S. Taylor et al., TOLL-LIKE RECEPTORS IN ISCHEMIA-REPERFUSION INJURY, Shock, vol.32, issue.1, pp.4-16, 2009.
DOI : 10.1097/SHK.0b013e318193e333

J. Klesney-tait, I. Turnbull, and M. Colonna, The TREM receptor family and signal integration, Nature Immunology, vol.20, issue.12, pp.1266-1273, 2006.
DOI : 10.1096/fj.05-5646com

R. Allcock, A. Barrow, S. Forbes, S. Beck, and J. Trowsdale, The human TREM gene cluster at 6p21.1 encodes both activating and inhibitory single IgV domain receptors and includes NKp44, European Journal of Immunology, vol.33, issue.2, pp.567-577, 2003.
DOI : 10.1002/immu.200310033

A. Bouchon, J. Dietrich, and M. Colonna, Cutting Edge: Inflammatory Responses Can Be Triggered by TREM-1, a Novel Receptor Expressed on Neutrophils and Monocytes, The Journal of Immunology, vol.164, issue.10, pp.4991-4995, 2000.
DOI : 10.4049/jimmunol.164.10.4991

A. Tessarz and A. Cerwenka, The TREM-1/DAP12 pathway, Immunology Letters, vol.116, issue.2, pp.111-116, 2008.
DOI : 10.1016/j.imlet.2007.11.021

A. Bouchon, F. Facchetti, M. Weigand, and M. Colonna, TREM-1 amplifies inflammation and is a crucial mediator of septic shock, Nature, vol.410, issue.6832, pp.1103-1107, 2001.
DOI : 10.1038/35074114

S. Gibot, F. Massin, L. Renard, P. , B. Mc et al., Surface and soluble triggering receptor expressed on myeloid cells-1: Expression patterns in murine sepsis, Critical Care Medicine, vol.33, issue.8, pp.1787-1793, 2005.
DOI : 10.1097/01.CCM.0000172614.36571.75

M. Kelker, T. Foss, W. Peti, L. Teyton, J. Kelly et al., TREM-1) at 1.47 Å, Crystal Structure of Human Triggering Receptor Expressed on Myeloid Cells J Mol Biol, vol.1342, pp.1237-1248, 2004.

I. Wong-baeza, N. Gonz-'alez-rold-'an, E. Ferat-osorio, N. Esquivel-callejas, R. Aduna-vicente et al., Triggering receptor expressed on myeloid cells (TREM-1) is regulated post-transcriptionally and its ligand is present in the sera of some septic patients, Clinical and Experimental Immunology, vol.171, issue.3, pp.448-455, 2006.
DOI : 10.4049/jimmunol.171.2.594

A. Washington, S. Gibot, I. Acevedo, J. Gattis, L. Quigley et al., TREM-like transcript-1 protects against inflammation-associated hemorrhage by facilitating platelet aggregation in mice and humans, Journal of Clinical Investigation, vol.119, issue.6, p.1489, 2009.
DOI : 10.1172/JCI36175DS1

M. Derive, Y. Bouazza, N. Sennoun, S. Marchionni, L. Quigley et al., Soluble Trem-like Transcript-1 Regulates Leukocyte Activation and Controls Microbial Sepsis, The Journal of Immunology, vol.188, issue.11, pp.5585-5592, 2012.
DOI : 10.4049/jimmunol.1102674

URL : http://www.jimmunol.org/content/jimmunol/188/11/5585.full.pdf

M. Nichols, N. Townsend, P. Scarborough, and M. Rayner, Cardiovascular disease in Europe 2014: epidemiological update, European Heart Journal, vol.60, issue.42, pp.2950-2959, 2014.
DOI : 10.1016/j.jacc.2012.08.962

URL : https://academic.oup.com/eurheartj/article-pdf/35/42/2950/17354923/ehu299.pdf

B. Sobel, G. Bresnahan, W. Shell, and R. Yoder, Estimation of Infarct Size in Man and its Relation to Prognosis, Circulation, vol.46, issue.4, pp.640-648, 1972.
DOI : 10.1161/01.CIR.46.4.640

E. Puymirat, T. Simon, P. Steg, F. Schiele, P. Guéret et al., Association of Changes in Clinical Characteristics and Management With Improvement in Survival Among Patients With ST-Elevation Myocardial Infarction, Investigators, FAST MI Investigators, pp.998-1006, 2000.
DOI : 10.1001/2012.jama.11348

M. Hall, S. Levant, and C. Defrances, Hospitalization for congestive heart failure: United States, NCHS Data Brief, pp.1-8, 2000.

D. Yellon and D. Hausenloy, Myocardial Reperfusion Injury, New England Journal of Medicine, vol.357, issue.11, p.1121, 2007.
DOI : 10.1056/NEJMra071667

URL : https://hal.archives-ouvertes.fr/hal-01608633

R. Jennings, H. Sommers, G. Smyth, H. Flack, and H. Linn, Myocardial necrosis induced by temporary occlusion of a coronary artery in the dog, Arch Pathol, vol.70, pp.68-78, 1960.

T. Scarabelli and R. Gottlieb, Functional and clinical repercussions of myocyte apoptosis in the multifaceted damage by ischemia/reperfusion injury: old and new concepts after 10 years of contributions, Cell Death and Differentiation, vol.11, pp.144-152, 2004.
DOI : 10.1038/sj.cdd.4401544

J. Zweier, Measurement of superoxide-derived free radicals in the reperfused heart. Evidence for a free radical mechanism of reperfusion injury, J Biol Chem, vol.263, pp.1353-1357, 1988.

G. Petrosillo, F. Ruggiero, and G. Paradies, from mitochondria, The FASEB Journal, vol.17, issue.15, pp.2202-2208, 2003.
DOI : 10.1016/S0959-440X(03)00008-3

D. Coquerel and F. Tamion, Myocardial ischaemia and reperfusion ??? pathophysiological aspects, R??animation, vol.20, issue.S2, pp.267-273, 2010.
DOI : 10.1007/s13546-010-0102-5

J. Cohn, R. Ferrari, and N. Sharpe, Cardiac remodeling???concepts and clinical implications: a consensus paper from an international forum on cardiac remodeling, Journal of the American College of Cardiology, vol.35, issue.3, pp.569-582, 2000.
DOI : 10.1016/S0735-1097(99)00630-0

H. Weisman, D. Bush, J. Mannisi, M. Weisfeldt, and B. Healy, Cellular mechanisms of myocardial infarct expansion, Circulation, vol.78, issue.1, pp.186-201, 1988.
DOI : 10.1161/01.CIR.78.1.186

J. Gajarsa and R. Kloner, Left ventricular remodeling in the post-infarction heart: a review of cellular, molecular mechanisms, and therapeutic modalities, Heart Failure Reviews, vol.121, issue.19, pp.13-21, 2011.
DOI : 10.1161/CIRCULATIONAHA.108.808071

N. Frangogiannis, The Mechanistic Basis of Infarct Healing, Antioxidants & Redox Signaling, vol.8, issue.11-12, pp.1907-1939, 2006.
DOI : 10.1089/ars.2006.8.1907

N. Frangogiannis, The Immune System and the Remodeling Infarcted Heart, Journal of Cardiovascular Pharmacology, vol.63, issue.3, pp.185-195, 2014.
DOI : 10.1097/FJC.0000000000000003

W. Blankesteijn, E. Creemers, E. Lutgens, J. Cleutjens, M. Daemen et al., Dynamics of cardiac wound healing following myocardial infarction: observations in genetically altered mice, Acta Physiologica Scandinavica, vol.145, issue.1, pp.75-82, 2001.
DOI : 10.1016/S0959-437X(98)80068-3

J. Cleutjens, The role of matrix metalloproteinases in heart disease, Cardiovascular Research, vol.32, issue.5, pp.816-821, 1996.
DOI : 10.1016/S0008-6363(96)00104-6

G. Gabbiani, Evolution and clinical implications of the myofibroblast concept, Cardiovascular Research, vol.38, issue.3, pp.545-548, 1998.
DOI : 10.1016/S0008-6363(98)00065-0

J. Janicki and G. Brower, The role of myocardial fibrillar collagen in ventricular remodeling and function, Journal of Cardiac Failure, vol.8, issue.6, pp.319-325, 2002.
DOI : 10.1054/jcaf.2002.129260

F. Spinale, Myocardial Matrix Remodeling and the Matrix Metalloproteinases: Influence on Cardiac Form and Function, Physiological Reviews, vol.87, issue.4, pp.1285-1342, 2007.
DOI : 10.1152/ajpheart.00160.2004

N. Kawamura, T. Kubota, S. Kawano, Y. Monden, A. Feldman et al., Blockade of NF-??B improves cardiac function and survival without affecting inflammation in TNF-??-induced cardiomyopathy, Cardiovascular Research, vol.66, issue.3, pp.520-529, 2005.
DOI : 10.1016/j.cardiores.2005.02.007

C. Long, The Role of Interleukin-1 in the Failing Heart, Heart Fail Rev, vol.6, pp.81-94, 2001.
DOI : 10.1007/978-1-4615-1449-7_3

R. García, K. Brown, R. Pavelec, K. Go, J. Covell et al., Abnormal cardiac wall motion and early matrix metalloproteinase activity, American Journal of Physiology-Heart and Circulatory Physiology, vol.288, issue.3, pp.1080-1087, 2005.
DOI : 10.1074/jbc.273.11.6550

S. Philip and G. Kundu, Osteopontin Induces Nuclear Factor ??B-mediated Promatrix Metalloproteinase-2 Activation through I??B??/IKK Signaling Pathways, and Curcumin (Diferulolylmethane) Down-regulates These Pathways, Journal of Biological Chemistry, vol.163, issue.16, pp.14487-14497, 2003.
DOI : 10.3109/00498259609046741

URL : http://www.jbc.org/content/278/16/14487.full.pdf

Z. Yang, T. Kyriakides, and P. Bornstein, Matricellular Proteins as Modulators of Cell-Matrix Interactions: Adhesive Defect in Thrombospondin 2-null Fibroblasts is a Consequence of Increased Levels of Matrix Metalloproteinase-2, Molecular Biology of the Cell, vol.11, issue.10, pp.3353-3364, 2000.
DOI : 10.1091/mbc.11.10.3353

W. Yuan and J. Varga, Transforming Growth Factor-?? Repression of Matrix Metalloproteinase-1 in Dermal Fibroblasts Involves Smad3, Journal of Biological Chemistry, vol.14, issue.42, pp.38502-38510, 2001.
DOI : 10.1038/29814

L. Guedez, W. Stetler-stevenson, L. Wolff, J. Wang, P. Fukushima et al., In vitro suppression of programmed cell death of B cells by tissue inhibitor of metalloproteinases-1., Journal of Clinical Investigation, vol.102, issue.11, pp.2002-2010, 1998.
DOI : 10.1172/JCI2881

S. Tyagi, S. Campbell, H. Reddy, E. Tjahja, and D. Voelker, Matrix metalloproteinase activity expression in infarcted, noninfarcted and dilated cardiomyopathic human hearts, Molecular and Cellular Biochemistry, vol.229, issue.suppl II, pp.13-21, 1996.
DOI : 10.1161/01.RES.67.1.23

S. Heymans, A. Luttun, D. Nuyens, G. Theilmeier, E. Creemers et al., Inhibition of plasminogen activators or matrix metalloproteinases prevents cardiac rupture but impairs therapeutic angiogenesis and causes cardiac failure, Nature Medicine, vol.267, issue.10, pp.1135-1142, 1999.
DOI : 10.1126/science.7863332

E. Wilson, S. Moainie, J. Baskin, A. Lowry, A. Deschamps et al., Region- and Type-Specific Induction of Matrix Metalloproteinases in Post-Myocardial Infarction Remodeling, Circulation, vol.107, issue.22, pp.2857-2863, 2003.
DOI : 10.1161/01.CIR.0000068375.40887.FA

R. Mukherjee, T. Brinsa, K. Dowdy, A. Scott, J. Baskin et al., Myocardial Infarct Expansion and Matrix Metalloproteinase Inhibition, Circulation, vol.107, issue.4, pp.618-625, 2003.
DOI : 10.1161/01.CIR.0000046449.36178.00

URL : http://circ.ahajournals.org/content/circulationaha/107/4/618.full.pdf

K. Apple, W. Yarbrough, R. Mukherjee, A. Deschamps, P. Escobar et al., Selective Targeting of Matrix Metalloproteinase Inhibition in Post-Infarction Myocardial Remodeling, Journal of Cardiovascular Pharmacology, vol.47, issue.2, pp.228-235, 2006.
DOI : 10.1097/01.fjc.0000200989.23987.b8

M. Hudson, P. Armstrong, W. Ruzyllo, J. Brum, L. Cusmano et al., Effects of Selective Matrix Metalloproteinase Inhibitor (PG-116800) to Prevent Ventricular Remodeling After Myocardial Infarction, Journal of the American College of Cardiology, vol.48, issue.1, pp.15-20, 2006.
DOI : 10.1016/j.jacc.2006.02.055

D. Brown, K. Desai, B. Vakili, C. Nouneh, H. Lee et al., Clinical and Biochemical Results of the Metalloproteinase Inhibition with Subantimicrobial Doses of Doxycycline to Prevent Acute Coronary Syndromes (MIDAS) Pilot Trial, Arteriosclerosis, Thrombosis, and Vascular Biology, vol.24, issue.4, pp.733-738, 2004.
DOI : 10.1161/01.ATV.0000121571.78696.dc

J. Yang, Y. Liu, X. Fan, Z. Li, and Y. Cheng, A pathway and network review on beta-adrenoceptor signaling and beta blockers in cardiac remodeling, Heart Failure Reviews, vol.65, issue.6, pp.799-814, 2014.
DOI : 10.1016/j.freeradbiomed.2013.06.033

V. Busatto, V. Cunha, M. Cicilini, and J. Mill, Differential effects of isoproterenol on the activity of angiotensin-converting enzyme in the rat heart and aorta, Brazilian Journal of Medical and Biological Research, vol.24, issue.3, pp.355-360, 1999.
DOI : 10.1161/01.HYP.8.12.1154

R. Doughty, G. Whalley, H. Walsh, G. Gamble, J. López-sendón et al., Effects of Carvedilol on Left Ventricular Remodeling After Acute Myocardial Infarction: The CAPRICORN Echo Substudy, Circulation, vol.109, issue.2, pp.201-206, 2004.
DOI : 10.1161/01.CIR.0000108928.25690.94

B. Groenning, J. Nilsson, L. Sondergaard, T. Fritz-hansen, H. Larsson et al., Antiremodeling effects on the left ventricle during beta-blockade with metoprolol in the treatment of chronic heart failure, Journal of the American College of Cardiology, vol.36, issue.7, pp.2072-2080, 2000.
DOI : 10.1016/S0735-1097(00)01006-8

W. Iraqi, P. Rossignol, M. Angioi, R. Fay, J. Nuée et al., Extracellular Cardiac Matrix Biomarkers in Patients With Acute Myocardial Infarction Complicated by Left Ventricular Dysfunction and Heart Failure: Insights From the Eplerenone Post-Acute Myocardial Infarction Heart Failure Efficacy and Survival Study (EPHESUS) Study, Circulation, vol.119, issue.18, pp.2471-2479, 2009.
DOI : 10.1161/CIRCULATIONAHA.108.809194

M. Konstam, M. Rousseau, M. Kronenberg, J. Udelson, J. Melin et al., Effects of the angiotensin converting enzyme inhibitor enalapril on the long-term progression of left ventricular dysfunction in patients with heart failure. SOLVD Investigators, SOLVD Investigators, pp.431-438, 1992.
DOI : 10.1161/01.CIR.86.2.431

R. Mckelvie, S. Yusuf, D. Pericak, A. Avezum, R. Burns et al., Comparison of Candesartan, Enalapril, and Their Combination in Congestive Heart Failure : Randomized Evaluation of Strategies for Left Ventricular Dysfunction (RESOLVD) Pilot Study: The RESOLVD Pilot Study Investigators, Circulation, vol.100, issue.10, pp.1056-1064, 1999.
DOI : 10.1161/01.CIR.100.10.1056

A. Chan, J. Sanderson, T. Wang, W. Lam, G. Yip et al., Aldosterone Receptor Antagonism Induces Reverse Remodeling When Added to Angiotensin Receptor Blockade in Chronic Heart Failure, Journal of the American College of Cardiology, vol.50, issue.7, pp.591-596, 2007.
DOI : 10.1016/j.jacc.2007.03.062

J. Mcmurray, S. Adamopoulos, S. Anker, A. Auricchio, M. Böhm et al., ESC Guidelines for the diagnosis and treatment of acute and chronic heart failure 2012 The Task Force for the Diagnosis and Treatment of Acute and Chronic Heart Failure 2012 of the European Society of Cardiology. Developed in collaboration with the Heart Failure Association (HFA) of the ESC, Eur Heart J, vol.33, pp.1787-1847, 2012.

N. Frangogiannis, Regulation of the Inflammatory Response in Cardiac Repair, Circulation Research, vol.110, issue.1, pp.159-173, 2012.
DOI : 10.1161/CIRCRESAHA.111.243162

M. Andrassy, H. Volz, J. Igwe, B. Funke, S. Eichberger et al., High-Mobility Group Box-1 in Ischemia-Reperfusion Injury of the Heart, Circulation, vol.117, issue.25, pp.3216-3226, 2008.
DOI : 10.1161/CIRCULATIONAHA.108.769331

L. Timmers, G. Pasterkamp, V. Hoog, . De, F. Arslan et al., The innate immune response in reperfused myocardium, Cardiovascular Research, vol.293, issue.4, pp.276-283, 2012.
DOI : 10.1152/ajpheart.00776.2007

J. Jordan, M. Montalto, and G. Stahl, Inhibition of Mannose-Binding Lectin Reduces Postischemic Myocardial Reperfusion Injury, Circulation, vol.104, issue.12, pp.1413-1418, 2001.
DOI : 10.1161/hc3601.095578

H. Weisman, T. Bartow, M. Leppo, H. Marsh, G. Carson et al., Soluble human complement receptor type 1: in vivo inhibitor of complement suppressing post-ischemic myocardial inflammation and necrosis, Science, vol.249, issue.4965, pp.146-151, 1990.
DOI : 10.1126/science.2371562

K. Hensley, K. Robinson, S. Gabbita, S. Salsman, and R. Floyd, Reactive oxygen species, cell signaling, and cell injury, Free Radical Biology and Medicine, vol.28, issue.10, pp.1456-1462, 2000.
DOI : 10.1016/S0891-5849(00)00252-5

W. Zhao, T. Zhao, Y. Chen, R. Ahokas, and Y. Sun, Reactive oxygen species promote angiogenesis in the infarcted rat heart, International Journal of Experimental Pathology, vol.28, issue.6, pp.621-629, 2009.
DOI : 10.1159/000115289

J. Gordon, J. Shaw, and L. Kirshenbaum, Multiple Facets of NF-??B in the Heart, Circulation Research, vol.108, issue.9, pp.1122-1132, 2011.
DOI : 10.1161/CIRCRESAHA.110.226928

E. Mezzaroma, S. Toldo, D. Farkas, I. Seropian, V. Tassell et al., The inflammasome promotes adverse cardiac remodeling following acute myocardial infarction in the mouse, Proceedings of the National Academy of Sciences, vol.293, issue.3, pp.19725-19730, 2011.
DOI : 10.1152/ajpheart.00291.2007

M. Kawaguchi, M. Takahashi, T. Hata, Y. Kashima, F. Usui et al., Inflammasome Activation of Cardiac Fibroblasts Is Essential for Myocardial Ischemia/Reperfusion Injury, Circulation, vol.123, issue.6, pp.594-604, 2011.
DOI : 10.1161/CIRCULATIONAHA.110.982777

N. Frangogiannis, M. Lindsey, L. Michael, K. Youker, R. Bressler et al., Resident Cardiac Mast Cells Degranulate and Release Preformed TNF-??, Initiating the Cytokine Cascade in Experimental Canine Myocardial Ischemia/Reperfusion, Circulation, vol.98, issue.7, pp.699-710, 1998.
DOI : 10.1161/01.CIR.98.7.699

T. Hamid, Y. Gu, R. Ortines, C. Bhattacharya, G. Wang et al., Divergent Tumor Necrosis Factor Receptor-Related Remodeling Responses in Heart Failure: Role of Nuclear Factor-??B and Inflammatory Activation, Circulation, vol.119, issue.10, pp.1386-1397, 2009.
DOI : 10.1161/CIRCULATIONAHA.108.802918

P. Fischer and D. Hilfiker-kleiner, Role of gp130-mediated signalling pathways in the heart and its impact on potential therapeutic aspects, British Journal of Pharmacology, vol.280, issue.6 Suppl, pp.414-427, 2008.
DOI : 10.4049/jimmunol.178.8.4832

D. Hilfiker-kleiner, P. Shukla, G. Klein, A. Schaefer, B. Stapel et al., Continuous Glycoprotein-130-Mediated Signal Transducer and Activator of Transcription-3 Activation Promotes Inflammation, Left Ventricular Rupture, and Adverse Outcome in Subacute Myocardial Infarction, Circulation, vol.122, issue.2, pp.145-155, 2010.
DOI : 10.1161/CIRCULATIONAHA.109.933127

N. Frangogiannis, The immune system and cardiac repair, Pharmacological Research, vol.58, issue.2, pp.88-111, 2008.
DOI : 10.1016/j.phrs.2008.06.007

T. Mocatta, A. Pilbrow, V. Cameron, R. Senthilmohan, C. Frampton et al., Plasma Concentrations of Myeloperoxidase Predict Mortality After Myocardial Infarction, Journal of the American College of Cardiology, vol.49, issue.20, pp.1993-2000, 2007.
DOI : 10.1016/j.jacc.2007.02.040

L. Ng, S. Khan, H. Narayan, P. Quinn, I. Squire et al., Proteinase 3 and prognosis of patients with acute myocardial infarction, Clinical Science, vol.120, issue.6, pp.231-238, 1979.
DOI : 10.1042/CS20100366

URL : https://hal.archives-ouvertes.fr/hal-00647879

A. Savchenko, J. Borissoff, K. Martinod, D. Meyer, S. Gallant et al., VWF-mediated leukocyte recruitment with chromatin decondensation by PAD4 increases myocardial ischemia/reperfusion injury in mice, Blood, vol.123, issue.1, pp.141-148, 2014.
DOI : 10.1182/blood-2013-07-514992

N. Maugeri, L. Campana, M. Gavina, C. Covino, D. Metrio et al., Activated platelets present high mobility group box 1 to neutrophils, inducing autophagy and promoting the extrusion of neutrophil extracellular traps, Journal of Thrombosis and Haemostasis, vol.86, issue.12, pp.2074-2088, 2014.
DOI : 10.1016/S0049-3848(97)00096-0

O. Soehnlein and L. Lindbom, Phagocyte partnership during the onset and resolution of inflammation, Nature Reviews Immunology, vol.8, issue.6, pp.427-439, 2010.
DOI : 10.4049/jimmunol.180.10.6868

M. Huang, D. Yang, M. Xiang, and J. Wang, Role of interleukin-6 in regulation of immune responses to remodeling after myocardial infarction, Heart Failure Reviews, vol.367, issue.1, pp.25-38, 2015.
DOI : 10.1056/NEJMoa1112802

K. Jung, P. Kim, F. Leuschner, R. Gorbatov, J. Kim et al., Endoscopic Time-Lapse Imaging of Immune Cells in Infarcted Mouse Hearts, Circulation Research, vol.112, issue.6, pp.891-899, 2013.
DOI : 10.1161/CIRCRESAHA.111.300484

F. Swirski and M. Nahrendorf, Leukocyte Behavior in Atherosclerosis, Myocardial Infarction, and Heart Failure, Science, vol.339, issue.6116, pp.161-166, 2013.
DOI : 10.1126/science.1230720

M. Nahrendorf, F. Swirski, E. Aikawa, L. Stangenberg, T. Wurdinger et al., The healing myocardium sequentially mobilizes two monocyte subsets with divergent and complementary functions, The Journal of Experimental Medicine, vol.52, issue.12, pp.3037-3047, 2007.
DOI : 10.1007/BF00788278

F. Swirski, M. Nahrendorf, M. Etzrodt, M. Wildgruber, V. Cortez-retamozo et al., Identification of Splenic Reservoir Monocytes and Their Deployment to Inflammatory Sites, Science, vol.25, issue.3, pp.612-616, 2009.
DOI : 10.1016/0002-9343(88)90206-9

S. Frantz and M. Nahrendorf, Cardiac macrophages and their role in ischaemic heart disease, Cardiovascular Research, vol.34, issue.11 Suppl 1, pp.240-248, 2014.
DOI : 10.1093/eurheartj/ehs366

M. Baldridge, K. King, and M. Goodell, Inflammatory signals regulate hematopoietic stem cells, Trends in Immunology, vol.32, issue.2, pp.57-65, 2011.
DOI : 10.1016/j.it.2010.12.003

URL : http://europepmc.org/articles/pmc3042730?pdf=render

H. Shinagawa and S. Frantz, Cellular Immunity and Cardiac Remodeling After Myocardial Infarction: Role of Neutrophils, Monocytes, and Macrophages, Current Heart Failure Reports, vol.34, issue.1 Pt 2, pp.247-254, 2015.
DOI : 10.1093/eurheartj/ehs366

I. Hilgendorf, L. Gerhardt, T. Tan, C. Winter, T. Holderried et al., Ly-6Chigh Monocytes Depend on Nr4a1 to Balance Both Inflammatory and Reparative Phases in the Infarcted Myocardium, Circulation Research, vol.114, issue.10, pp.1611-1622, 2014.
DOI : 10.1161/CIRCRESAHA.114.303204

U. Hofmann and S. Frantz, Role of Lymphocytes in Myocardial Injury, Healing, and Remodeling After Myocardial Infarction, Circulation Research, vol.116, issue.2, pp.354-367, 2015.
DOI : 10.1161/CIRCRESAHA.116.304072

Y. Zouggari, H. Ait-oufella, P. Bonnin, T. Simon, A. Sage et al., B lymphocytes trigger monocyte mobilization and impair heart function after acute myocardial infarction, Nature Medicine, vol.360, issue.10, pp.1273-1280, 2013.
DOI : 10.1056/NEJMoa0808227

URL : http://europepmc.org/articles/pmc4042928?pdf=render

Z. Yang, Y. Day, M. Toufektsian, Y. Xu, S. Ramos et al., Myocardial Infarct-Sparing Effect of Adenosine A2A Receptor Activation Is due to Its Action on CD4+ T Lymphocytes, Circulation, vol.114, issue.19, pp.2056-2064, 2006.
DOI : 10.1161/CIRCULATIONAHA.106.649244

U. Hofmann, N. Beyersdorf, J. Weirather, A. Podolskaya, J. Bauersachs et al., Activation of CD4+ T Lymphocytes Improves Wound Healing and Survival After Experimental Myocardial Infarction in Mice, Circulation, vol.125, issue.13, pp.1652-1663, 2012.
DOI : 10.1161/CIRCULATIONAHA.111.044164

J. Weirather, U. Hofmann, N. Beyersdorf, G. Ramos, B. Vogel et al., Foxp3+ CD4+ T Cells Improve Healing After Myocardial Infarction by Modulating Monocyte/Macrophage Differentiation, Circulation Research, vol.115, issue.1, 2014.
DOI : 10.1161/CIRCRESAHA.115.303895

URL : http://circres.ahajournals.org/content/circresaha/115/1/55.full.pdf

P. Hiebert, D. Wu, and D. Granville, Granzyme B degrades extracellular matrix and contributes to delayed wound closure in apolipoprotein E knockout mice, Cell Death & Differentiation, vol.4, issue.10, pp.1404-1414, 2013.
DOI : 10.1002/prot.340040306

H. Kondo, Y. Hojo, R. Tsuru, Y. Nishimura, H. Shimizu et al., Elevation of Plasma Granzyme B Levels After Acute Myocardial Infarction, Circulation Journal, vol.73, issue.3, pp.503-507, 2009.
DOI : 10.1253/circj.CJ-08-0668

N. Varda-bloom, J. Leor, D. Ohad, Y. Hasin, M. Amar et al., Cytotoxic T Lymphocytes Are Activated Following Myocardial Infarction and Can Recognize and Kill Healthy Myocytes In Vitro, Journal of Molecular and Cellular Cardiology, vol.32, issue.12, pp.2141-2149, 2000.
DOI : 10.1006/jmcc.2000.1261

E. Wan, X. Yeap, S. Dehn, R. Terry, M. Novak et al., Enhanced Efferocytosis of Apoptotic Cardiomyocytes Through Myeloid-Epithelial-Reproductive Tyrosine Kinase Links Acute Inflammation Resolution to Cardiac Repair After Infarction, Circulation Research, vol.113, issue.8, pp.1004-1012, 2013.
DOI : 10.1161/CIRCRESAHA.113.301198

A. Saxena, M. Dobaczewski, V. Rai, Z. Haque, W. Chen et al., Regulatory T cells are recruited in the infarcted mouse myocardium and may modulate fibroblast phenotype and function, American Journal of Physiology-Heart and Circulatory Physiology, vol.142, issue.8, pp.1233-1242, 2014.
DOI : 10.1038/nm.3284

M. Ikeuchi, H. Tsutsui, T. Shiomi, H. Matsusaka, S. Matsushima et al., Inhibition of TGF-? signaling exacerbates early cardiac dysfunction but prevents late remodeling after infarction, Cardiovascular Research, vol.64, issue.3, pp.526-535, 2004.
DOI : 10.1016/j.cardiores.2004.07.017

T. Kempf, A. Zarbock, C. Widera, S. Butz, A. Stadtmann et al., GDF-15 is an inhibitor of leukocyte integrin activation required for survival after myocardial infarction in mice, Nature Medicine, vol.146, issue.5, pp.581-588, 2011.
DOI : 10.1084/jem.20072660

C. Cochain, C. Auvynet, L. Poupel, J. Vilar, E. Dumeau et al., The Chemokine Decoy Receptor D6 Prevents Excessive Inflammation and Adverse Ventricular Remodeling After Myocardial Infarction, Arteriosclerosis, Thrombosis, and Vascular Biology, vol.32, issue.9, pp.2206-2213, 2012.
DOI : 10.1161/ATVBAHA.112.254409

URL : http://atvb.ahajournals.org/content/atvbaha/32/9/2206.full.pdf

W. Chen, A. Saxena, N. Li, J. Sun, A. Gupta et al., Endogenous IRAK-M Attenuates Postinfarction Remodeling Through Effects on Macrophages and Fibroblasts, Arteriosclerosis, Thrombosis, and Vascular Biology, vol.32, issue.11, pp.2598-2608, 2012.
DOI : 10.1161/ATVBAHA.112.300310

URL : http://atvb.ahajournals.org/content/atvbaha/32/11/2598.full.pdf

I. Seropian, S. Toldo, V. Tassell, B. Abbate, and A. , Anti-Inflammatory Strategies for Ventricular Remodeling Following ST-Segment Elevation Acute Myocardial Infarction, Journal of the American College of Cardiology, vol.63, issue.16, pp.1593-1603, 2014.
DOI : 10.1016/j.jacc.2014.01.014

URL : https://doi.org/10.1016/j.jacc.2014.01.014

K. Baran, M. Nguyen, G. Mckendall, C. Lambrew, G. Dykstra et al., Double-Blind, Randomized Trial of an Anti-CD18 Antibody in Conjunction With Recombinant Tissue Plasminogen Activator for Acute Myocardial Infarction: Limitation of Myocardial Infarction Following Thrombolysis in Acute Myocardial Infarction (LIMIT AMI) Study, Circulation, vol.104, issue.23, pp.2778-2783, 2001.
DOI : 10.1161/hc4801.100236

D. Faxon, R. Gibbons, N. Chronos, P. Gurbel, and F. Sheehan, The effect of blockade of the CD11/CD18 integrin receptor on infarct size in patients with acute myocardial infarction treated with direct angioplasty: the results of the HALT-MI study, Journal of the American College of Cardiology, vol.40, issue.7, pp.1199-1204, 2002.
DOI : 10.1016/S0735-1097(02)02136-8

K. Mahaffey, C. Granger, J. Nicolau, W. Ruzyllo, W. Weaver et al., Effect of Pexelizumab, an Anti-C5 Complement Antibody, as Adjunctive Therapy to Fibrinolysis in Acute Myocardial Infarction: The COMPlement inhibition in myocardial infarction treated with thromboLYtics (COMPLY) Trial, Circulation, vol.108, issue.10, pp.1176-1183, 2003.
DOI : 10.1161/01.CIR.0000087404.53661.F8

C. Granger, K. Mahaffey, W. Weaver, P. Theroux, J. Hochman et al., Pexelizumab, an Anti-C5 Complement Antibody, as Adjunctive Therapy to Primary Percutaneous Coronary Intervention in Acute Myocardial Infarction: The COMplement inhibition in Myocardial infarction treated with Angioplasty (COMMA) Trial, Circulation, vol.108, issue.10, pp.1184-1190, 2003.
DOI : 10.1161/01.CIR.0000087447.12918.85

P. Armstrong, C. Granger, P. Adams, C. Hamm, H. Jr et al., Van de Werf F, others. Pexelizumab for acute ST-elevation myocardial infarction in patients undergoing primary percutaneous coronary intervention: a randomized controlled trial, JAMA J Am Med Assoc, vol.297, p.43, 2007.

A. Ross, R. Gibbons, G. Stone, R. Kloner, and A. R. Randomized, A Randomized, Double-Blinded, Placebo-Controlled Multicenter Trial of Adenosine as an Adjunct to Reperfusion in the Treatment of Acute Myocardial Infarction (AMISTAD-II), Journal of the American College of Cardiology, vol.45, issue.11, pp.1775-1780, 2005.
DOI : 10.1016/j.jacc.2005.02.061

R. Kloner, M. Forman, R. Gibbons, A. Ross, R. Alexander et al., Impact of time to therapy and reperfusion modality on the efficacy of adenosine in acute myocardial infarction: the AMISTAD-2 trial, European Heart Journal, vol.27, issue.20, pp.2400-2405, 2006.
DOI : 10.1093/eurheartj/ehl094

J. Headrick, B. Hack, and K. Ashton, Acute adenosinergic cardioprotection in ischemic-reperfused hearts, American Journal of Physiology-Heart and Circulatory Physiology, vol.285, issue.5, pp.1797-1818, 2003.
DOI : 10.1161/01.CIR.88.5.2359

N. Frangogiannis, Inflammation in cardiac injury, repair and regeneration, Current Opinion in Cardiology, vol.30, issue.3, pp.240-245, 2015.
DOI : 10.1097/HCO.0000000000000158

F. Arslan, M. Smeets, O. Neill, L. Keogh, B. Mcguirk et al., Myocardial Ischemia/Reperfusion Injury Is Mediated by Leukocytic Toll-Like Receptor-2 and Reduced by Systemic Administration of a Novel Anti-Toll-Like Receptor-2 Antibody, Circulation, vol.121, issue.1, pp.80-90, 2010.
DOI : 10.1161/CIRCULATIONAHA.109.880187

J. Oyama, B. Jr, C. Liu, X. Pu, M. Kobzik et al., Reduced Myocardial Ischemia-Reperfusion Injury in Toll-Like Receptor 4-Deficient Mice, Circulation, vol.109, issue.6, pp.784-789, 2004.
DOI : 10.1161/01.CIR.0000112575.66565.84

R. Lefering and E. Neugebauer, Steroid controversy in sepsis and septic shock, Critical Care Medicine, vol.23, issue.7, p.1294, 1995.
DOI : 10.1097/00003246-199507000-00021

E. Abraham, P. Laterre, J. Garbino, S. Pingleton, T. Butler et al., Lenercept (p55 tumor necrosis factor receptor fusion protein) in severe sepsis and early septic shock: A randomized, double-blind, placebo-controlled, multicenter phase III trial with 1,342 patients, Critical Care Medicine, vol.29, issue.3, p.503, 2001.
DOI : 10.1097/00003246-200103000-00006

C. Fisher, J. Dhainaut, S. Opal, J. Pribble, R. Balk et al., Recombinant human interleukin 1 receptor antagonist in the treatment of patients with sepsis syndrome. Results from a randomized, double-blind, placebo-controlled trial. Phase III rhIL-1ra Sepsis Syndrome Study Group, JAMA: The Journal of the American Medical Association, vol.271, issue.23, pp.1836-1843, 1994.
DOI : 10.1001/jama.271.23.1836

S. Opal, P. Laterre, B. Francois, S. Larosa, D. Angus et al., Effect of Eritoran, an Antagonist of MD2-TLR4, on Mortality in Patients With Severe Sepsis, JAMA, vol.309, issue.11, pp.1154-1162, 2013.
DOI : 10.1001/jama.2013.2194

P. Eichacker, C. Parent, A. Kalil, C. Esposito, X. Cui et al., Risk and the Efficacy of Antiinflammatory Agents, American Journal of Respiratory and Critical Care Medicine, vol.166, issue.9, pp.1197-1205, 2002.
DOI : 10.1172/JCI112902

M. Derive, A. Boufenzer, Y. Bouazza, F. Groubatch, C. Alauzet et al., Effects of a TREM-Like Transcript 1???Derived Peptide During Hypodynamic Septic Shock in Pigs, Shock, vol.39, issue.2, pp.176-182, 2013.
DOI : 10.1097/SHK.0b013e31827bcdfb

URL : https://hal.archives-ouvertes.fr/hal-01272749

N. Frangogiannis, Targeting the Inflammatory Response in Healing Myocardial Infarcts, Current Medicinal Chemistry, vol.13, issue.16, pp.1877-1893, 2006.
DOI : 10.2174/092986706777585086

N. Frangogiannis, The inflammatory response in myocardial injury, repair and remodelling, Nature Reviews Cardiology, vol.147, issue.5, pp.255-265, 2014.
DOI : 10.1016/j.trsl.2012.01.023

S. Gibot, M. Kolopp-sarda, M. Béné, P. Bollaert, A. Lozniewski et al., A Soluble Form of the Triggering Receptor Expressed on Myeloid Cells-1 Modulates the Inflammatory Response in Murine Sepsis, The Journal of Experimental Medicine, vol.10, issue.11, pp.1419-1426, 2004.
DOI : 10.7326/0003-4819-141-1-200407060-00009

M. Derive, A. Boufenzer, and S. Gibot, Attenuation of Responses to Endotoxin by the Triggering Receptor Expressed on Myeloid Cells-1 Inhibitor LR12 in Nonhuman Primate, Anesthesiology, vol.120, issue.4, pp.935-942, 2014.
DOI : 10.1097/ALN.0000000000000078

URL : https://hal.archives-ouvertes.fr/hal-01738448

B. Weber, S. Schuster, D. Zysset, S. Rihs, N. Dickgreber et al., TREM-1 Deficiency Can Attenuate Disease Severity without Affecting Pathogen Clearance Available from, PLoS Pathog, vol.10, 2014.
DOI : 10.1371/journal.ppat.1003900

URL : https://doi.org/10.1371/journal.ppat.1003900

J. Klesney-tait, K. Keck, X. Li, S. Gilfillan, K. Otero et al., Transepithelial migration of neutrophils into the lung requires TREM-1, Journal of Clinical Investigation, vol.123, issue.1, pp.138-149, 2013.
DOI : 10.1172/JCI64181DS1

P. Lim, S. Moutereau, T. Simon, R. Gallet, V. Probst et al., Usefulness of Fetuin-A and C-Reactive Protein Concentrations for Prediction of Outcome in Acute Coronary Syndromes (from the French Registry of Acute ST-Elevation Non-ST-Elevation Myocardial Infarction [FAST-MI]), The American Journal of Cardiology, vol.111, issue.1, pp.31-37, 2013.
DOI : 10.1016/j.amjcard.2012.08.042

T. Simon, C. Verstuyft, M. Mary-krause, L. Quteineh, E. Drouet et al., Genetic Determinants of Response to Clopidogrel and Cardiovascular Events, New England Journal of Medicine, vol.360, issue.4, pp.363-375, 2009.
DOI : 10.1056/NEJMoa0808227

P. Christia and N. Frangogiannis, Targeting inflammatory pathways in myocardial infarction, European Journal of Clinical Investigation, vol.87, issue.9, pp.986-995, 2013.
DOI : 10.1161/01.CIR.87.2.526

G. Fröhlich, P. Meier, S. White, D. Yellon, and D. Hausenloy, Myocardial reperfusion injury: looking beyond primary PCI, European Heart Journal, vol.55, issue.23, pp.1714-1722, 2013.
DOI : 10.1016/j.jacc.2009.10.052

F. Arslan, D. De-kleijn, L. Timmers, P. Doevendans, and G. Pasterkamp, Bridging Innate Immunity and Myocardial Ischemia/Reperfusion Injury: The Search for Therapeutic Targets, Current Pharmaceutical Design, vol.14, issue.12, pp.1205-1216, 2008.
DOI : 10.2174/138161208784246090

F. Arslan, D. De-kleijn, and G. Pasterkamp, Innate immune signaling in cardiac ischemia, Nature Reviews Cardiology, vol.182, issue.5, pp.292-300, 2011.
DOI : 10.4049/jimmunol.0803842

Y. Feng and W. Chao, Toll-Like Receptors and Myocardial Inflammation, International Journal of Inflammation, vol.169, issue.6, p.170352, 2011.
DOI : 10.1097/ALN.0b013e31820a4d78

S. Gibot, C. Alauzet, F. Massin, N. Sennoune, G. Faure et al., Modulation of the Triggering Receptor Expressed on Myeloid Cells???1 Pathway during Pneumonia in Rats, The Journal of Infectious Diseases, vol.194, issue.7, pp.975-983, 2006.
DOI : 10.1086/506950

K. Kamei, T. Yasuda, T. Ueda, F. Qiang, Y. Takeyama et al., Role of triggering receptor expressed on myeloid cells-1 in experimental severe acute pancreatitis, Journal of Hepato-Biliary-Pancreatic Sciences, vol.187, issue.3, pp.305-312, 2010.
DOI : 10.1086/374754

S. Gibot, F. Massin, C. Alauzet, M. Derive, C. Montemont et al., EFFECTS OF THE TREM 1 PATHWAY MODULATION DURING HEMORRHAGIC SHOCK IN RATS, Shock, vol.32, issue.6, p.633, 2009.
DOI : 10.1097/SHK.0b013e3181a53842

M. Schenk, A. Bouchon, F. Seibold, and C. Mueller, TREM-1???expressing intestinal macrophages crucially amplify chronic inflammation in experimental colitis and inflammatory bowel diseases, Journal of Clinical Investigation, vol.117, issue.10, pp.3097-3106, 2007.
DOI : 10.1172/JCI30602DS1

C. Collins, D. La, H. Yang, F. Massin, S. Gibot et al., Elevated synovial expression of triggering receptor expressed on myeloid cells 1 in patients with septic arthritis or rheumatoid arthritis, Annals of the Rheumatic Diseases, vol.68, issue.11, pp.1768-1774, 2009.
DOI : 10.1136/ard.2008.089557

C. Bisson, F. Massin, P. Lefevre, N. Thilly, N. Miller et al., Increased gingival crevicular fluid levels of soluble triggering receptor expressed on myeloid cells (sTREM) -1 in severe periodontitis, Journal of Clinical Periodontology, vol.36, issue.53, pp.1141-1148, 2012.
DOI : 10.1097/CCM.0b013e31817b8824

R. Roberts, V. Demello, and B. Sobel, Deleterious effects of methylprednisolone in patients with myocardial infarction, Circulation, vol.53, pp.204-206, 1976.

O. How, A. Røsner, A. Kildal, T. Stenberg, P. Gjessing et al., Dobutamine-norepinephrine, but not vasopressin, restores the ventriculoarterial matching in experimental cardiogenic shock, Translational Research, vol.156, issue.5, pp.273-281, 2010.
DOI : 10.1016/j.trsl.2010.07.011

F. Mccall, K. Telukuntla, V. Karantalis, V. Suncion, A. Heldman et al., Myocardial infarction and intramyocardial injection models in swine, Nature Protocols, vol.3, issue.8, pp.1479-1496, 2012.
DOI : 10.1016/S0735-1097(99)00126-6

R. Fincke, J. Hochman, A. Lowe, V. Menon, J. Slater et al., Cardiac power is the strongest hemodynamic correlate of mortality in cardiogenic shock: A report from the SHOCK trial registry, Journal of the American College of Cardiology, vol.44, issue.2, pp.340-348, 2004.
DOI : 10.1016/j.jacc.2004.03.060

A. Abbate, B. Van-tassell, G. Biondi-zoccai, M. Kontos, J. Grizzard et al., Effects of Interleukin-1 Blockade With Anakinra on Adverse Cardiac Remodeling and Heart Failure After Acute Myocardial Infarction [from the Virginia Commonwealth University-Anakinra Remodeling Trial (2) (VCU-ART2) Pilot Study], Effects of Interleukin-1 Blockade With Anakinra on Adverse Cardiac Remodeling and Heart Failure After Acute Myocardial Infarction [from the Virginia Commonwealth University-Anakinra Remodeling TrialVCU-ART2) Pilot Study], pp.1394-1400, 2013.
DOI : 10.1016/j.amjcard.2013.01.287

M. Dirksen, G. Laarman, M. Simoons, and D. Duncker, Reperfusion injury in humans: A review of clinical trials on reperfusion injury inhibitory strategies, Cardiovascular Research, vol.74, issue.3, pp.343-355, 2007.
DOI : 10.1016/j.cardiores.2007.01.014

N. Webster and H. Galley, Immunomodulation in the critically ill, British Journal of Anaesthesia, vol.103, issue.1, pp.70-81, 2009.
DOI : 10.1093/bja/aep128

T. Kempf, A. Zarbock, D. Vestweber, and K. Wollert, Anti-inflammatory mechanisms and therapeutic opportunities in myocardial infarct healing, Journal of Molecular Medicine, vol.55, issue.4, pp.361-369, 2012.
DOI : 10.1016/j.jacc.2009.06.059

W. Xiao, M. Mindrinos, J. Seok, J. Cuschieri, A. Cuenca et al., A genomic storm in critically injured humans, The Journal of Experimental Medicine, vol.61, issue.13, pp.2581-2590, 2011.
DOI : 10.1038/nature08780

J. Jordan, Z. Zhao, and J. Vinten-johansen, The role of neutrophils in myocardial ischemia???reperfusion injury, Cardiovascular Research, vol.43, issue.4, pp.860-878, 1999.
DOI : 10.1016/S0008-6363(99)00187-X

H. Jaeschke and C. Smith, Mechanisms of neutrophil-induced parenchymal cell injury, Journal of Leukocyte Biology, vol.61, issue.6, pp.647-653, 1997.
DOI : 10.1002/jlb.61.6.647

S. Tavener, E. Long, S. Robbins, K. Mcrae, H. Van-remmen et al., Immune Cell Toll-Like Receptor 4 Is Required for Cardiac Myocyte Impairment During Endotoxemia, Circulation Research, vol.95, issue.7, pp.700-707, 2004.
DOI : 10.1161/01.RES.0000144175.70140.8c

URL : http://circres.ahajournals.org/content/circresaha/95/7/700.full.pdf

V. Braunersreuther and V. Jaquet, Reactive Oxygen Species in Myocardial Reperfusion Injury: From Physiopathology to Therapeutic Approaches, Current Pharmaceutical Biotechnology, vol.13, issue.1, pp.97-114, 2012.
DOI : 10.2174/138920112798868782

G. Blaudszun, M. Licker, and D. Morel, : a sensitive, continuous, load-independent contractility index, Experimental Physiology, vol.287, issue.10, pp.1446-1456, 2013.
DOI : 10.1152/ajpheart.00624.2003

URL : http://onlinelibrary.wiley.com/doi/10.1113/expphysiol.2013.073833/pdf