X. Bibliographie-]-d, P. Annane, M. C. Aegerter, E. B. Jars-guincestre, . Guidet-]-s et al., « Ventricular myocyte caspases are directly responsible for endotoxininduced cardiac dysfunction « Surviving Sepsis Campaign: international guidelines for management of severe sepsis and septic shock « Efficacy and safety of recombinant human activated protein C for severe sepsis « Definitions for sepsis and organ failure and guidelines for the use of innovative therapies in sepsis reappraisal of the epidemiology and outcome of severe sepsis in French intensive care units, Current epidemiology of septic shock: the CUB-Réa Network The ACCP/SCCM Consensus Conference Committee. American College of Chest Physicians/Society of Critical Care Medicine », ChestSIS International Sepsis Definitions Conference Profound but reversible myocardial depression in patients with septic shock », Annals of Internal Medicine, pp.165-172, 1984.

R. Moreno, The use of maximum SOFA score to quantify organ dysfunction/failure in intensive care. Results of a prospective, multicentre study, Intensive Care Medicine, vol.25, issue.7, pp.686-696, 1999.
DOI : 10.1007/s001340050931

A. Kumar, C. Haery, and J. E. Parrillo, MYOCARDIAL DYSFUNCTION IN SEPTIC SHOCK, Critical Care Clinics, vol.16, issue.2, pp.251-287, 2000.
DOI : 10.1016/S0749-0704(05)70110-X

K. Wei and S. Kaul, The coronary microcirculation in health and disease, Cardiology Clinics, vol.22, issue.2, pp.221-231, 2004.
DOI : 10.1016/j.ccl.2004.02.005

A. B. Groeneveld, A. A. Van-lambalgen, G. C. Van-den-bos, W. Bronsveld, J. J. Nauta et al., Maldistribution of heterogeneous coronary blood flow during canine endotoxin shock, Cardiovascular Research, vol.25, issue.1, pp.80-88, 1991.
DOI : 10.1093/cvr/25.1.80

M. A. Solomon, Myocardial energy metabolism and morphology in a canine model of sepsis, American Journal of Physiology-Heart and Circulatory Physiology, vol.266, issue.2, pp.757-768, 1994.
DOI : 10.1152/ajpheart.1994.266.2.H757

W. S. Madorin, T. Rui, N. Sugimoto, O. Handa, G. Cepinskas et al., Cardiac Myocytes Activated by Septic Plasma Promote Neutrophil Transendothelial Migration: Role of Platelet-Activating Factor and the Chemokines LIX and KC, Circulation Research, vol.94, issue.7, pp.944-951, 2004.
DOI : 10.1161/01.RES.0000124395.20249.AE

F. Chagnon, M. Bentourkia, R. Lecomte, and M. Lessard, Endotoxin-induced heart dysfunction in rats: Assessment of myocardial perfusion and permeability and the role of fluid resuscitation*, Critical Care Medicine, vol.34, issue.1, pp.127-133, 2006.
DOI : 10.1097/01.CCM.0000190622.02222.DF

C. J. Fernandes-júnior, M. Iervolino, R. A. Neves, E. L. Sampaio, and E. E. Knobel, Interstitial myocarditis in sepsis, The American Journal of Cardiology, vol.74, issue.9, p.958, 1994.
DOI : 10.1016/0002-9149(94)90597-5

G. Schlag, H. Redl, C. J. Van-vuuren, and E. J. Davies, « Hyperdynamic sepsis in baboons: II. Relation of organ damage to severity of sepsis evaluated by a newly developed morphological scoring system, Circulatory Shock, vol.38, issue.4, pp.253-263, 1992.

M. Hersch, A. A. Gnidec, A. D. Bersten, M. Troster, F. S. Rutledge et al., Histologic and ultrastructural changes in nonpulmonary organs during early hyperdynamic sepsis, Surgery, vol.107, issue.4, pp.397-410, 1990.

L. Gotloib, A. Shostak, P. Galdi, and J. Jaichenko, Fudin, « Loss of microvascular negative charges accompanied by interstitial edema in septic rats' heart », Circulatory Shock, vol.36, issue.1, pp.45-56, 1992.

J. E. Langenfeld, G. W. Machiedo, M. Lyons, B. F. Rush, G. Dikdan et al., « Correlation between red blood cell deformability and changes in hemodynamic function, Surgery, vol.116, issue.5, pp.859-867, 1994.

D. M. Bers, Cardiac excitation???contraction coupling, Nature, vol.272, issue.6868, pp.198-205, 2002.
DOI : 10.1161/hh1701.095716

H. Fauvel, P. Marchetti, C. Chopin, P. Formstecher, and E. R. Nevière, Differential effects of caspase inhibitors on endotoxin-induced myocardial dysfunction and heart apoptosis, American Journal of Physiology-Heart and Circulatory Physiology, vol.10, issue.4, pp.1608-1614, 2001.
DOI : 10.1161/01.CIR.97.3.276

R. Nevière, H. Fauvel, C. Chopin, P. Formstecher, and E. P. Marchetti, Caspase Inhibition Prevents Cardiac Dysfunction and Heart Apoptosis in a Rat Model of Sepsis, American Journal of Respiratory and Critical Care Medicine, vol.160, issue.1, pp.218-225, 2001.
DOI : 10.1073/pnas.96.24.13795

T. E. Mcdonald, M. N. Grinman, C. M. Carthy, and K. R. Walley, Endotoxin infusion in rats induces apoptotic and survival pathways in hearts, American Journal of Physiology-Heart and Circulatory Physiology, vol.38, issue.5, pp.2053-2061, 2000.
DOI : 10.1006/jmcc.1997.0614

K. L. Laugwitz, Blocking Caspase-Activated Apoptosis Improves Contractility in Failing Myocardium, Human Gene Therapy, vol.12, issue.17, pp.2051-2063, 2001.
DOI : 10.1089/10430340152677403

A. Moretti, Essential myosin light chain as a target for caspase-3 in failing myocardium, Proceedings of the National Academy of Sciences of the United States of America, pp.11860-11865, 2002.
DOI : 10.1073/pnas.092022999

C. Communal, M. Sumandea, P. De-tombe, J. Narula, R. J. Solaro et al., Functional consequences of caspase activation in cardiac myocytes, Proceedings of the National Academy of Sciences of the United States of America, pp.6252-6256, 2002.
DOI : 10.1016/S0008-6363(99)00268-0

B. L. Schwab, Cleavage of plasma membrane calcium pumps by caspases: a link between apoptosis and necrosis, Cell Death & Differentiation, vol.261, issue.8, pp.818-831, 2002.
DOI : 10.1074/jbc.270.24.14643

A. Lepple-wienhues, Stimulation of CD95 (Fas) blocks T lymphocyte calcium channels through sphingomyelinase and sphingolipids, Proceedings of the National Academy of Sciences of the United States of America, pp.13795-13800, 1999.
DOI : 10.1084/jem.180.4.1547

S. M. Hassoun, Prevention of endotoxin-induced sarcoplasmic reticulum calcium leak improves mitochondrial and myocardial dysfunction*, Critical Care Medicine, vol.36, issue.9, pp.2590-2596, 2008.
DOI : 10.1097/CCM.0b013e3181844276

R. Neviere, Caspase-dependent protein phosphatase 2A activation contributes to endotoxin-induced cardiomyocyte contractile dysfunction*, Critical Care Medicine, vol.38, issue.10, pp.2031-2036, 2010.
DOI : 10.1097/CCM.0b013e3181eedafb

S. M. Hassoun, Sphingosine impairs mitochondrial function by opening permeability transition pore, Mitochondrion, vol.6, issue.3, pp.149-154, 2006.
DOI : 10.1016/j.mito.2006.05.001

M. P. Fink and . Cytopathic-hypoxia, Cytopathic hypoxia: Is oxygen use impaired in sepsis as a result of an acquired intrinsic derangement in cellular respiration?, Critical Care Clinics, vol.18, issue.1, pp.165-175, 2002.
DOI : 10.1016/S0749-0704(03)00071-X

D. Brealey, Association between mitochondrial dysfunction and severity and outcome of septic shock, The Lancet, vol.360, issue.9328, pp.219-223, 2002.
DOI : 10.1016/S0140-6736(02)09459-X

D. Brealey, Mitochondrial dysfunction in a long-term rodent model of sepsis and organ failure, American Journal of Physiology-Regulatory, Integrative and Comparative Physiology, vol.286, issue.3, pp.491-497, 2004.
DOI : 10.1016/S0005-2728(99)00031-6

E. D. Crouser, Mitochondrial dysfunction in septic shock and multiple organ dysfunction syndrome, Mitochondrion, vol.4, issue.5-6, pp.5-6, 2004.
DOI : 10.1016/j.mito.2004.07.023

E. D. Crouser, M. W. Julian, D. V. Blaho, D. R. Pfeiffer, and . Endotoxin, Endotoxin-induced mitochondrial damage correlates with impaired respiratory activity, Critical Care Medicine, vol.30, issue.2, pp.276-284, 2002.
DOI : 10.1097/00003246-200202000-00002

J. A. Watts, J. A. Kline, L. R. Thornton, R. M. Grattan, and S. S. Brar, Metabolic dysfunction and depletion of mitochondria in hearts of septic rats, Journal of Molecular and Cellular Cardiology, vol.36, issue.1, pp.141-150, 2004.
DOI : 10.1016/j.yjmcc.2003.10.015

F. G. Soriano, Potential role of poly(adenosine 5???-diphosphate-ribose) polymerase activation in the pathogenesis of myocardial contractile dysfunction associated with human septic shock, Critical Care Medicine, vol.34, issue.4, pp.1073-1079, 2006.
DOI : 10.1097/01.CCM.0000206470.47721.8D

H. B. Suliman, M. S. Carraway, K. E. Welty-wolf, A. R. Whorton, and C. A. Piantadosi, Lipopolysaccharide Stimulates Mitochondrial Biogenesis via Activation of Nuclear Respiratory Factor-1, Journal of Biological Chemistry, vol.67, issue.42, pp.42-41510, 2003.
DOI : 10.1074/jbc.272.30.18732

H. B. Suliman, K. E. Welty-wolf, M. Carraway, L. Tatro, and C. A. Piantadosi, Lipopolysaccharide induces oxidative cardiac mitochondrial damage and biogenesis, Lipopolysaccharide induces oxidative cardiac mitochondrial damage and biogenesis, pp.279-288, 2004.
DOI : 10.1016/j.cardiores.2004.07.005

C. Batandier, E. Fontaine, C. Kériel, and X. M. Leverve, Determination of mitochondrial reactive oxygen species: methodological aspects, Journal of Cellular and Molecular Medicine, vol.126, issue.2, pp.175-187, 2002.
DOI : 10.1016/S0891-5849(99)00061-1

URL : https://hal.archives-ouvertes.fr/inserm-00389962

P. Ferdinandy, H. Danial, I. Ambrus, R. A. Rothery, and E. R. Schulz, Peroxynitrite Is a Major Contributor to Cytokine-Induced Myocardial Contractile Failure, Circulation Research, vol.87, issue.3, pp.241-247, 2000.
DOI : 10.1161/01.RES.87.3.241

M. G. Vila-petroff, A. Younes, J. Egan, E. G. Lakatta, and S. J. Sollott, Activation of Distinct cAMP-Dependent and cGMP-Dependent Pathways by Nitric Oxide in Cardiac Myocytes, Circulation Research, vol.84, issue.9, pp.1020-1031, 1999.
DOI : 10.1161/01.RES.84.9.1020

R. A. Kelly, J. L. Balligand, and T. W. Smith, Nitric Oxide and Cardiac Function, Nitric oxide and cardiac function, pp.363-380, 1996.
DOI : 10.1161/01.RES.79.3.363

M. S. Finkel, C. V. Oddis, T. D. Jacob, S. C. Watkins, B. G. Hattler et al., Negative inotropic effects of cytokines on the heart mediated by nitric oxide, Negative inotropic effects of cytokines on the heart mediated by nitric oxide, pp.387-389, 1992.
DOI : 10.1126/science.1631560

A. Kumar, Role of nitric oxide and cGMP in human septic serum-induced depression of cardiac myocyte contractility, American Journal of Physiology-Regulatory, Integrative and Comparative Physiology, vol.268, issue.37, pp.265-276, 1999.
DOI : 10.1172/JCI116834

K. Sugishita, Cellular Basis for the Acute Inhibitory Effects of IL-6 and TNF- ?? on Excitation-contraction Coupling, Journal of Molecular and Cellular Cardiology, vol.31, issue.8, pp.1457-1467, 1999.
DOI : 10.1006/jmcc.1999.0989

D. J. Pinsky, B. Cai, X. Yang, C. Rodriguez, R. R. Sciacca et al., The lethal effects of cytokine-induced nitric oxide on cardiac myocytes are blocked by nitric oxide synthase antagonism or transforming growth factor beta., Journal of Clinical Investigation, vol.95, issue.2, pp.677-685, 1995.
DOI : 10.1172/JCI117713

G. Kreymann, S. Grosser, P. Buggisch, C. Gottschall, S. Matthaei et al., Oxygen consumption and resting metabolic rate in sepsis, sepsis syndrome, and septic shock, Critical Care Medicine, vol.21, issue.7, pp.1012-1019, 1993.
DOI : 10.1097/00003246-199307000-00015

A. Rudiger and M. Singer, Mechanisms of sepsis-induced cardiac dysfunction, Critical Care Medicine, vol.35, issue.6, pp.1599-1608, 2007.
DOI : 10.1097/01.CCM.0000266683.64081.02

P. Boekstegers, S. Weidenhöfer, and T. Kapsner, Skeletal muscle partial pressure of oxygen in patients with sepsis, Critical Care Medicine, vol.22, issue.4, pp.640-650, 1994.
DOI : 10.1097/00003246-199404000-00021

P. Boekstegers, S. Weidenhöfer, G. Pilz, and E. K. Werdan, Periphere Sauerstoffverf??gbarkeit im Skelettmuskel bei Sepsis und septischem Schock: Vergleich mit begrenzter Infektion und kardiogenem Schock, Infection, vol.24, issue.5, pp.317-323, 1991.
DOI : 10.1007/978-1-4684-1188-1_42

R. E. Cunnion, G. L. Schaer, M. M. Parker, C. Natanson, and J. E. Parrillo, The coronary circulation in human septic shock, Circulation, vol.73, issue.4, pp.637-644, 1986.
DOI : 10.1161/01.CIR.73.4.637

P. Rossi, L. Tauzin, M. Weiss, J. Rostain, and J. Sainty, Could hyperoxic ventilation impair oxygen delivery in septic patients?, Clinical Physiology and Functional Imaging, vol.50, issue.3, pp.180-184, 2007.
DOI : 10.1097/00005344-200308000-00014

J. F. Dhainaut, Coronary hemodynamics and myocardial metabolism of lactate, free fatty acids, glucose, and ketones in patients with septic shock, Circulation, vol.75, issue.3, pp.533-541, 1987.
DOI : 10.1161/01.CIR.75.3.533

R. J. Levy, Evidence of myocardial hibernation in the septic heart*, Critical Care Medicine, vol.33, issue.12, pp.2752-2756, 2005.
DOI : 10.1097/01.CCM.0000189943.60945.77

L. H. Opie, Cellular Basis for Therapeutic Choices in Heart Failure, Circulation, vol.110, issue.17, pp.2559-2561, 2004.
DOI : 10.1161/01.CIR.0000146803.14063.F7

D. Annane, Inappropriate Sympathetic Activation at Onset of Septic Shock, American Journal of Respiratory and Critical Care Medicine, vol.17, issue.2, pp.458-465, 1999.
DOI : 10.1161/01.CIR.95.6.1449

J. K. Bocking, W. J. Sibbald, R. L. Holliday, and S. Scott, Viidik, « Plasma catecholamine levels and pulmonary dysfunction in sepsis, Surgery, Gynecology & Obstetrics, vol.148, issue.5, pp.715-719, 1979.

G. Bernardin, F. Tiger, R. Fouché, and E. M. Mattéi, Continuous noninvasive measurement of aortic blood flow in critically ill patients with a new esophageal echo-Doppler system, Journal of Critical Care, vol.13, issue.4, pp.177-183, 1998.
DOI : 10.1016/S0883-9441(98)90003-X

M. Iwase, Cardiac functional and structural alterations induced by endotoxin in rats: Importance of platelet-activating factor, Critical Care Medicine, vol.29, issue.3, pp.609-617, 2001.
DOI : 10.1097/00003246-200103000-00025

P. Y. Hahn, P. Wang, S. M. Tait, Z. F. Ba, S. S. Reich et al., SUSTAINED ELEVATION IN CIRCULATING CATECHOLAMINE LEVELS DURING POLYMICROBIAL SEPSIS, Sustained elevation in circulating catecholamine levels during polymicrobial sepsis, pp.269-273, 1995.
DOI : 10.1097/00024382-199510000-00007

J. M. Hare, E. Loh, M. A. Creager, and W. S. Colucci, Nitric Oxide Inhibits the Positive Inotropic Response to ??-Adrenergic Stimulation in Humans With Left Ventricular Dysfunction, Circulation, vol.92, issue.8, pp.2198-2203, 1995.
DOI : 10.1161/01.CIR.92.8.2198

E. Barth, P. Radermacher, C. Thiemermann, S. Weber, and M. Georgieff, Role of inducible nitric oxide synthase in the reduced responsiveness of the myocardium to catecholamines in a hyperdynamic, murine model of septic shock*, Critical Care Medicine, vol.34, issue.2, pp.307-313, 2006.
DOI : 10.1097/01.CCM.0000199070.46812.21

N. H. Ismail, E. J. Cohn, and D. L. Mollitt, « Nitric oxide synthase inhibition negates septic-induced alterations in cytoplasmic calcium homeostasis and membrane dynamics », The American Surgeon, pp.20-23, 1997.

C. Tang and M. S. Liu, « Initial externalization followed by internalization of betaadrenergic receptors in rat heart during sepsis, The American Journal of Physiology, vol.270, issue.1, pp.254-263, 1996.

R. E. Shepherd, C. H. Lang, and K. H. Mcdonough, Myocardial adrenergic responsiveness after lethal and nonlethal doses of endotoxin, American Journal of Physiology-Heart and Circulatory Physiology, vol.252, issue.2, pp.410-416, 1987.
DOI : 10.1152/ajpheart.1987.252.2.H410

T. Gulick, M. K. Chung, S. J. Pieper, L. G. Lange, and G. F. Schreiner, Interleukin 1 and tumor necrosis factor inhibit cardiac myocyte beta-adrenergic responsiveness., Proceedings of the National Academy of Sciences of the United States of America, pp.6753-6757, 1989.
DOI : 10.1073/pnas.86.17.6753

URL : http://www.pnas.org/content/86/17/6753.full.pdf

N. Matsuda, Y. Hattori, Y. Akaishi, Y. Suzuki, O. Kemmotsu et al., Impairment of Cardiac ??-Adrenoceptor Cellular Signaling by Decreased Expression of Gs?? in Septic Rabbits, Anesthesiology, vol.93, issue.6, pp.1465-1473, 2000.
DOI : 10.1097/00000542-200012000-00019

M. Böhm, R. Kirchmayr, P. Gierschik, and E. E. Erdmann, Increase of myocardial inhibitory G-proteins in catecholamine-refractory septic shock or in septic multiorgan failure, The American Journal of Medicine, vol.98, issue.2, pp.183-186, 1995.
DOI : 10.1016/S0002-9343(99)80402-1

L. Wu, G Protein and Adenylate Cyclase Complex-Mediated Signal Transduction in the Rat Heart During Sepsis, Shock, vol.19, issue.6, pp.533-537, 2003.
DOI : 10.1097/01.shk.0000055816.40894.cd

. Abi-gerges, Endotoxin Injection in Rat, American Journal of Respiratory and Critical Care Medicine, vol.265, issue.4, pp.1196-1204, 1999.
DOI : 10.1056/NEJM199901213400307

J. E. Parrillo, C. Burch, J. H. Shelhamer, M. M. Parker, C. Natanson et al., A circulating myocardial depressant substance in humans with septic shock. Septic shock patients with a reduced ejection fraction have a circulating factor that depresses in vitro myocardial cell performance., Journal of Clinical Investigation, vol.76, issue.4, pp.1539-1553, 1985.
DOI : 10.1172/JCI112135

J. M. Reilly, R. E. Cunnion, C. Burch-whitman, M. M. Parker, J. H. Shelhamer et al., A Circulating Myocardial Depressant Substance is Associated with Cardiac Dysfunction and Peripheral Hypoperfusion (Lactic Acidemia) in Patients with Septic Shock, Chest, vol.95, issue.5, pp.1072-1080, 1989.
DOI : 10.1378/chest.95.5.1072

N. Pathan, C. Sandiford, S. E. Harding, and E. M. Levin, Characterization of a myocardial depressant factor in meningococcal septicemia*, Critical Care Medicine, vol.30, issue.10, pp.2191-2198, 2002.
DOI : 10.1097/00003246-200210000-00003

A. Kumar, V. Thota, L. Dee, J. Olson, E. Uretz et al., Tumor necrosis factor alpha and interleukin 1beta are responsible for in vitro myocardial cell depression induced by human septic shock serum, Journal of Experimental Medicine, vol.183, issue.3, pp.949-958, 1996.
DOI : 10.1084/jem.183.3.949

B. Vallet and E. Wiel, Endothelial cell dysfunction and coagulation, Endothelial cell dysfunction and coagulation, pp.36-41, 2001.
DOI : 10.1097/00003246-200107001-00015

F. Fourrier, « Septic shock, multiple organ failure, and disseminated intravascular coagulation. Compared patterns of antithrombin III, protein C, and protein S deficiencies, pp.816-823, 1992.
DOI : 10.1378/chest.101.3.816

A. C. Mavrommatis, Activation of the fibrinolytic system and utilization of the coagulation inhibitors in sepsis: comparison with severe sepsis and septic shock, Intensive Care Medicine, vol.27, issue.12, pp.1853-1859, 2001.
DOI : 10.1007/s00134-001-1139-8

E. De-jonge, « Tissue factor pathway inhibitor dose-dependently inhibits coagulation activation without influencing the fibrinolytic and cytokine response during human endotoxemia, pp.1124-1129, 2000.

M. Riewald, R. J. Petrovan, A. Donner, B. M. Mueller, and E. W. Ruf, Activation of Endothelial Cell Protease Activated Receptor 1 by the Protein C Pathway, Science, vol.296, issue.5574, pp.1880-1882, 2002.
DOI : 10.1126/science.1071699

D. E. Joyce, L. Gelbert, A. Ciaccia, B. Dehoff, and B. W. , Gene Expression Profile of Antithrombotic Protein C Defines New Mechanisms Modulating Inflammation and Apoptosis, Journal of Biological Chemistry, vol.95, issue.14, pp.11199-11203, 2001.
DOI : 10.1093/glycob/4.2.221

E. Bellissant and D. Annane, Effect of hydrocortisone on phenylephrine??? mean arterial pressure dose-response relationship in septic shock, Clinical Pharmacology & Therapeutics, vol.68, issue.3, pp.293-303, 2000.
DOI : 10.1067/mcp.2000.109354

T. M. Mckenna, Enhanced vascular effects of cyclic GMP in septic rat aorta, American Journal of Physiology-Regulatory, Integrative and Comparative Physiology, vol.254, issue.3, pp.436-442, 1988.
DOI : 10.1152/ajpregu.1988.254.3.R436

J. D. Macmicking, Altered responses to bacterial infection and endotoxic shock in mice lacking inducible nitric oxide synthase, Cell, vol.81, issue.4, pp.641-650, 1995.
DOI : 10.1016/0092-8674(95)90085-3

A. López, Multiple-center, randomized, placebo-controlled, double-blind study of the nitric oxide synthase inhibitor 546C88: Effect on survival in patients with septic shock*, Critical Care Medicine, vol.32, issue.1, pp.21-30, 2004.
DOI : 10.1097/01.CCM.0000105581.01815.C6

W. L. Smith, D. L. Dewitt, and R. M. Garavito, Cyclooxygenases: Structural, Cellular, and Molecular Biology, Cyclooxygenases: structural, cellular, and molecular biology, pp.145-182, 2000.
DOI : 10.1146/annurev.biochem.69.1.145

M. Adib-conquy and J. Cavaillon, Stress molecules in sepsis and systemic inflammatory response syndrome, FEBS Letters, vol.7, issue.19, pp.3723-3733, 2007.
DOI : 10.1016/S1473-3099(07)70052-X

M. Wanecek, E. Weitzberg, and A. Rudehill, The endothelin system in septic and endotoxin shock, European Journal of Pharmacology, vol.407, issue.1-2, pp.1-2, 2000.
DOI : 10.1016/S0014-2999(00)00675-0

S. Moncada and E. A. Higgs, Endogenous nitric oxide: physiology, pathology and clinical relevance, European Journal of Clinical Investigation, vol.8, issue.4, pp.361-374, 1991.
DOI : 10.1097/00000441-189305000-00001

D. M. Dudzinski and T. Michel, Life history of eNOS: Partners and pathways, Cardiovascular Research, vol.75, issue.2, pp.247-260, 2007.
DOI : 10.1016/j.cardiores.2007.03.023

M. Zhou, P. Wang, and I. H. Chaudry, Endothelial nitric oxide synthase is downregulated during hyperdynamic sepsis, Biochimica et Biophysica Acta (BBA) - General Subjects, vol.1335, issue.1-2, pp.1-2, 1997.
DOI : 10.1016/S0304-4165(96)00139-0

J. Leclerc, A single endotoxin injection in the rabbit causes prolonged blood vessel dysfunction and a procoagulant state, Critical Care Medicine, vol.28, issue.11, pp.3672-3678, 2000.
DOI : 10.1097/00003246-200011000-00023

K. Bhagat, R. Moss, J. Collier, and E. P. Vallance, stunning " following a brief exposure to endotoxin: a mechanism to link infection and infarction?, Cardiovascular Research, vol.32, issue.5, pp.822-829, 1996.

K. Bhagat and J. Collier, Local Venous Responses to Endotoxin in Humans, Circulation, vol.94, issue.3, pp.490-497, 1996.
DOI : 10.1161/01.CIR.94.3.490

R. A. Kloner, A. M. Hutter, J. T. Emmick, M. I. Mitchell, J. Denne et al., Time course of the interaction between tadalafil and nitrates, Journal of the American College of Cardiology, vol.42, issue.10, pp.1855-1860, 2003.
DOI : 10.1016/j.jacc.2003.09.023

M. J. Winn, B. Vallet, N. K. Asante, S. E. Curtis, and S. M. Cain, Effects of NG-substituted arginines on coronary vascular function after endotoxin, Effects of NGsubstituted arginines on coronary vascular function after endotoxin, pp.424-431, 1985.
DOI : 10.1152/jappl.1993.75.1.424

S. D. Chauhan, G. Seggara, P. A. Vo, R. J. Macallister, A. J. Hobbs et al., Protection against lipopolysaccharide-induced endothelial dysfunction in resistance and conduit vasculature of iNOS knockout mice, The FASEB Journal, vol.17, issue.6, pp.773-775, 2003.
DOI : 10.1096/fj.02-0668fje

M. Mcmillan, B. Chernow, and B. L. Roth, « Hepatic alpha 1-adrenergic receptor alteration in a rat model of chronic sepsis, Circulatory Shock, vol.19, issue.2, pp.185-193, 1986.

J. F. Buckley, M. Singer, and L. H. Clapp, Role of KATP channels in sepsis, Cardiovascular Research, vol.72, issue.2, pp.220-230, 2006.
DOI : 10.1016/j.cardiores.2006.07.011

E. Rivers, Early Goal-Directed Therapy in the Treatment of Severe Sepsis and Septic Shock, New England Journal of Medicine, vol.345, issue.19, pp.1368-1377, 2001.
DOI : 10.1056/NEJMoa010307

M. Leone, B. Vallet, J. Teboul, J. Mateo, O. Bastien et al., Survey of the use of catecholamines by French physicians, Survey of the use of catecholamines by French physicians, pp.984-988, 2004.
DOI : 10.1007/s00134-004-2172-1

I. J. Kopin, Catecholamine metabolism: basic aspects and clinical significance, Pharmacological Reviews, vol.37, issue.4, pp.333-364, 1985.

W. E. Clutter and P. E. Cryer, « Plasma dose-response studies with noradrenaline and adrenaline in man, Progress in Biochemical Pharmacology, vol.17, pp.84-89, 1980.

J. S. Gregory, M. F. Bonfiglio, J. F. Dasta, T. E. Reilley, M. C. Townsend et al., Experience with phenylephrine as a component of the pharmacologie support of septic shock, Critical Care Medicine, vol.19, issue.11, pp.1395-1400, 1991.
DOI : 10.1097/00003246-199111000-00016

H. Reinelt, Impact of exogenous beta-adrenergic receptor stimulation on hepatosplanchnic oxygen kinetics and metabolic activity in septic shock, Critical Care Medicine, vol.27, issue.2, pp.325-331, 1999.
DOI : 10.1097/00003246-199902000-00039

B. Levy, Bench-to-bedside review: Is there a place for epinephrine in septic shock?, Critical Care, vol.9, issue.6, pp.561-565, 2005.
DOI : 10.1186/cc3901

D. Annane, Norepinephrine plus dobutamine versus epinephrine alone for management of septic shock: a randomised trial, The Lancet, vol.370, issue.9588, pp.676-684, 2007.
DOI : 10.1016/S0140-6736(07)61344-0

A. R. Rezaie, Exosite-Dependent Regulation of the Protein C Anticoagulant Pathway, Trends in Cardiovascular Medicine, vol.13, issue.1, pp.8-15, 2003.
DOI : 10.1016/S1050-1738(02)00191-3

F. B. Taylor, G. T. Peer, M. S. Lockhart, G. Ferrell, and C. T. Esmon, Endothelial cell protein C receptor plays an important role in protein C activation in vivo, Blood, vol.97, issue.6, pp.1685-1688, 2001.
DOI : 10.1182/blood.V97.6.1685

S. N. Faust, R. S. Heyderman, and E. M. Levin, Coagulation in severe sepsis: A central role for thrombomodulin and activated protein C, Critical Care Medicine, vol.29, issue.7, pp.62-67, 2001.
DOI : 10.1097/00003246-200107001-00022

N. Sennoun, Recombinant human activated protein C improves endotoxemia-induced endothelial dysfunction: a blood-free model in isolated mouse arteries, American Journal of Physiology-Heart and Circulatory Physiology, vol.297, issue.1, pp.277-282, 2009.
DOI : 10.1016/j.thromres.2004.09.011

D. E. Joyce and B. W. , Recombinant human activated protein C attenuates the inflammatory response in endothelium and monocytes by modulating nuclear factor-??B, Critical Care Medicine, vol.30, issue.Supplement, pp.288-293, 2002.
DOI : 10.1097/00003246-200205001-00019

M. Brueckmann, Recombinant human activated protein C upregulates cyclooxygenase-2 expression in endothelial cells via binding to endothelial cell protein C receptor and activation of protease-activated receptor-1, Thrombosis and Haemostasis, vol.93, issue.4, pp.743-750, 2005.
DOI : 10.1160/TH04-08-0511

D. J. Stearns-kurosawa, Plasma levels of endothelial protein C receptor respond to anticoagulant treatment, Blood, vol.99, issue.2, pp.526-530, 2002.
DOI : 10.1182/blood.V99.2.526

T. Cheng, Activated protein C blocks p53-mediated apoptosis in ischemic human brain endothelium and is neuroprotective, Nature Medicine, vol.102, issue.3, pp.338-342, 2003.
DOI : 10.1172/JCI2029

D. S. Skundric, B. Bealmear, and R. P. Lisak, Induced upregulation of IL-1, IL-1RA and IL-1R type I gene expression by Schwann cells, Journal of Neuroimmunology, vol.74, issue.1-2, pp.1-2, 1997.
DOI : 10.1016/S0165-5728(96)00200-7

S. Stosi?-grujici? and M. L. Luki?, « Glucocorticoid-induced keratinocyte-derived interleukin-1 receptor antagonist(s), Immunology, vol.75, issue.2, pp.293-298, 1992.

C. Gabay, M. F. Smith, W. P. Eidlen, and . Arend, Interleukin 1 receptor antagonist (IL-1Ra) is an acute-phase protein., Journal of Clinical Investigation, vol.99, issue.12, pp.2930-2940, 1997.
DOI : 10.1172/JCI119488

T. Van-der-poll, A. E. Barber, S. M. Coyle, and S. F. Lowry, « Hypercortisolemia increases plasma interleukin-10 concentrations during human endotoxemia--a clinical research center study, The Journal of Clinical Endocrinology and Metabolism, vol.81, issue.10, pp.3604-3606, 1996.

A. Marchant, Methylprednisolone differentially regulates IL-10 and tumour necrosis factor (TNF) production during murine endotoxaemia, Clinical and Experimental Immunology, vol.106, issue.1, pp.91-96, 1996.
DOI : 10.1046/j.1365-2249.1996.d01-799.x

T. M. Mckenna, Prolonged exposure of rat aorta to low levels of endotoxin in vitro results in impaired contractility. Association with vascular cytokine release., Journal of Clinical Investigation, vol.86, issue.1, pp.160-168, 1990.
DOI : 10.1172/JCI114679

F. Buttgereit, I. Brink, B. Thiele, G. R. Burmester, F. Hiepe et al., « Effects of methylprednisolone and 21-aminosteroids on mitogen-induced interleukin-6 and tumor necrosis factor-alpha production in human peripheral blood mononuclear cells, The Journal of Pharmacology and Experimental Therapeutics, vol.275, issue.2, pp.850-853, 1995.

S. K. Arya, F. Wong-staal, and R. C. Gallo, « Dexamethasone-mediated inhibition of human T cell growth factor and gamma-interferon messenger RNA », Journal of Immunology, vol.133, issue.1, pp.273-276, 1950.

M. John, Inhaled Corticosteroids Increase Interleukin-10 but Reduce Macrophage Inflammatory Protein-1 ?? , Granulocyte-Macrophage Colony-stimulating Factor, and Interferon- ?? Release from Alveolar Macrophages in Asthma, American Journal of Respiratory and Critical Care Medicine, vol.153, issue.1, pp.256-262, 1998.
DOI : 10.1146/annurev.iy.11.040193.001121

V. Brinkmann and C. Kristofic, « Regulation by corticosteroids of Th1 and Th2 cytokine production in human CD4+ effector T cells generated from CD45RO-and CD45RO+ subsets, Journal of Immunology, vol.155, issue.7, pp.3322-3328, 1950.

T. Calandra, MIF as a glucocorticoid-induced modulator of cytokine production, Nature, vol.377, issue.6544, pp.68-71, 1995.
DOI : 10.1038/377068a0

J. Wu, F. Q. Cunha, F. Y. Liew, and W. Y. Weiser, « IL-10 inhibits the synthesis of migration inhibitory factor and migration inhibitory factor-mediated macrophage activation, Journal of Immunology, vol.151, issue.8, pp.4325-4332, 1950.

Q. Zhu and S. Solomon, Isolation and mode of action of rabbit corticostatic (antiadrenocorticotropin) peptides, Endocrinology, vol.130, issue.3, pp.1413-1423, 1992.

M. Perretti and R. J. Flower, « Modulation of IL-1-induced neutrophil migration by dexamethasone and lipocortin 1 », Journal of Immunology, vol.150, issue.3, pp.992-999, 1950.

J. M. Bailey, A. N. Makheja, J. Pash, and E. M. Verma, Corticosteroids suppress cyclooxygenase messenger RNA levels and prostanoid synthesis in cultured vascular cells, Biochemical and Biophysical Research Communications, vol.157, issue.3, pp.1159-1163, 1988.
DOI : 10.1016/S0006-291X(88)80995-1

M. W. Radomski, R. M. Palmer, and E. S. Moncada, Glucocorticoids inhibit the expression of an inducible, but not the constitutive, nitric oxide synthase in vascular endothelial cells., Proceedings of the National Academy of Sciences of the United States of America, pp.10043-10047, 1990.
DOI : 10.1073/pnas.87.24.10043

P. J. Barnes, Anti-inflammatory Actions of Glucocorticoids: Molecular Mechanisms, Clinical Science, vol.94, issue.6, pp.557-572, 1979.
DOI : 10.1042/cs0940557

M. Hochstrasser, Protein Degradation or Regulation: Ub the Judge, Cell, vol.84, issue.6, pp.813-815, 1996.
DOI : 10.1016/S0092-8674(00)81058-2

D. Annane and J. Cavaillon, Corticosteroids in Sepsis: From Bench to Bedside?, Shock, vol.20, issue.3, pp.197-207, 2003.
DOI : 10.1097/01.shk.0000079423.72656.2f

J. P. Grünfeld and L. Eloy, Glucocorticoids modulate vascular reactivity in the rat, Hypertension, vol.10, issue.6, pp.608-618, 1987.
DOI : 10.1161/01.HYP.10.6.608

S. M. Gardiner, P. A. Kemp, J. E. March, and E. T. Bennett, Regional haemodynamic responses to infusion of lipopolysaccharide in conscious rats: effects of pre- or post-treatment with glibenclamide, British Journal of Pharmacology, vol.114, issue.8, pp.1772-1778, 1999.
DOI : 10.1111/j.1476-5381.1995.tb13343.x

B. Levy, Vascular hyporesponsiveness to vasopressors in septic shock: from bench to bedside, Intensive Care Medicine, vol.345, issue.12, pp.2019-2029, 2010.
DOI : 10.1161/01.CIR.95.5.1122

URL : https://hal.archives-ouvertes.fr/hal-00666836

A. Steiner, R. Locher, A. Sachinidis, and E. W. Vetter, Cortisol-stimulated phosphoinositide metabolism in vascular smooth muscle cells: a role for glucocorticoids in blood pressure control?, Journal of Hypertension, vol.7, issue.6, pp.140-141, 1989.
DOI : 10.1097/00004872-198900076-00066

P. M. Williamson, J. L. Kohlhagen, G. J. Mangos, J. A. Whitworth, and J. J. Kelly, Acute effects of hydrocortisone on plasma nitrate/nitrite activity and forearm vasodilator responsiveness in normal human subjects, Clinical and Experimental Pharmacology and Physiology, vol.27, issue.3, pp.162-166, 2005.
DOI : 10.1046/j.1440-1681.2000.03354.x

K. Bhagat and J. Collier, Local Venous Responses to Endotoxin in Humans, Circulation, vol.94, issue.3, pp.490-497, 1996.
DOI : 10.1161/01.CIR.94.3.490

D. Annane, Impaired pressor sensitivity to noradrenaline in septic shock patients with and without impaired adrenal function reserve, British Journal of Clinical Pharmacology, vol.329, issue.6, pp.589-597, 1998.
DOI : 10.1056/NEJM199312303292706

D. Annane, Effect of Treatment With Low Doses of Hydrocortisone and Fludrocortisone on Mortality in Patients With Septic Shock, JAMA, vol.288, issue.7, pp.862-871, 2002.
DOI : 10.1001/jama.288.7.862

P. E. Bollaert, C. Charpentier, B. Levy, M. Debouverie, and G. Audibert, Reversal of late septic shock with supraphysiologic doses of hydrocortisone, Critical Care Medicine, vol.26, issue.4, pp.645-650, 1998.
DOI : 10.1097/00003246-199804000-00010

J. Briegel, Stress doses of hydrocortisone reverse hyperdynamic septic shock, Critical Care Medicine, vol.27, issue.4, pp.723-732, 1999.
DOI : 10.1097/00003246-199904000-00025

D. Keh, Immunologic and Hemodynamic Effects of ???Low-Dose??? Hydrocortisone in Septic Shock, American Journal of Respiratory and Critical Care Medicine, vol.167, issue.4, pp.512-520, 2003.
DOI : 10.1001/archinte.1997.00440250031004

M. Oppert, Low-dose hydrocortisone improves shock reversal and reduces cytokine levels in early hyperdynamic septic shock*, Critical Care Medicine, vol.33, issue.11, pp.2457-2464, 2005.
DOI : 10.1097/01.CCM.0000186370.78639.23

C. M. Bamberger, A. M. Bamberger, M. De-castro, and G. P. Chrousos, Glucocorticoid receptor beta, a potential endogenous inhibitor of glucocorticoid action in humans., Journal of Clinical Investigation, vol.95, issue.6, pp.2435-2441, 1995.
DOI : 10.1172/JCI117943

I. Grad and D. Picard, The glucocorticoid responses are shaped by molecular chaperones, Molecular and Cellular Endocrinology, vol.275, issue.1-2, pp.2-12, 2007.
DOI : 10.1016/j.mce.2007.05.018

M. J. Tsai and B. W. Malley, Molecular Mechanisms of Action of Steroid/Thyroid Receptor Superfamily Members, Annual Review of Biochemistry, vol.63, issue.1, pp.451-486, 1994.
DOI : 10.1146/annurev.bi.63.070194.002315

N. Auphan, J. A. Didonato, C. Rosette, A. Helmberg, and E. M. Karin, Immunosuppression by Glucocorticoids: Inhibition of NF-kappaB Activity Through Induction of IkappaB Sy nthesis, Science, vol.270, issue.5234, pp.286-290, 1995.
DOI : 10.1126/science.270.5234.286

R. I. Scheinman, P. C. Cogswell, A. K. Lofquist, and A. S. Baldwin, Role of Transcriptional Activation of IkappaBalpha in Mediation of Immunosuppression by Glucocorticoi ds, Science, vol.270, issue.5234, pp.283-286, 1995.
DOI : 10.1126/science.270.5234.283

S. Heck, K. Bender, M. Kullmann, M. Göttlicher, P. Herrlich et al., Ikappa Balpha -independent downregulation of NF-kappa B activity by glucocorticoid receptor, The EMBO Journal, vol.16, issue.15, pp.4698-4707, 1997.
DOI : 10.1093/emboj/16.15.4698

A. Dostert and T. Heinzel, « Negative glucocorticoid receptor response elements and their role in glucocorticoid action, Current Pharmaceutical Design, vol.10, issue.23, pp.2807-2816, 2004.

P. R. Mittelstadt and J. D. , Ashwell, « Inhibition of AP-1 by the glucocorticoid-inducible protein GILZ, The Journal of Biological Chemistry, vol.276, pp.31-29603, 2001.

F. Buttgereit, K. G. Saag, M. Cutolo, J. A. Da-silva, and J. W. Bijlsma, The molecular basis for the effectiveness, toxicity, and resistance to glucocorticoids: focus on the treatment of rheumatoid arthritis, Scandinavian Journal of Rheumatology, vol.2001, issue.81, pp.14-21, 2005.
DOI : 10.1002/1097-4644(20010401)81:1<149::AID-JCB1031>3.0.CO;2-W

B. Bartholome, Membrane glucocorticoid receptors (mGCR) are expressed in normal human peripheral blood mononuclear cells and up-regulated after in vitro stimulation and in patients with rheumatoid arthritis, The FASEB Journal, vol.18, issue.1, pp.70-80, 2004.
DOI : 10.1016/S0039-128X(98)00097-X

M. Löwenberg, Rapid immunosuppressive effects of glucocorticoids mediated through Lck and Fyn, Blood, vol.106, issue.5, pp.1703-1710, 2005.
DOI : 10.1182/blood-2004-12-4790

J. D. Croxtall, Q. Choudhury, and R. J. Flower, Glucocorticoids act within minutes to inhibit recruitment of signalling factors to activated EGF receptors through a receptor-dependent, transcription-independent mechanism, British Journal of Pharmacology, vol.10, issue.2, pp.289-298, 2000.
DOI : 10.3181/00379727-217-44252

D. Annane, Corticosteroids in the Treatment of Severe Sepsis and Septic Shock in Adults, JAMA, vol.301, issue.22, pp.2362-2375, 2009.
DOI : 10.1001/jama.2009.815

M. Vinclair, Duration of adrenal inhibition following a??single dose of etomidate in critically ill patients, Intensive Care Medicine, vol.54, issue.4, pp.714-719, 2008.
DOI : 10.1007/s00134-007-0970-y

URL : https://hal.archives-ouvertes.fr/inserm-00398796

M. Deuschle, Steroid Synthesis Inhibition with Ketoconazole and its Effect upon the Regulation of the Hypothalamus???Pituitary???Adrenal System in Healthy Humans, Neuropsychopharmacology, vol.34, issue.2, pp.379-383, 2003.
DOI : 10.1016/S0022-3956(00)00016-9

M. Jäättelä, V. Ilvesmäki, R. Voutilainen, U. H. Stenman, and E. E. Saksela, Tumor Necrosis Factor as a Potent Inhibitor of Adrenocorticotropin-Induced Cortisol Production and Steroidogenic P450 Enzyme Gene Expression in Cultured Human Fetal Adrenal Cells*, Endocrinology, vol.128, issue.1, pp.623-629, 1991.
DOI : 10.1210/endo-128-1-623

M. Jäättelä, O. Carpén, U. H. Stenman, and E. E. Saksela, « Regulation of ACTH-induced steroidogenesis in human fetal adrenals by rTNF-alpha, Molecular and Cellular Endocrinology, vol.68, pp.2-3, 1990.

R. Natarajan, S. Ploszaj, R. Horton, and E. J. Nadler, Tumor Necrosis Factor and Interleukin-1 Are Potent Inhibitors of Angiotensin-II-Induced Aldosterone Synthesis*, Endocrinology, vol.125, issue.6, pp.3084-3089, 1989.
DOI : 10.1210/endo-125-6-3084

H. J. Van-leeuwen, E. C. Heezius, G. M. Dallinga, J. A. Van-strijp, J. Verhoef et al., Lipoprotein metabolism in patients with severe sepsis, Critical Care Medicine, vol.31, issue.5, pp.1359-1366, 2003.
DOI : 10.1097/01.CCM.0000059724.08290.51

P. H. Van-der-voort, R. T. Gerritsen, A. J. Bakker, E. C. Boerma, M. A. Kuiper et al., HDL-cholesterol level and cortisol response to synacthen in critically ill patients, Intensive Care Medicine, vol.27, issue.12, pp.2199-2203, 2003.
DOI : 10.1007/BF03350334

J. Chien, J. Jerng, C. Yu, and P. Yang, Low serum level of high-density lipoprotein cholesterol is a poor prognostic factor for severe sepsis*, Critical Care Medicine, vol.33, issue.8, pp.1688-1693, 2005.
DOI : 10.1097/01.CCM.0000171183.79525.6B

S. Weitzman and S. Berger, Clinical Trial Design in Studies of Corticosteroids for Bacterial Infections, Annals of Internal Medicine, vol.81, issue.1, pp.36-42, 1974.
DOI : 10.7326/0003-4819-81-1-36

W. Schumer, Steroids in the Treatment of Clinical Septic Shock, Annals of Surgery, vol.184, issue.3, pp.333-341, 1976.
DOI : 10.1097/00000658-197609000-00011

R. Lefering and E. A. Neugebauer, Steroid controversy in sepsis and septic shock, Critical Care Medicine, vol.23, issue.7, pp.1294-1303, 1995.
DOI : 10.1097/00003246-199507000-00021

C. L. Sprung, Hydrocortisone Therapy for Patients with Septic Shock, New England Journal of Medicine, vol.358, issue.2, pp.111-124, 2008.
DOI : 10.1056/NEJMoa071366

R. P. Dellinger, Surviving Sepsis Campaign, Critical Care Medicine, vol.41, issue.2, pp.296-327, 2008.
DOI : 10.1097/CCM.0b013e31827e83af

URL : http://pdfs.journals.lww.com/ccmjournal/2013/02000/Surviving_Sepsis_Campaign___International.24.pdf?token=method|ExpireAbsolute;source|Journals;ttl|1504299991983;payload|mY8D3u1TCCsNvP5E421JYK6N6XICDamxByyYpaNzk7FKjTaa1Yz22MivkHZqjGP4kdS2v0J76WGAnHACH69s21Csk0OpQi3YbjEMdSoz2UhVybFqQxA7lKwSUlA502zQZr96TQRwhVlocEp/sJ586aVbcBFlltKNKo+tbuMfL73hiPqJliudqs17cHeLcLbV/CqjlP3IO0jGHlHQtJWcICDdAyGJMnpi6RlbEJaRheGeh5z5uvqz3FLHgPKVXJzd9ia1/MJJUFmWp1b9urv13HGxLCxqSptnCmTonIrDcMk=;hash|q4R7gkVEeDtraL4DxyGu6w==

D. Rittirsch, M. S. Huber-lang, M. A. Flierl, and P. A. Ward, Immunodesign of experimental sepsis by cecal ligation and puncture, Nature Protocols, vol.22, issue.1, pp.31-36, 2009.
DOI : 10.1164/ajrccm.164.5.2010073

K. A. Wichterman, A. E. Baue, and I. H. Chaudry, Sepsis and septic shock???A review of laboratory models and a proposal, Journal of Surgical Research, vol.29, issue.2, pp.189-201, 1980.
DOI : 10.1016/0022-4804(80)90037-2

E. A. Deitch, ANIMAL MODELS OF SEPSIS AND SHOCK, Shock, vol.9, issue.1, pp.1-11, 1998.
DOI : 10.1097/00024382-199801000-00001

A. Mansart, P. E. Bollaert, C. Seguin, B. Levy, D. Longrois et al., Hemodynamic Effects of Early Versus Late Glucocorticosteroid Administration in Experimental Septic Shock, Shock, vol.19, issue.1, pp.38-44, 2003.
DOI : 10.1097/00024382-200301000-00008

N. Sennoun, Activated protein C improves lipopolysaccharide-induced cardiovascular dysfunction by decreasing tissular inflammation and oxidative stress, Critical Care Medicine, vol.37, issue.1, pp.246-255, 2009.

C. T. Esmon, The endothelial protein C receptor, Current Opinion in Hematology, vol.13, issue.5, pp.382-385, 2006.
DOI : 10.1097/01.moh.0000239712.93662.35

P. Bilbault, Influence of drotrecogin alpha (activated) infusion on the variation of Bax/Bcl-2 and Bax/Bcl-xl ratios in circulating mononuclear cells: A cohort study in septic shock patients, Critical Care Medicine, vol.35, issue.1, pp.69-75, 2007.
DOI : 10.1097/01.CCM.0000251133.26979.F4

M. R. Looney and M. A. , Matthay, « Bench-to-bedside review: the role of activated protein C in maintaining endothelial tight junction function and its relationship to organ injury, Critical Care, vol.10, issue.6, p.239, 2006.
DOI : 10.1186/cc5099

V. Regnault and B. Levy, « Recombinant activated protein C in sepsis: endothelium protection or endothelium therapy?, Critical Care, vol.11, issue.1, p.103, 2007.
DOI : 10.1186/cc5135

R. Favory, S. Lancel, X. Maréchal, S. Tissier, and E. R. Neviere, Cardiovascular protective role for activated protein C during endotoxemia in rats, Intensive Care Medicine, vol.95, issue.6, pp.899-905, 2006.
DOI : 10.1056/NEJM198908033210503

H. Isobe, Activated Protein C Prevents Endotoxin-Induced Hypotension in Rats by Inhibiting Excessive Production of Nitric Oxide, Circulation, vol.104, issue.10, pp.1171-1175, 2001.
DOI : 10.1161/hc3501.093799

X. Monnet, B. Lamia, N. Anguel, C. Richard, G. Bonmarchand et al., Rapid and beneficial hemodynamic effects of activated protein C in septic shock patients, Intensive Care Medicine, vol.33, issue.11, pp.1573-1576, 2005.
DOI : 10.1007/s00134-003-1731-1

A. Hafezi-moghadam, Acute cardiovascular protective effects of corticosteroids are mediated by non-transcriptional activation of endothelial nitric oxide synthase, Nature Medicine, vol.101, issue.5, pp.473-479, 2002.
DOI : 10.1161/01.CIR.101.9.1019

H. Prigent and V. Maxime, Annane, « Science review: mechanisms of impaired adrenal function in sepsis and molecular actions of glucocorticoids, Critical Care, vol.8, issue.4, pp.243-252, 2004.
DOI : 10.1186/cc2878

A. Mansart, P. Bollaert, B. Levy, M. Nicolas, and J. Mallié, Comparative effects of dexamethasone and l-canavanine in experimental septic shock, European Journal of Pharmacology, vol.475, issue.1-3, pp.1-3, 2003.
DOI : 10.1016/S0014-2999(03)02035-1

M. O. Maybauer, Recombinant human activated protein C attenuates cardiovascular and microcirculatory dysfunction in acute lung injury and septic shock, Critical Care, vol.14, issue.6, p.217, 2010.
DOI : 10.1186/cc9342

N. Matsuda, Y. Hayashi, Y. Takahashi, and E. Y. Hattori, Phosphorylation of Endothelial Nitric-Oxide Synthase Is Diminished in Mesenteric Arteries from Septic Rabbits Depending on the Altered Phosphatidylinositol 3-Kinase/Akt Pathway: Reversal Effect of Fluvastatin Therapy, Journal of Pharmacology and Experimental Therapeutics, vol.319, issue.3, pp.1348-1354, 2006.
DOI : 10.1124/jpet.106.109785

A. Cauwels, Nitric oxide in shock, Nitric oxide in shock, pp.557-565, 2007.
DOI : 10.1038/sj.ki.5002340

P. Pacher, J. S. Beckman, and E. L. Liaudet, Nitric Oxide and Peroxynitrite in Health and Disease, Physiological Reviews, vol.87, issue.1, pp.315-424, 2007.
DOI : 10.1089/152308601750100443

L. Connelly, M. Madhani, and A. J. Hobbs, Resistance to Endotoxic Shock in Endothelial Nitric-oxide Synthase (eNOS) Knock-out Mice, Journal of Biological Chemistry, vol.34, issue.11, pp.10040-10046, 2005.
DOI : 10.1097/00004872-200311000-00020

A. Vieillard-baron, S. Prin, K. Chergui, and O. Dubourg, Hemodynamic Instability in Sepsis, American Journal of Respiratory and Critical Care Medicine, vol.168, issue.11, pp.1270-1276, 2003.
DOI : 10.1164/rccm.200202-146CC

M. Levi, T. Van-der-poll, R. Dellinger, M. Levy, J. Carlet et al., Inflammation and coagulation Surviving Sepsis Campaign: international guidelines for management of severe sepsis and septic shock, Crit Care Med Intensive Care Med, vol.38, issue.34, pp.26-34, 2008.

G. Bernard, J. Vincent, P. Laterre, S. Larosa, J. Dhainaut et al., Efficacy and Safety of Recombinant Human Activated Protein C for Severe Sepsis, New England Journal of Medicine, vol.344, issue.10, pp.699-709, 2001.
DOI : 10.1056/NEJM200103083441001

L. Mosnier, B. Zlokovic, and J. Griffin, The cytoprotective protein C pathway, Blood, vol.109, issue.8, pp.3161-3172, 2007.
DOI : 10.1182/blood-2006-09-003004

D. Joyce, L. Gelbert, A. Ciaccia, B. Dehoff, and B. Grinnell, Gene Expression Profile of Antithrombotic Protein C Defines New Mechanisms Modulating Inflammation and Apoptosis, Journal of Biological Chemistry, vol.95, issue.14, 2001.
DOI : 10.1093/glycob/4.2.221

N. Day, N. Phu, N. Mai, D. Bethell, T. Chau et al., Effects of dopamine and epinephrine infusions on renal hemodynamics in severe malaria and severe sepsis, Critical Care Medicine, vol.28, issue.5, pp.1353-1362, 2000.
DOI : 10.1097/00003246-200005000-00016

P. Bilbault, T. Lavaux, A. Launoy, M. Gaub, N. Meyer et al., Influence of drotrecogin alpha (activated) infusion on the variation of Bax/Bcl-2 and Bax/Bcl-xl ratios in circulating mononuclear cells: A cohort study in septic shock patients, Critical Care Medicine, vol.35, issue.1, pp.69-75, 2007.
DOI : 10.1097/01.CCM.0000251133.26979.F4

M. Looney and M. Matthay, Bench-to-bedside review: the role of activated protein C in maintaining endothelial tight junction function and its relationship to organ injury, Critical Care, vol.10, issue.6, p.239, 2006.
DOI : 10.1186/cc5099

V. Regnault and B. Levy, Recombinant activated protein C in sepsis: endothelium protection or endothelium therapy?, Crit Care, vol.11, issue.103, 2007.

R. Favory, S. Lancel, X. Marechal, S. Tissier, and R. Neviere, Cardiovascular protective role for activated protein C during endotoxemia in rats, Intensive Care Medicine, vol.95, issue.6, pp.899-905, 2006.
DOI : 10.1056/NEJM198908033210503

H. Isobe, K. Okajima, M. Uchiba, A. Mizutani, N. Harada et al., Activated Protein C Prevents Endotoxin-Induced Hypotension in Rats by Inhibiting Excessive Production of Nitric Oxide, Circulation, vol.104, issue.10, pp.1171-1175, 2001.
DOI : 10.1161/hc3501.093799

A. Gupta, G. Rhodes, D. Berg, B. Gerlitz, B. Molitoris et al., Activated protein C ameliorates LPS-induced acute kidney injury and downregulates renal INOS and angiotensin 2, American Journal of Physiology-Renal Physiology, vol.293, issue.1, pp.245-254, 2007.
DOI : 10.1016/j.amjmed.2005.01.069

X. Monnet, B. Lamia, N. Anguel, C. Richard, G. Bonmarchand et al., Rapid and beneficial hemodynamic effects of activated protein C in septic shock patients, Intensive Care Medicine, vol.33, issue.11, pp.1573-1576, 2005.
DOI : 10.1007/s00134-003-1731-1

N. Sennoun, F. Meziani, O. Dessebe, V. Cattan, C. S. Montemont et al., Activated protein C improves lipopolysaccharideinduced cardiovascular dysfunction by decreasing tissular inflammation and oxidative stress, Crit Care Med, vol.37, pp.246-255, 2009.

D. Annane, E. Bellissant, P. Bollaert, J. Briegel, M. Confalonieri et al., Corticosteroids in the Treatment of Severe Sepsis and Septic Shock in Adults, JAMA, vol.301, issue.22, pp.2362-2375, 2009.
DOI : 10.1001/jama.2009.815

P. Marik, S. Pastores, D. Annane, G. Meduri, C. Sprung et al., Recommendations for the diagnosis and management of corticosteroid insufficiency in critically ill adult patients: Consensus statements from an international task force by the American College of Critical Care Medicine, Critical Care Medicine, vol.36, issue.6, pp.1937-1949, 2008.
DOI : 10.1097/CCM.0b013e31817603ba

A. Hafezi-moghadam, T. Simoncini, Z. Yang, F. Limbourg, J. Plumier et al., Acute cardiovascular protective effects of corticosteroids are mediated by non-transcriptional activation of endothelial nitric oxide synthase, Nature Medicine, vol.101, issue.5, pp.473-479, 2002.
DOI : 10.1161/01.CIR.101.9.1019

H. Prigent, V. Maxime, and D. Annane, Science review: mechanisms of impaired adrenal function in sepsis and molecular actions of glucocorticoids, Critical Care, vol.8, issue.4, pp.243-252, 2004.
DOI : 10.1186/cc2878

A. Mansart, P. Bollaert, B. Levy, M. Nicolas, and J. Mallie, Comparative effects of dexamethasone and l-canavanine in experimental septic shock, European Journal of Pharmacology, vol.475, issue.1-3, pp.61-67, 2003.
DOI : 10.1016/S0014-2999(03)02035-1

N. Sennoun, C. Baron-menguy, M. Burban, T. Lecompte, R. Andriantsitohaina et al., Recombinant human activated protein C improves endotoxemia-induced endothelial dysfunction: a blood-free model in isolated mouse arteries, American Journal of Physiology-Heart and Circulatory Physiology, vol.297, issue.1, pp.277-282, 2009.
DOI : 10.1016/j.thromres.2004.09.011

A. Mansart, P. Bollaert, C. Seguin, B. Levy, D. Longrois et al., Hemodynamic Effects of Early Versus Late Glucocorticosteroid Administration in Experimental Septic Shock, Shock, vol.19, issue.1, pp.38-44, 2003.
DOI : 10.1097/00024382-200301000-00008

M. Maybauer, D. Maybauer, J. Fraser, C. Szabo, M. Westphal et al., Recombinant human activated protein C attenuates cardiovascular and microcirculatory dysfunction in acute lung injury and septic shock, Critical Care, vol.14, issue.6, p.217, 2010.
DOI : 10.1186/cc9342

N. Matsuda, Y. Hayashi, Y. Takahashi, and Y. Hattori, Phosphorylation of Endothelial Nitric-Oxide Synthase Is Diminished in Mesenteric Arteries from Septic Rabbits Depending on the Altered Phosphatidylinositol 3-Kinase/Akt Pathway: Reversal Effect of Fluvastatin Therapy, Journal of Pharmacology and Experimental Therapeutics, vol.319, issue.3, pp.1348-1354, 2006.
DOI : 10.1124/jpet.106.109785

A. Cauwels, Nitric oxide in shock, Kidney International, vol.72, issue.5, pp.557-565, 2007.
DOI : 10.1038/sj.ki.5002340

P. Pacher, J. Beckman, and L. Liaudet, Nitric Oxide and Peroxynitrite in Health and Disease, Physiological Reviews, vol.87, issue.1, pp.315-424, 2007.
DOI : 10.1089/152308601750100443

D. Dudzinski and T. Michel, Life history of eNOS: Partners and pathways, Cardiovascular Research, vol.75, issue.2, pp.247-260, 2007.
DOI : 10.1016/j.cardiores.2007.03.023

B. Levy, C. S. Sennoun, N. Ducrocq, N. Kimmoun, A. Asfar et al., Vascular hyporesponsiveness to vasopressors in septic shock: from bench to bedside, Intensive Care Medicine, vol.345, issue.12, pp.2019-2029, 2010.
DOI : 10.1161/01.CIR.95.5.1122

URL : https://hal.archives-ouvertes.fr/hal-00666836

L. Connelly, M. Madhani, and A. Hobbs, Resistance to Endotoxic Shock in Endothelial Nitric-oxide Synthase (eNOS) Knock-out Mice, Journal of Biological Chemistry, vol.34, issue.11, pp.10040-10046, 2005.
DOI : 10.1097/00004872-200311000-00020

V. Strunk, K. Hahnenkamp, M. Schneuing, L. Fischer, and G. Rich, Selective iNOS Inhibition Prevents Hypotension in Septic Rats While Preserving Endothelium-Dependent Vasodilation, Anesthesia and Analgesia, vol.92, issue.3, pp.681-687, 2001.
DOI : 10.1213/00000539-200103000-00025

J. Buckley, M. Singer, and L. Clapp, Role of KATP channels in sepsis, Cardiovascular Research, vol.72, issue.2, pp.220-230, 2006.
DOI : 10.1016/j.cardiores.2006.07.011

A. Vieillard-baron, S. Prin, K. Chergui, O. Dubourg, and F. Jardin, Hemodynamic Instability in Sepsis, American Journal of Respiratory and Critical Care Medicine, vol.168, issue.11, pp.1270-1276, 2003.
DOI : 10.1164/rccm.200202-146CC

E. Rivers, B. Nguyen, S. Havstad, J. Ressler, A. Muzzin et al., Early Goal-Directed Therapy in the Treatment of Severe Sepsis and Septic Shock, New England Journal of Medicine, vol.345, issue.19, pp.1368-1377, 2001.
DOI : 10.1056/NEJMoa010307