]. Sakata, Y. Hoit, B. Liggett, S. Walsh, R. Dorn et al., Decompensation of Pressure-Overload Hypertrophy in G??q-Overexpressing Mice, Circulation, vol.97, issue.15, pp.1488-95, 1998.
DOI : 10.1161/01.CIR.97.15.1488

N. Frey and E. Olson, Cardiac Hypertrophy: The Good, the Bad, and the Ugly, Annual Review of Physiology, vol.65, issue.1, pp.45-79, 2003.
DOI : 10.1146/annurev.physiol.65.092101.142243

G. Dorn and T. Force, Protein kinase cascades in the regulation of cardiac hypertrophy, Journal of Clinical Investigation, vol.115, issue.3, pp.527-564, 2005.
DOI : 10.1172/JCI24178

B. Wilkins, Y. Dai, O. Bueno, S. Parsons, and J. Xu, Calcineurin/NFAT Coupling Participates in Pathological, but not Physiological, Cardiac Hypertrophy, Circulation Research, vol.94, issue.1, pp.110-118, 2004.
DOI : 10.1161/01.RES.0000109415.17511.18

]. Mudd, S. Finkelstein, J. Irrevere, F. Laster, and L. , Homocystinuria: An Enzymatic Defect, Science, vol.143, issue.3613, pp.1443-1448, 1964.
DOI : 10.1126/science.143.3613.1443

K. Mccully, Vascular pathology of homocysteinemia: implications for the pathogenesis of arteriosclerosis

]. Boushey, C. Beresford, S. Omenn, G. Motulsky, and A. , A quantitative assessment of plasma homocysteine as a risk factor for vascular disease. Probable benefits of increasing folic acid intakes, JAMA: The Journal of the American Medical Association, vol.274, issue.13, pp.1049-57, 1995.
DOI : 10.1001/jama.274.13.1049

L. Brattström, B. Israelsson, B. Norrving, D. Bergqvist, and J. Thome, Impaired homocysteine metabolism in early-onset cerebral and peripheral occlusive arterial disease Effects of pyridoxine and folic acid treatment, Atherosclerosis, vol.81, issue.1, pp.51-60, 1990.
DOI : 10.1016/0021-9150(90)90058-Q

S. Mudd and J. Poole, Labile methyl balances for normal humans on various dietary regimens, Metabolism, vol.24, issue.6, pp.721-755, 1975.
DOI : 10.1016/0026-0495(75)90040-2

]. Mudd, S. Ebert, M. Scriver, and C. , Labile methyl group balances in the human: The role of sarcosine, Metabolism, vol.29, issue.8, pp.707-727, 1980.
DOI : 10.1016/0026-0495(80)90192-4

R. Cook and C. Wagner, Glycine N-methyltransferase is a folate binding protein of rat liver cytosol., Proceedings of the National Academy of Sciences, vol.81, issue.12, pp.3631-3635, 1984.
DOI : 10.1073/pnas.81.12.3631

S. Miner, J. Evroski, and C. Dec, Clinical chemistry and molecular biology of homocysteine metabolism: An update, Clinical Biochemistry, vol.30, issue.3, pp.189-201, 1997.
DOI : 10.1016/S0009-9120(96)00172-5

J. Miller, M. Nadeau, J. Smith, D. Smith, and J. Selhub, -adenosylmethionine's co-ordinate regulation of homocysteine metabolism, Biochemical Journal, vol.298, issue.2, pp.415-424, 1994.
DOI : 10.1042/bj2980415

J. Miller, J. Ribaya-mercado, R. Russell, D. Shephard, and F. Morrow, Effect of vitamin B-6 deficiency on fasting plasma homocysteine concentrations, The American Journal of Clinical Nutrition, vol.55, issue.6, pp.1154-60, 1992.
DOI : 10.1093/ajcn/55.6.1154

J. Miller, M. Nadeau, D. Smith, and J. Selhub, Vitamin B???6 deficiency vs folate deficiency: comparison of responses to methionine loading in rats, The American Journal of Clinical Nutrition, vol.59, issue.5, pp.1033-1042, 1994.
DOI : 10.1093/ajcn/59.5.1033

S. Kang, J. Zhou, P. Wong, J. Kowalisyn, and G. Strokosch, Intermediate homocysteinemia: a thermolabile variant of methylenetetrahydrofolatereductase, Am J Hum Genet, vol.43, pp.414-435, 1988.

K. Dalery, S. Lusier-cacan, J. Selhub, J. Davignon, and . Latoury, Homocysteine and coronary artery disease in French Canadian subjects: Relation with vitamins B12, B6, pyridoxal phosphate, and folate, The American Journal of Cardiology, vol.75, issue.16, pp.1107-1118, 1995.
DOI : 10.1016/S0002-9149(99)80739-5

]. Franken, D. Boers, G. Blom, H. Trijbels, and J. , Effect of various regimens of vitamin B6 and folic acid on mild hyperhomocysteinaemia in vascular patients, Journal of Inherited Metabolic Disease, vol.309, issue.1, pp.159-62, 1994.
DOI : 10.1007/BF00735426

S. Kang, P. Wong, and M. Norusis, Homocysteinemia due to folate deficiency, Metabolism, vol.36, issue.5, pp.458-62, 1987.
DOI : 10.1016/0026-0495(87)90043-6

H. Naurath, E. Joosten, R. Riezler, S. Stabler, and R. Allen, Effects of vitamin B12, folate, and vitamin B6 supplements in elderly people with normal serum vitamin concentrations, The Lancet, vol.346, issue.8967, pp.858-867, 1995.
DOI : 10.1016/S0140-6736(95)92113-3

J. Ubbink, W. Vermaak, A. Van-der-merwe, P. Becker, and R. Delport, Vitamin Requirements for the Treatment of Hyperhomocysteinemia in Humans, The Journal of Nutrition, vol.124, issue.10, pp.1927-1960, 1994.
DOI : 10.1093/jn/124.10.1927

O. Stanger, W. Herrmann, K. Pietrzik, B. Fowler, and J. Geisel, ???ber den rationellen klinischen Umgang mit Homocystein, Fols???ure und B-Vitaminen bei kardiovaskul???ren und thrombotischen Erkrankungen, Zeitschrift f???r Kardiologie, vol.93, issue.6, pp.439-53, 2004.
DOI : 10.1007/s00392-004-0075-3

J. Selhub, P. Jacques, P. Wilson, D. Rush, and I. Rosenberg, Vitamin Status and Intake as Primary Determinants of Homocysteinemia in an Elderly Population, JAMA: The Journal of the American Medical Association, vol.270, issue.22, pp.2693-2701, 1993.
DOI : 10.1001/jama.1993.03510220049033

A. Chango, G. Potier-de-courcy, F. Boisson, J. Guilland, and F. Barbé, 10-methylenetetrahydrofolate reductas (MHTFR) Commonmutations, folate status and plasma homocysteine in healthy French adults of the SU.VI.MAX cohort, Br J Nutr, vol.5, issue.84, pp.891-894, 2000.

D. Harmon, D. Shields, J. Woodside, D. Mcmaster, and J. Yarnell, Methionine synthase D919G polymorphism is a significant but modest determinant of circulating homocysteine concentrations, Genetic Epidemiology, vol.90, issue.4, pp.298-309, 1999.
DOI : 10.1093/qjmed/90.8.551

D. Gaughan, L. Kluijtmans, S. Barbaux, D. Mcmaster, and I. Young, The methionine synthase reductase (MTRR) A66 polymorphism is a novel genetic determinant of plasma homocystéine concentrations

P. Jacques, . Bostomag, R. Williams, R. Ellison, and J. Eckfeldt, Relation Between Folate Status, a Common Mutation in Methylenetetrahydrofolate Reductase, and Plasma Homocysteine Concentrations, Circulation, vol.93, issue.1, pp.7-9, 1996.
DOI : 10.1161/01.CIR.93.1.7

U. Refsum, P. Ueland, O. Nygard, and S. Vollset, HOMOCYSTEINE AND CARDIOVASCULAR DISEASE, Annual Review of Medicine, vol.49, issue.1, pp.31-63, 1998.
DOI : 10.1146/annurev.med.49.1.31

K. Mccully, Growth disorders and homocysteine metabolism, Ann Clin Lab Sci, vol.5, pp.147-52, 1975.

B. Nedrebo, U. Ericsson, O. Nygard, H. Refsum, and P. Ueland, Plasma total homocysteine levels in hyperthyroid and hypothyroid patients, Metabolism, vol.47, issue.1, pp.89-93, 1998.
DOI : 10.1016/S0026-0495(98)90198-6

G. Upchurch, G. Welch, A. Fabiani, J. Freedman, and J. Johnson, Homocysteine decreases bioavailable nitric oxide by a mechanism involving glutathione peroxidase, J Biol Chem, vol.27, pp.17012-17019, 1997.

P. Ho, S. Collins, S. Dhitavat, D. Ortiz, and D. Ashline, Homocysteine potentiates ??-amyloid neurotoxicity: role of oxidative stress, Journal of Neurochemistry, vol.67, issue.2, pp.249-53, 2001.
DOI : 10.1046/j.1471-4159.1996.67041419.x

J. Stamler, J. Osborne, O. Jaraki, L. Rabbani, and M. Mullins, Adverse vascular effects of homocysteine are modulated by endothelium-derived relaxing factor and related oxides of nitrogen., Journal of Clinical Investigation, vol.91, issue.1, pp.308-326, 1993.
DOI : 10.1172/JCI116187

G. Welch, G. Upchurch, R. Farivar, A. Pigazz, and K. Vu, Homocysteine-induced nitric oxide production in vascular smooth-muscle cells by NF-kappa B-dependent transcriptional activation of Nos2, Proc Assoc Am Physicians, vol.110, pp.22-31, 1998.

G. Upchurch, G. Welch, and L. J. Homocysteine, EDRF, and endothelial function, [39]. Jacobsen DW. Homocysteine and vitamins in cardiovascular disease, pp.12901833-12901876, 1996.

X. Zhang, H. Li, H. Jin, Z. Ebin, and S. Brodsky, Effects of homocysteine on endothelial nitric oxide production, American Journal of Physiology-Renal Physiology, vol.277, issue.4, pp.671-679, 2000.
DOI : 10.1161/01.CIR.83.2.391

A. Tawakol, T. Omland, G. M. Wu, J. Creager, and M. , Hyperhomocyst(e)inemia Is Associated With Impaired Endothelium-Dependent Vasodilation in Humans, Circulation, vol.95, issue.5, pp.1119-1140, 1997.
DOI : 10.1161/01.CIR.95.5.1119

M. Dalton, P. Gadson, R. Wrenn, and T. Rosenquist, Homocysteine signal cascade: production of phospholipids, activation of protein kinase C, and the induction of c-fos and c-myb in smooth muscle cells., The FASEB Journal, vol.11, issue.8, pp.703-714, 1997.
DOI : 10.1096/fasebj.11.8.9240971

. Suzukiy, M. Lorenzi, S. Shi, R. Day, and J. Blumberg, Homocysteine exerts cell type-specific inhibition of AP-1 transcription factor, Free Radical Biology and Medicine, vol.28, issue.1, pp.39-45, 2000.
DOI : 10.1016/S0891-5849(99)00200-2

M. Hofman, E. Lalla, Y. Lu, M. Gleason, and B. Wolf, Hyperhomocysteinemia enhances vascular inflammation and accelerates atherosclerosis in a murine model, Journal of Clinical Investigation, vol.107, issue.6, pp.675-83, 2001.
DOI : 10.1172/JCI10588

K. Mccully, A. Olszewski, and M. Vezeridis, Homocysteine and lipid metabolism in atherogenesis: effect of the homocysteine thiolactonyl derivatives, thioretinaco and thioretinamide, Atherosclerosis, vol.83, issue.2-3, pp.197-206, 1990.
DOI : 10.1016/0021-9150(90)90165-F

P. Harpel, X. Zhang, and W. Borth, Homocysteine and Hemostasis: Pathogenetic Mechanisms Predisposing to Thrombosis, The Journal of Nutrition, vol.126, issue.suppl_4, pp.1285-1294, 1996.
DOI : 10.1093/jn/126.suppl_4.1285S

A. Emsley, J. J. Gomes, G. Angelini, G. Planer, and F. , Investigation of the inhibitory effects of homocysteine and copper on nitric oxide-mediated relaxation of rat isolated aorta, British Journal of Pharmacology, vol.96, issue.4, pp.1034-1074, 1999.
DOI : 10.1161/01.CIR.96.8.2542

J. Chambers, A. Mcgregor, J. Jean-marie, O. Obeid, and J. Kooner, Demonstration of Rapid Onset Vascular Endothelial Dysfunction After Hyperhomocysteinemia : An Effect Reversible With Vitamin C Therapy, Circulation, vol.99, issue.9, pp.1156-60, 1999.
DOI : 10.1161/01.CIR.99.9.1156

P. Mannisto, I. Ulmanen, K. Lundstrom, J. Taskinen, and J. Tenhunen, Characteristics of catechol O-methyltransferase (COMT) and properties of selective COMT inhibitors, Prog Drug Res, vol.39, pp.291-350, 1992.

J. Tsai, M. Perella, M. Yoshizumi, C. Hsier, and E. Haber, Promotion of vascular smooth muscle cell growth by homocysteine: a link to atherosclerosis., Proceedings of the National Academy of Sciences, vol.91, issue.14, pp.6369-73, 1994.
DOI : 10.1073/pnas.91.14.6369

M. Khan, M. Yamauchi, S. Srisawasdi, D. Stiner, and S. Doty, Homocysteine decreases chondrocyte-mediated matrix mineralization in differentiating chick limb-bud mesenchymal cell micro-mass cultures, Bone, vol.28, issue.4, pp.387-98, 2001.
DOI : 10.1016/S8756-3282(01)00409-4

R. Guéant-rodriguez, Y. Juillière, M. Nippert, I. Abdelmouttaleb, and B. Herbeth, Left ventricular systolic dysfunction is an independent predictor of homocysteine in angiographically documented patients with or without coronary artery lesions, Journal of Thrombosis and Haemostasis, vol.5, issue.suppl 4, pp.1209-1225, 2007.
DOI : 10.1152/ajpheart.00475.2002

J. Sundström, R. Vasan, T. James, J. Narula, N. Haider et al., Homocysteine and heart failure: a review of investigations from the Framingham Heart Study Reed JC. Mechanisms of apoptosis Apoptosis in cardiac disease, Apoptosis in myocytes in end-stage heart failure, pp.987-992, 1996.

G. Olivetti, R. Abbi, F. Quaini, J. Kajstura, and W. Cheng, Apoptosis in the Failing Human Heart, New England Journal of Medicine, vol.336, issue.16, pp.1131-1172, 1997.
DOI : 10.1056/NEJM199704173361603

P. Kang and S. Izumo, Apoptosis and Heart Failure : A Critical Review of the Literature, Circulation Research, vol.86, issue.11, pp.1107-1120, 2000.
DOI : 10.1161/01.RES.86.11.1107

Z. Li, O. Bing, X. Long, K. Robinson, and E. Lakatta, Increased cardiomyocyte apoptosis during the transition to heart failure in the spontaneously hypertensive rat, American Journal of Physiology-Heart and Circulatory Physiology, vol.272, issue.5, pp.2313-2322, 1997.
DOI : 10.1152/ajpheart.1997.272.5.H2313

H. Hirota, J. Chen, U. Betz, K. Rajewsky, and Y. Gu, Loss of a gp130 Cardiac Muscle Cell Survival Pathway Is a Critical Event in the Onset of Heart Failure during Biomechanical Stress, Cell, vol.97, issue.2, pp.189-98, 1999.
DOI : 10.1016/S0092-8674(00)80729-1

D. Wencker, M. Chandra, K. Nguyen, W. Miao, and S. Garantziotis, A mechanistic role for cardiac myocyte apoptosis in heart failure, Journal of Clinical Investigation, vol.111, issue.10, pp.1497-504, 2003.
DOI : 10.1172/JCI17664

I. Dubus, J. Samuel, F. Marotte, C. Delcayre, and L. Rappaport, Beta-adrenergic agonists stimulate the synthesis of noncontractile but not contractile proteins in cultured myocytes isolated from adult rat heart, Circulation Research, vol.66, issue.3, pp.867-74, 1990.
DOI : 10.1161/01.RES.66.3.867

C. Singh, K. Pimentel, D. Colucci, W. Bisognano, J. Weinberger et al., Norepinephrine stimulates apoptosis in adult rat ventricular myocytes by activation of the beta-adrenergic pathway Myocardial-directed overexpression of the human beta(1)-adrenergic receptor in transgenic mice, Circulation J Mol Cell Cardiol, vol.9867, issue.32, pp.1329-1363, 1998.

R. Xiao, P. Avdonin, Y. Zhou, H. Cheng, and S. Akhter, Coupling of ??2-Adrenoceptor to Gi Proteins and Its Physiological Relevance in Murine Cardiac Myocytes, Circulation Research, vol.84, issue.1, pp.43-52, 1999.
DOI : 10.1161/01.RES.84.1.43

Y. Wang, S. Huang, V. Sah, R. J. Brown, and J. , Cardiac Muscle Cell Hypertrophy and Apoptosis Induced by Distinct Members of the p38 Mitogen-activated Protein Kinase Family, Journal of Biological Chemistry, vol.272, issue.4, pp.2161-2169, 1998.
DOI : 10.1016/S0092-8674(00)81878-4

W. Colucci and K. Singh, p38 mitogen-activated protein kinase pathway protects adult rat ventricular myocytes against beta-adrenergic receptor-stimulated apoptosis. Evidence for Gi-dependent activation

A. Remondino, S. Kwon, C. Communal, D. Pimentel, and D. Sawyer, beta-Adrenergic Receptor-Stimulated Apoptosis in Cardiac Myocytes Is Mediated by Reactive Oxygen Species/c-Jun NH2-Terminal Kinase-Dependent Activation of the Mitochondrial Pathway, Circulation Research, vol.92, issue.2, pp.136-144, 2003.
DOI : 10.1161/01.RES.0000054624.03539.B4

M. Singh, B. Menon, Z. Xie, and W. Colucci, beta1 integrins expression in adult rat ventricular myocytes and its role in the regulation of beta-adrenergic receptor-stimulated apoptosis Neurohormonal regulation of myocardial cell apoptosis during the development of heart failure, J Cell Biochem J Cell Physiol, vol.8973, issue.186, pp.381-389, 2001.

Z. Mallat, A. Tedgui, F. Fontaliran, R. Frank, and M. Durigon, Evidence of Apoptosis in Arrhythmogenic Right Ventricular Dysplasia, New England Journal of Medicine, vol.335, issue.16, pp.1190-1196, 1996.
DOI : 10.1056/NEJM199610173351604

J. Narula, P. Pandey, E. Arbustini, N. Haider, and N. Narula, Apoptosis in heart failure: Release of cytochrome c from mitochondria and activation of caspase-3 in human cardiomyopathy, Proceedings of the National Academy of Sciences, vol.57, issue.12, pp.8144-8153, 1999.
DOI : 10.1253/jcj.57.1150

D. Cesselli, I. Jakoniuk, L. Barlucchi, A. Beltrami, and T. Hintze, Oxidative Stress-Mediated Cardiac Cell Death Is a Major Determinant of Ventricular Dysfunction and Failure in Dog Dilated Cardiomyopathy, Circulation Research, vol.89, issue.3, pp.279-86, 2001.
DOI : 10.1161/hh1501.094115

G. Condorelli, R. Roncarati, R. J. Pisani, A. Stassi, and G. , Heart-targeted overexpression of caspase3 in mice increases infarct size and depresses cardiac function, Proceedings of the National Academy of Sciences, vol.6, issue.11, pp.9977-82, 2001.
DOI : 10.1038/sj.cdd.4400596

P. Kang, A. Haunstetter, H. Aoki, A. Usheva, and S. Izumo, Morphological and Molecular Characterization of Adult Cardiomyocyte Apoptosis During Hypoxia and Reoxygenation, Circulation Research, vol.87, issue.2, pp.118-143, 2000.
DOI : 10.1161/01.RES.87.2.118

K. Suzuki, S. Kostin, V. Person, A. Elsasser, and J. Schaper, Time Course of the Apoptotic Cascade and Effects of Caspase Inhibitors in Adult Rat Ventricular Cardiomyocytes, Journal of Molecular and Cellular Cardiology, vol.33, issue.5, pp.983-94, 2001.
DOI : 10.1006/jmcc.2001.1364

T. Ide, H. Tsutsui, S. Kinugawa, N. Suematsu, and S. Hayashidani, Direct Evidence for Increased Hydroxyl Radicals Originating From Superoxide in the Failing Myocardium, Circulation Research, vol.86, issue.2, pp.152-159, 2000.
DOI : 10.1161/01.RES.86.2.152

P. Ferdinandy and R. Schulz, Peroxynitrite: Toxic or Protective in the Heart?, Circulation Research, vol.88, issue.2, pp.12-15, 2001.
DOI : 10.1161/01.RES.88.2.e12

T. Ide, H. Tsutsui, S. Kinugawa, H. Utsumi, and D. Kang, Mitochondrial Electron Transport Complex I Is a Potential Source of Oxygen Free Radicals in the Failing Myocardium, Circulation Research, vol.85, issue.4, pp.357-63, 1999.
DOI : 10.1161/01.RES.85.4.357

M. Benderdour, G. Charron, D. Deblois, B. Comte, D. Rosiers et al., -isocitrate Dehydrogenase Is Inactivated through 4-Hydroxynonenal Adduct Formation, Journal of Biological Chemistry, vol.30, issue.182, pp.45154-45163, 2003.
DOI : 10.1016/S0022-2828(03)00145-7

T. Ide, H. Tsutsui, S. Hayashidani, D. Kang, and N. Suematsu, Mitochondrial DNA Damage and Dysfunction Associated With Oxidative Stress in Failing Hearts After Myocardial Infarction, Circulation Research, vol.88, issue.5, pp.529-564, 2001.
DOI : 10.1161/01.RES.88.5.529

H. Friedl, G. Till, U. Ryan, and P. Ward, Mediator-induced activation of xanthine oxidase in endothelial cells., The FASEB Journal, vol.3, issue.13, pp.2512-2520, 1989.
DOI : 10.1096/fasebj.3.13.2806779

S. Sakuma, Y. Fujimoto, Y. Sakamoto, T. Uchiyama, and K. Yoshioka, Peroxynitrite Induces the Conversion of Xanthine Dehydrogenase to Oxidase in Rabbit Liver, Biochemical and Biophysical Research Communications, vol.230, issue.2, pp.476-485, 1997.
DOI : 10.1006/bbrc.1996.5983

J. Jong, M. Van-der, A. Nieukoop, T. Huizer, and R. Stroeve, Xanthine oxidoreductase activity in perfused hearts of various species, including humans, Circulation Research, vol.67, issue.3, pp.770-773, 1990.
DOI : 10.1161/01.RES.67.3.770

T. Cappola, D. Kass, G. Nelson, R. Berger, and G. Rosas, Allopurinol Improves Myocardial Efficiency in Patients With Idiopathic Dilated Cardiomyopathy, Circulation, vol.104, issue.20, pp.2407-2418, 2001.
DOI : 10.1161/hc4501.098928

U. Ekelund, R. Harrison, O. Shokek, R. Thakkar, and R. Tunin, Intravenous Allopurinol Decreases Myocardial Oxygen Consumption and Increases Mechanical Efficiency in Dogs With Pacing-Induced Heart Failure, Circulation Research, vol.85, issue.5
DOI : 10.1161/01.RES.85.5.437

G. Ellis, R. Anderson, D. Lang, D. Blackman, and R. Morris, Neutrophil superoxide anion???generating capacity, endothelial function and oxidative stress in chronic heart failure: effects of short- and long-term vitamin C therapy, Journal of the American College of Cardiology, vol.36, issue.5, pp.1474-82, 2000.
DOI : 10.1016/S0735-1097(00)00916-5

L. De-biase, P. Pignatelli, L. Lenti, G. Tocci, and F. Piccioni, - production, Thrombosis and Haemostasis, vol.90, pp.317-342, 2003.
DOI : 10.1160/TH03-02-0105

D. Sorescu and K. Griendling, Reactive Oxygen Species, Mitochondria, and NAD(P)H Oxidases in the Development and Progression of Heart Failure, Congestive Heart Failure, vol.100, issue.4, pp.132-172, 2002.
DOI : 10.1161/01.CIR.100.3.292

T. Bleeke, H. Zhang, N. Madamanchi, C. Patterson, and J. Faber, Catecholamine-Induced Vascular Wall Growth Is Dependent on Generation of Reactive Oxygen Species, Circulation Research, vol.94, issue.1, pp.37-45, 2004.
DOI : 10.1161/01.RES.0000109412.80157.7D

L. Xiao, D. Pimentel, J. Wang, K. Singh, and W. Colucci, -adrenoceptor signaling in adult rat cardiac myocytes, American Journal of Physiology-Cell Physiology, vol.49, issue.4, pp.926-960, 2002.
DOI : 10.1016/S0014-2999(99)00222-8

J. Li, N. Gall, D. Grieve, M. Chen, and A. Shah, Activation of NADPH Oxidase During Progression of Cardiac Hypertrophy to Failure, Hypertension, vol.40, issue.4, pp.477-84, 2002.
DOI : 10.1161/01.HYP.0000032031.30374.32

C. Maack, T. Kartes, H. Kilter, H. Schäfers, and G. Nickenig, Oxygen Free Radical Release in Human Failing Myocardium Is Associated With Increased Activity of Rac1-GTPase and Represents a Target for Statin Treatment, Circulation, vol.108, issue.13, pp.1567-74, 2003.
DOI : 10.1161/01.CIR.0000091084.46500.BB

C. Heymes, J. Bendall, P. Ratajczak, A. Cave, and J. Samuel, Increased myocardial NADPH oxidase activity in human heart failure, Journal of the American College of Cardiology, vol.41, issue.12, pp.2164-71, 2003.
DOI : 10.1016/S0735-1097(03)00471-6

M. Terashima, Y. Ohashi, H. Azumi, K. Otsui, and H. Kaneda, Impact of NAD(P)H Oxidase-Derived Reactive Oxygen Species on Coronary Arterial Remodeling: A Comparative Intravascular Ultrasound and Histochemical Analysis of Atherosclerotic Lesions, Circulation: Cardiovascular Interventions, vol.2, issue.3, pp.196-204, 2009.
DOI : 10.1161/CIRCINTERVENTIONS.108.799502

M. Keith, A. Geranmayegan, M. Sole, R. Kurian, and A. Robinson, Increased Oxidative Stress in Patients With Congestive Heart Failure 11This study was supported by a grant jointly sponsored by the Medical Research Council of Canada, Ottawa and Bayer Pharmaceuticals, Etobicoke, Ontario, Canada., Journal of the American College of Cardiology, vol.31, issue.6, pp.1352-1358, 1998.
DOI : 10.1016/S0735-1097(98)00101-6

R. Lebovitz, H. Zhang, H. Vogel, J. Cartwright, . Jr et al., Neurodegeneration, myocardial injury, and perinatal death in mitochondrial superoxide dismutase-deficient mice., Proceedings of the National Academy of Sciences, vol.93, issue.18, pp.9782-9789, 1996.
DOI : 10.1073/pnas.93.18.9782

T. Shiomi, H. Tsutsui, H. Matsusaka, K. Murakami, and S. Hayashidani, Overexpression of Glutathione Peroxidase Prevents Left Ventricular Remodeling and Failure After Myocardial Infarction in Mice, Circulation, vol.109, issue.4, pp.544-553, 2004.
DOI : 10.1161/01.CIR.0000109701.77059.E9

A. Steiber, J. Kerner, and C. Hoppel, Carnitine: a nutritional, biosynthetic, and functional perspective, Molecular Aspects of Medicine, vol.25, issue.5-6, pp.455-73, 2004.
DOI : 10.1016/j.mam.2004.06.006

S. Friedman and G. Fraenkel, Reversible enzymatic acetylation of carnitine, Archives of Biochemistry and Biophysics, vol.59, issue.2, pp.491-501, 1955.
DOI : 10.1016/0003-9861(55)90515-4

K. Buchan and D. Hassall, PPAR agonists as direct modulators of the vessel wall in cardiovascular disease, Medicinal Research Reviews, vol.77, issue.5, pp.350-366, 2000.
DOI : 10.1161/01.ATV.13.4.571

G. Chinetti, F. Gbaguidi, S. Griglio, Z. Mallat, and M. Antonucci, CLA-1/SR-BI Is Expressed in Atherosclerotic Lesion Macrophages and Regulated by Activators of Peroxisome Proliferator-Activated Receptors, Circulation, vol.101, issue.20, pp.2411-2417, 2000.
DOI : 10.1161/01.CIR.101.20.2411

D. Jones, X. Ding, and R. Daynes, Nuclear Receptor Peroxisome Proliferator-activated Receptor ?? (PPAR??) Is Expressed in Resting Murine Lymphocytes, Journal of Biological Chemistry, vol.270, issue.9, pp.6838-6845, 2002.
DOI : 10.1161/01.HYP.36.5.851

]. Finck, B. Kelly, and D. , PGC-1 coactivators: inducible regulators of energy metabolism in health and disease, Journal of Clinical Investigation, vol.116, issue.3, pp.615-637, 2006.
DOI : 10.1172/JCI27794

M. Sano, Y. Izumi, K. Helenius, M. Asakura, and D. Rossi, M??nage-??-Trois 1 Is Critical for the Transcriptional Function of PPAR?? Coactivator 1, Cell Metabolism, vol.5, issue.2, pp.129-171, 2007.
DOI : 10.1016/j.cmet.2007.01.003

G. Martin, K. Schoonjans, A. Lefebvre, B. Staels, and J. Auwerx, Coordinate Regulation of the Expression of the Fatty Acid Transport Protein and Acyl-CoA Synthetase Genes by PPAR?? and PPAR?? Activators, Journal of Biological Chemistry, vol.270, issue.45, pp.28210-28217, 1997.
DOI : 10.1172/JCI117717

B. Frohnert, T. Hui, and D. Bernlohr, Identification of a Functional Peroxisome Proliferator-responsive Element in the Murine Fatty Acid Transport Protein Gene, Journal of Biological Chemistry, vol.267, issue.7, pp.3970-3977, 1999.
DOI : 10.1016/0960-0760(93)90058-5

K. Schoonjans, M. Watanabe, H. Suzuki, A. Mahfoudi, and G. Krey, Induction of the Acyl-Coenzyme A Synthetase Gene by Fibrates and Fatty Acids Is Mediated by a Peroxisome Proliferator Response Element in the C Promoter, Journal of Biological Chemistry, vol.267, issue.33, pp.19269-19276, 1995.
DOI : 10.1146/annurev.nu.14.070194.000503

J. Brandt, F. Djouai, and D. Kelly, Fatty Acids Activate Transcription of the Muscle Carnitine Palmitoyltransferase I Gene in Cardiac Myocytes via the Peroxisome Proliferator-activated Receptor ??, Journal of Biological Chemistry, vol.235, issue.37, pp.23786-23792, 1998.
DOI : 10.1074/jbc.273.15.8560

C. Mascaró, E. Acosta, J. Ortiz, P. Marrero, and F. Hegardt, Control of Human Muscle-type Carnitine Palmitoyltransferase I Gene Transcription by Peroxisome Proliferator-activated Receptor, Journal of Biological Chemistry, vol.269, issue.15, pp.8560-8563, 1998.
DOI : 10.2337/diab.46.7.1230

A. Minnich, N. Tian, L. Byan, and G. Bilder, A potent PPAR?? agonist stimulates mitochondrial fatty acid ??-oxidation in liver and skeletal muscle, American Journal of Physiology-Endocrinology and Metabolism, vol.255, issue.2, pp.270-279, 2001.
DOI : 10.1074/jbc.273.49.32901

A. Gilde, K. Van-der-lee, P. Willemsen, G. Chinetti, and F. Van-der-leij, Peroxisome Proliferator-Activated Receptor (PPAR) alpha and PPARbeta/delta, but not PPARgamma, Modulate the Expression of Genes Involved in Cardiac Lipid Metabolism, Circulation Research, vol.92, issue.5, pp.518-524, 2003.
DOI : 10.1161/01.RES.0000060700.55247.7C

T. Gulick, S. Cresci, T. Caira, D. Moore, and D. Kelly, The peroxisome proliferator-activated receptor regulates mitochondrial fatty acid oxidative enzyme gene expression., Proceedings of the National Academy of Sciences, vol.91, issue.23, pp.11012-11018, 1994.
DOI : 10.1073/pnas.91.23.11012

T. Aoyama, J. Peters, N. Iritani, T. Nakajima, and K. Furihata, Altered Constitutive Expression of Fatty Acid-metabolizing Enzymes in Mice Lacking the Peroxisome Proliferator-activated Receptor ?? (PPAR??), Journal of Biological Chemistry, vol.83, issue.10, pp.5678-84, 1998.
DOI : 10.1016/0002-9343(87)90873-4

J. Ortiz, J. Mallolas, C. Nicot, J. Bofarull, and J. Rodríguez, Isolation of pig mitochondrial 3-hydroxy-3- methylglutaryl-CoA synthase gene promoter : characterization of a peroxisome proliferator-responsive element

J. Rodriguez, G. Gil-gomez, F. Hegardt, and D. Haro, Peroxisome proliferator-activated receptor mediates induction of the mitochondrial 3-hydroxy-3-methylglutaryl-CoA synthase gene by fatty acids, J Biol Chem, vol.269, pp.18767-72, 1994.

J. L. Breslow, Transgenic mouse models of lipoprotein metabolism and atherosclerosis., Proceedings of the National Academy of Sciences, vol.90, issue.18, pp.8314-8322, 1993.
DOI : 10.1073/pnas.90.18.8314

V. Schumaker, M. Phillips, and J. Chatterton, Apolipoprotein B and Low-Density Lipoprotein Structure: Implications for Biosynthesis of Triglyceride-Rich Lipoproteins, Adv Protein Chem, vol.45, pp.205-253, 1994.
DOI : 10.1016/S0065-3233(08)60641-5

R. Shulman, P. Herbert, K. Wehrly, and D. Fredrickson, Thf complete amino acid sequence of C-I (apoLp- Ser), an apolipoprotein from human very low density lipoproteins, J Biol Chem, vol.250, pp.182-90, 1975.

S. Rall, . Jr, K. Weisgraber, and R. Mahley, Human apolipoprotein E. The complete amino acid sequence

B. Staels, J. Dallongeville, J. Auwerx, K. Schoonjans, and E. Leitersdorf, Mechanism of Action of Fibrates on Lipid and Lipoprotein Metabolism, Circulation, vol.98, issue.19, pp.2088-93, 1998.
DOI : 10.1161/01.CIR.98.19.2088

N. Vu-dac, K. Schoonjans, B. Laine, J. Fruchart, and J. Auwerx, Negative regulation of the human apolipoprotein A-I promoter by fibrates can be attenuated by the interaction of the peroxisome proliferatoractivated receptor with its response element, J Biol Chem, vol.269, pp.31012-31020, 1994.

N. Vu-dac, K. Schoonjans, V. Kosykh, J. Dallongeville, and J. Fruchart, Fibrates increase human apolipoprotein A-II expression through activation of the peroxisome proliferator-activated receptor., Journal of Clinical Investigation, vol.96, issue.2, pp.741-50, 1995.
DOI : 10.1172/JCI118118

J. Peters, N. Hennuyer, B. Staels, J. Fruchart, and C. Fievet, Alterations in Lipoprotein Metabolism in Peroxisome Proliferator-activated Receptor ??-deficient Mice, Journal of Biological Chemistry, vol.8, issue.43, pp.27307-27319, 1997.
DOI : 10.1074/jbc.270.33.19269

B. Staels, J. Peinado-onsurbe, and J. Auwerx, Down-regulation of hepatic lipase gene expression and activity by fenofibrate, Biochimica et Biophysica Acta (BBA) - Lipids and Lipid Metabolism, vol.1123, issue.2, pp.227-257, 1992.
DOI : 10.1016/0005-2760(92)90115-C

B. Staels, A. Van-tol, G. Skretting, and J. Auwerx, Lecithin: cholesterol acyltransferase gene expression is regulated in a tissue-selective manner by fibrates, J Lipid Res, vol.33, pp.727-762, 1992.

M. Bouly, D. Masson, B. Gross, X. Jiang, and C. Fievet, Induction of the Phospholipid Transfer Protein Gene Accounts for the High Density Lipoprotein Enlargement in Mice Treated with Fenofibrate, Journal of Biological Chemistry, vol.37, issue.28, pp.25841-25848, 2001.
DOI : 10.1097/00041433-199906000-00007

P. Mardones, A. Pilon, M. Bouly, D. Duran, and T. Nishimoto, Fibrates Down-regulate Hepatic Scavenger Receptor Class B Type I Protein Expression in Mice, Journal of Biological Chemistry, vol.41, issue.10, pp.7884-90, 2003.
DOI : 10.1074/jbc.M002310200

F. Heller and C. Harvengt, Effects of clofibrate, bezafibrate, fenofibrate and probucol on plasma lipolytic enzymes in normolipaemic subjects, European Journal of Clinical Pharmacology, vol.40, issue.1, pp.57-63, 1983.
DOI : 10.1007/BF00544015

C. Malmendier, J. Lontie, C. Delcroix, D. Dubois, and T. Magot, Apolipoproteins C-II and C-III metabolism in hypertriglyceridemic patients Effect of a drastic triglyceride reduction by combined diet restriction and fenofibrate administration, Atherosclerosis, vol.77, issue.2-3, pp.139-188, 1989.
DOI : 10.1016/0021-9150(89)90075-0

I. Wakabayashi and H. Masuda, Lipoprotein (a) as a determinant of arterial stiffness in elderly patients with type 2 diabetes mellitus, Clinica Chimica Acta, vol.373, issue.1-2, pp.127-158, 2006.
DOI : 10.1016/j.cca.2006.05.018

Y. Wang and R. Fitch, Vascular Stiffness: Measurements, Mechanisms and Implications, Current Vascular Pharmacology, vol.2, issue.4, pp.379-84, 2004.
DOI : 10.2174/1570161043385448

A. Levitas, E. Muhammad, G. Harel, A. Saada, and V. Caspi, Familial neonatal isolated cardiomyopathy caused by a mutation in the flavoprotein subunit of succinate dehydrogenase, European Journal of Human Genetics, vol.93, issue.10, pp.1160-1165, 2010.
DOI : 10.1007/BF00711676

J. Pohjoismäki, S. Goffart, R. Taylor, D. Turnbull, and A. Suomalainen, Developmental and Pathological Changes in the Human Cardiac Muscle Mitochondrial DNA Organization, Replication and Copy Number, PLoS ONE, vol.5, issue.5
DOI : 10.1371/journal.pone.0010426.s002

J. Marin-garcia, M. Goldenthal, and G. Moe, Mitochondrial pathology in cardiac failure, Cardiovascular Research, vol.49, issue.1, pp.10426-14417, 2001.
DOI : 10.1016/S0008-6363(00)00241-8

I. Schrijver, L. Pique, I. Traynis, C. Scharfe, and A. Sehnert, Mitochondrial DNA analysis by multiplex denaturing high-performance liquid chromatography and selective sequencing in pediatric patients with cardiomyopathy, Genetics in Medicine, vol.355, issue.2, pp.118-144, 2009.
DOI : 10.1016/S0140-6736(99)05225-3

D. Bonnet, P. De-lonlay, I. Gautier, R. P. Rötig, and A. , Efficiency of metabolic screening in childhood cardiomyopathies, European Heart Journal, vol.19, issue.5, pp.790-793, 1998.
DOI : 10.1053/euhj.1997.0818

A. Buchwald, H. Till, C. Unterberg, R. Oberschmidt, and H. Figulla, Alterations of the mitochondrial respiratory chain in human dilated cardiomyopathy, European Heart Journal, vol.11, issue.6, pp.509-525, 1990.
DOI : 10.1093/oxfordjournals.eurheartj.a059743

C. Izgi, C. Cevik, R. Bakal, and M. Ozkan, Severe obstructive hypertrophic cardiomyopathy occurring secondary to mitochondrial disease, Turk Kardiyol Dern Ars, vol.37, pp.332-338, 2009.

B. Maron, V. Ferrans, and W. Roberts, Ultrastructural features of degenerated cardiac muscle cells in patients with cardiac hypertrophy, Am J Pathol, vol.79, pp.387-434, 1975.

Y. Maingourd, J. Kachaner, L. Fermont, A. Batisse, and D. Sidi, Severe hypotrophic and hypokinetic myocardiopathies secondary to obstructive malformations of the left heart infants, Arch Mal Coeur Vaiss, vol.76, pp.484-92, 1983.

C. Antozzi and M. Zeviani, Cardiomyopathies in disorders of oxidative metabolism, Cardiovascular Research, vol.35, issue.2, pp.184-99, 1997.
DOI : 10.1016/S0008-6363(97)00141-7

A. Papadimitriou, H. Neustein, S. Dimauro, R. Stanton, and N. Bresolin, Histiocytoid Cardiomyopathy of Infancy, Pediatric Research, vol.18, issue.10, pp.1023-1031, 1984.
DOI : 10.1203/00006450-198418100-00023

M. Van-bilsen, F. Van-nieuwenhoven, and G. Van-der-vusse, Metabolic remodelling of the failing heart: beneficial or detrimental?, Cardiovascular Research, vol.314, issue.1, pp.420-428, 2009.
DOI : 10.1124/jpet.104.082636

E. Norton, E. Jackson, M. Turner, R. Virmani, and M. Forman, The effects of intravenous infusions of selective adenosine A1-receptor and A2-receptor agonists on myocardial reperfusion injury, American Heart Journal, vol.123, issue.2, pp.332-340, 1992.
DOI : 10.1016/0002-8703(92)90643-A

W. Schlack, M. Schäfer, A. Uebing, S. Schäfer, and U. Borchard, Adenosine A2-Receptor Activation at Reperfusion Reduces Infarct Size and Improves Myocardial Wall Function in Dog Heart, Journal of Cardiovascular Pharmacology, vol.22, issue.1, pp.89-96, 1993.
DOI : 10.1097/00005344-199307000-00015

C. Murry, R. Jennings, and K. Reimer, Preconditioning with ischemia: a delay of lethal cell injury in ischemic myocardium, Circulation, vol.74, issue.5, pp.1124-1160, 1986.
DOI : 10.1161/01.CIR.74.5.1124

D. Yellon, G. Baxter, and M. Marber, Angina reassessed: pain or protector?, The Lancet, vol.347, issue.9009, pp.1059-62, 1996.
DOI : 10.1016/S0140-6736(96)90613-3

W. Kiesman, E. Elzein, and J. Zablocki, A1 Adenosine Receptor Antagonists, Agonists, and Allosteric Enhancers, Handb Exp Pharmacol, vol.193, pp.25-58, 2009.
DOI : 10.1007/978-3-540-89615-9_2

S. Blaise, J. Alberto, E. Nedelec, A. Ayav, and G. Pourie, Mild Neonatal Hypoxia Exacerbates the Effects of Vitamin-Deficient Diet on Homocysteine Metabolism in Rats, Pediatric Research, vol.22, issue.6, pp.777-82, 2005.
DOI : 10.1007/s007260200000

URL : https://hal.archives-ouvertes.fr/hal-00466555

O. Frankfurt, Immunoassay for single-stranded DNA in apoptotic cells, Methods Mol Biol, vol.282, pp.85-101, 2004.

J. Edge, J. Hodgin, J. Hagaman, and O. Smithies, A noninvasive computerized tail-cuff system for measuring blood pressure in mice, Hypertension, vol.25, pp.1111-1116, 1995.

O. Lowry, N. Rosebrough, A. Farr, and R. Randall, Protein measurement with the Folin phenol reagent, J Biol Chem, vol.193, pp.265-75, 1951.

Z. Chen, S. Chakraborty, and R. Banerjee, Demonstration That Mammalian Methionine Synthases Are Predominantly Cobalamin-loaded, Journal of Biological Chemistry, vol.86, issue.33, pp.19246-19255, 1995.
DOI : 10.1016/S0140-6736(95)90165-5

C. Kutzbach and E. Stokstad, Mammalian methylenetetrahydrofolate reductase Partial purification, properties, and inhibition by S-adenosylmethionine, Biochimica et Biophysica Acta (BBA) - Enzymology, vol.250, issue.3, pp.459-477, 1971.
DOI : 10.1016/0005-2744(71)90247-6

D. Paglia and W. Valentine, Studies on the quantitative and qualitative characterization of erythrocyte glutathione peroxidase, J Lab Clin Med, vol.70, pp.158-69, 1967.

J. Liu, H. Yeo, S. Doniger, and B. Ames, Assay of Aldehydes from Lipid Peroxidation: Gas Chromatography???Mass Spectrometry Compared to Thiobarbituric Acid, Analytical Biochemistry, vol.245, issue.2, pp.161-167, 1997.
DOI : 10.1006/abio.1996.9990

S. Krähenbühl, C. Talos, S. Fischer, and J. Reichen, Toxicity of bile acids on the electron transport chain of isolated rat liver mitochondria*1, Hepatology, vol.19, issue.2, pp.471-479, 1994.
DOI : 10.1016/0270-9139(94)90027-2

M. Malgat, G. Durrieu, and J. Mazat, Enzymatic and Polarographic Measurements of the Respiratory Chain Complexes, pp.357-377, 1999.
DOI : 10.1007/978-3-642-59884-5_27

P. Rustin, D. Chretien, T. Bourgeron, B. Gérard, and A. Rötig, Biochemical and molecular investigations in respiratory chain deficiencies, Clinica Chimica Acta, vol.228, issue.1, pp.35-51, 1994.
DOI : 10.1016/0009-8981(94)90055-8

J. Folch, M. Lees, S. Stanley, and G. , A simple method for the isolation and purification of total lipides from animal tissues, J Biol Chem, vol.226, pp.497-509, 1957.

H. Ono, H. Katagiri, H. Yahagi, N. Sakoda, and H. , Hepatic Akt Activation Induces Marked Hypoglycemia, Hepatomegaly, and Hypertriglyceridemia With Sterol Regulatory Element Binding Protein Involvement, Diabetes, vol.52, issue.12, pp.2905-2923, 2003.
DOI : 10.2337/diabetes.52.12.2905

S. Giannattasio, S. Gagliardi, M. Samaja, and E. Marra, Simultaneous determination of purine nucleotides, their metabolites and ??-nicotinamide adenine dinucleotide in cerebellar granule cells by ion-pair high performance liquid chromatography, Brain Research Protocols, vol.10, issue.3, pp.168-74, 2003.
DOI : 10.1016/S1385-299X(02)00215-5

M. Katayama, Y. Matsuda, K. Shimokawa, S. Tanabe, and S. Kaneko, Simultaneous determination of six adenyl purines in human plasma by high-performance liquid chromatography with fluorescence derivatization, Journal of Chromatography B: Biomedical Sciences and Applications, vol.760, issue.1, pp.159-63, 2001.
DOI : 10.1016/S0378-4347(01)00265-1

R. Guéant-rodriguez, Y. Juillière, M. Nippert, I. Abdelmouttaleb, and B. Herbeth, Left ventricular systolic dysfunction is an independent predictor of homocysteine in angiographically documented patients with or without coronary artery lesions, Journal of Thrombosis and Haemostasis, vol.5, issue.suppl 4, pp.1209-1225, 2007.
DOI : 10.1152/ajpheart.00475.2002

W. Herrmann, M. Herrmann, J. Joseph, and S. Tyagi, Abstract, Clinical Chemical Laboratory Medicine, vol.1156, issue.12, pp.1633-1677, 2007.
DOI : 10.1515/CCLM.2003.222

URL : https://hal.archives-ouvertes.fr/hal-01375359

M. Herrmann, S. Müller, I. Kindermann, L. Günther, and J. König, Plasma B vitamins and their relation to the severity of chronic heart failure, The American Journal of Clinical Nutrition, vol.26, issue.1, pp.117-140, 2007.
DOI : 10.1093/eurheartj/ehi442

P. Chen, R. Poddar, E. Tipa, P. Dibello, and C. Moravec, Homocysteine metabolism in cardiovascular cells and tissues: implications for hyperhomocysteinemia and cardiovascular disease, Advances in Enzyme Regulation, vol.39, issue.1, pp.93-109, 1999.
DOI : 10.1016/S0065-2571(98)00029-6

S. Blaise, E. Nédélec, H. Schroeder, J. Alberto, and C. Bossenmeyer-pourié, Gestational Vitamin B Deficiency Leads to Homocysteine-Associated Brain Apoptosis and Alters Neurobehavioral Development in Rats, The American Journal of Pathology, vol.170, issue.2, pp.667-79, 2007.
DOI : 10.2353/ajpath.2007.060339

URL : https://hal.archives-ouvertes.fr/hal-00466548

D. G. Gardner, S. Chen, D. J. Glenn, and C. Grigsby, Molecular Biology of the Natriuretic Peptide System: Implications for Physiology and Hypertension, Hypertension, vol.49, issue.3, pp.419-445, 2007.
DOI : 10.1161/01.HYP.0000258532.07418.fa

S. Kalra, R. Ahuja, E. Mutti, D. Veber, and S. Seetharam, Cobalamin-mediated regulation of transcobalamin receptor levels in rat organs, Archives of Biochemistry and Biophysics, vol.463, issue.1, pp.128-160, 2007.
DOI : 10.1016/j.abb.2007.03.011

P. Chen, R. Poddar, E. Tipa, P. Dibello, and C. Moravec, Homocysteine metabolism in cardiovascular cells and tissues: implications for hyperhomocysteinemia and cardiovascular disease Nutritional modulation of gene expression and homocysteine utilization by vitamin B12, Adv Enzyme Regul J Biol Chem, vol.39183, issue.278, pp.93-10920778, 1999.

S. Ortiou, J. Alberto, J. Guéant, and M. Merten, Homocysteine Increases Methionine Synthase mRNA Level in Caco-2 Cells, Cellular Physiology and Biochemistry, vol.14, issue.4-6, pp.407-421, 2004.
DOI : 10.1159/000080350

Y. Ho, J. Magnenat, M. Gargano, and J. Cao, The nature of antioxidant defense mechanisms: a lesson from transgenic studies, Environmental Health Perspectives, vol.106, issue.Suppl 5, pp.1219-1247, 1998.
DOI : 10.1289/ehp.98106s51219

G. Upchurch, . Jr, G. Welch, A. Fabian, J. Freedman et al., Homocyst(e)ine Decreases Bioavailable Nitric Oxide by a Mechanism Involving Glutathione Peroxidase, Journal of Biological Chemistry, vol.110, issue.27, pp.17012-17019, 1997.
DOI : 10.1172/JCI118771

E. Nishio and Y. Watanabe, Homocysteine as a modulator of platelet-derived growth factor action in vascular smooth muscle cells: a possible role for hydrogen peroxide, British Journal of Pharmacology, vol.70, issue.2, pp.269-74, 1997.
DOI : 10.1042/bj3140021

K. Robert, J. Nehme, E. Bourdon, G. Pivert, and B. Friguet, Cystathionine ?? Synthase Deficiency Promotes Oxidative Stress, Fibrosis, and Steatosis in Mice Liver, Gastroenterology, vol.128, issue.5, pp.1405-1420, 2005.
DOI : 10.1053/j.gastro.2005.02.034

S. Battaglia-hsu, N. Akchiche, N. Noel, J. Alberto, and E. Jeannesson, Vitamin B12 deficiency reduces proliferation and promotes differentiation of neuroblastoma cells and up-regulates PP2A, proNGF, and TACE, Proceedings of the National Academy of Sciences, vol.391, issue.1, pp.21930-21935, 2009.
DOI : 10.1007/s00216-008-1953-8

E. Ventura, R. Durant, A. Jaussent, M. Picot, and M. Morena, Homocysteine and inflammation as main determinants of oxidative stress in the elderly, Free Radical Biology and Medicine, vol.46, issue.6, pp.737-781, 2009.
DOI : 10.1016/j.freeradbiomed.2008.11.002

E. Rogers, S. Chen, and C. A. , Folate Deficiency and Plasma Homocysteine during Increased Oxidative Stress, New England Journal of Medicine, vol.357, issue.4, pp.421-423, 2007.
DOI : 10.1056/NEJMc066569

]. Werstuck, G. Lentz, S. Dayal, S. Hossain, G. Sood et al., Homocysteine-induced endoplasmic reticulum stress causes dysregulation of the cholesterol and triglyceride biosynthetic pathways, Journal of Clinical Investigation, vol.107, issue.10, pp.1263-73, 2001.
DOI : 10.1172/JCI11596

]. Chanson, A. Rock, E. Martin, J. Liotard, A. Brachet et al., Preferential response of glutathione-related enzymes to folate-dependent changes in the redox state of rat liver, European Journal of Nutrition, vol.55, issue.Suppl 3, pp.204-216, 2007.
DOI : 10.1080/07315724.1993.10718293

]. Chambers, J. Ueland, P. Wright, M. Doré, C. Refsum et al., Investigation of Relationship Between Reduced, Oxidized, and Protein-Bound Homocysteine and Vascular Endothelial Function in Healthy Human Subjects, Circulation Research, vol.89, issue.2
DOI : 10.1161/hh1401.093459

]. Zhang, Q. Zeng, X. Guo, J. , and W. X. , Oxidant stress mechanism of homocysteine potentiating Con A-induced proliferation in murine splenic T lymphocytes, Cardiovascular Research, vol.53, issue.4, pp.1035-1077, 2002.
DOI : 10.1016/S0008-6363(01)00541-7

]. Mansoor, M. Bergmark, C. Svardal, A. Lønning, P. Ueland et al., Redox Status and Protein Binding of Plasma Homocysteine and Other Aminothiols in Patients With Early-Onset Peripheral Vascular Disease : Homocysteine and Peripheral Vascular Disease, Arteriosclerosis, Thrombosis, and Vascular Biology, vol.15, issue.2, pp.232-272, 1995.
DOI : 10.1161/01.ATV.15.2.232

Q. Zhang, X. Zeng, J. Guo, W. X. Chambers, J. Ueland et al., Oxidant stress mechanism of homocysteine potentiating Con A-induced proliferation in murine splenic T lymphocytes, Cardiovascular Research, vol.53, issue.4, pp.1035-1077, 0199.
DOI : 10.1016/S0008-6363(01)00541-7

D. Hoffman, J. Haning, W. Cornatzer, N. Veal, C. Hsieh et al., Effects of a methyl-deficient diet on rat liver phosphatidylcholine biosynthesis Inhibition of lipopolysaccharide-stimulated TNF-alpha promoter activity by S-adenosylmethionine and 5'-methylthioadenosine, Can J Biochem Am J Physiol Gastrointest Liver Physiol, vol.59201287, pp.543-50, 1981.

R. Ventura-clapier, A. Garnier, and V. Veksler, Transcriptional control of mitochondrial biogenesis: the central role of PGC-1??, Cardiovascular Research, vol.279, issue.45, pp.208-225, 2008.
DOI : 10.1074/jbc.M406217200

URL : https://hal.archives-ouvertes.fr/inserm-00290124

S. Murugappa and S. Kunapuli, Farabee MJ. The Nature of ATP and Biological Energy The role of ADP receptors in platelet function, Serial on the Internet Front Biosci, vol.7, issue.11, pp.1977-86, 2002.

T. Corydon, J. Hansen, P. Bross, and T. Jensen, Down-regulation of Hsp60 expression by RNAi impairs folding of medium-chain acyl-CoA dehydrogenase wild-type and disease-associated proteins, Molecular Genetics and Metabolism, vol.85, issue.4, pp.260-70, 2005.
DOI : 10.1016/j.ymgme.2005.04.003

J. Ibdah, H. Paul, Y. Zhao, S. Binford, and K. Salleng, Lack of mitochondrial trifunctional protein in mice causes neonatal hypoglycemia and sudden death, Journal of Clinical Investigation, vol.107, issue.11, pp.1403-1412, 2001.
DOI : 10.1172/JCI12590

C. Meng, J. X. Xia, L. Shen, S. Wang, and X. , Alterations of Mitochondrial Enzymes Contribute to Cardiac Hypertrophy before Hypertension Development in Spontaneously Hypertensive Rats, Journal of Proteome Research, vol.8, issue.5, pp.2463-75, 2009.
DOI : 10.1021/pr801059u

V. Lamounier-zepter, C. Look, J. Alvarez, T. Christ, and U. Ravens, Adipocyte Fatty Acid-Binding Protein Suppresses Cardiomyocyte Contraction: A New Link Between Obesity and Heart Disease, Circulation Research, vol.105, issue.4, pp.326-360, 2009.
DOI : 10.1161/CIRCRESAHA.109.200501

P. Burton, C. Hogben, C. Joannou, A. Clark, and J. Hsuan, Heart Fatty Acid Binding Protein Is a Novel Regulator of Cardiac Myocyte Hypertrophy, Biochemical and Biophysical Research Communications, vol.205, issue.3, pp.1822-1850, 1994.
DOI : 10.1006/bbrc.1994.2882

M. Spaniol, P. Kaufmann, K. Beier, J. Wuthrich, and M. Torok, Mechanisms of liver steatosis in rats with systemic carnitine deficiency due to treatment with trimethylhydraziniumpropionate, Journal of Lipid Research, vol.42, issue.1, pp.144-53, 2003.
DOI : 10.1095/biolreprod64.6.1689

U. Delabar, D. Kloor, G. Luippold, and B. Mühlbauer, Simultaneous determination of adenosine, Sadenosylhomocysteine and S-adenosylmethionine in biological samples using solid-phase extraction and highperformance liquid chromatography, J Chromatogr B Biomed Sci Appl, vol.724, pp.231-239, 1999.

D. Paglia and W. Valentine, Studies on the quantitative and qualitative characterization of erythrocyte glutathione peroxidase, J Lab Clin Med, vol.70, pp.158-69, 1967.

C. Stanley, Carnitine Deficiency Disorders in Children, Annals of the New York Academy of Sciences, vol.93, issue.1, pp.42-51, 2004.
DOI : 10.1196/annals.1320.004

J. Nezu, I. Tamai, A. Oku, R. Ohashi, and H. Yabuuchi, Primary systemic carnitine deficiency is caused by mutations in a gene encoding sodium ion-dependent carnitine transporter, Nature Genetics, vol.325, issue.1, pp.91-95, 1999.
DOI : 10.1038/325641a0

X. Wu, W. Huang, P. Prasad, P. Seth, and D. Rajan, Functional characteristics and tissue distribution pattern of organic cation transporter 2 (OCTN2), an organic cation/carnitine transporter, J Pharmacol Exp Ther, vol.290, pp.1482-92, 1999.

R. Takahashi, T. Asai, H. Murakami, R. Murakami, and M. Tsuzuki, Pressure Overload-Induced Cardiomyopathy in Heterozygous Carrier Mice of Carnitine Transporter Gene Mutation, Hypertension, vol.50, issue.3, pp.497-502, 2007.
DOI : 10.1161/HYPERTENSIONAHA.107.088609

M. Kuwajima, K. Lu, M. Sei, A. Ono, and M. Hayashi, Characteristics of Cardiac Hypertrophy in the Juvenile Visceral Steatosis Mouse with Systemic Carnitine Deficiency, Journal of Molecular and Cellular Cardiology, vol.30, issue.4, pp.773-81, 1998.
DOI : 10.1006/jmcc.1998.0641

J. Nezu, I. Tamai, A. Oku, R. Ohashi, and H. Yabuuchi, Primary systemic carnitine deficiency is caused by mutations in a gene encoding sodium ion-dependent carnitine transporter, Nature Genetics, vol.325, issue.1, pp.91-95, 1999.
DOI : 10.1038/325641a0

A. Lamhonwah, J. Wong, C. Tam, L. Mai, and I. Tein, Organic cation/carnitine transporter family expression patterns in adult murine heart, Pathology - Research and Practice, vol.205, issue.6, pp.395-402, 2009.
DOI : 10.1016/j.prp.2008.12.010

J. Folch, M. Lees, S. Stanley, and G. , A simple method for the isolation and purification of total lipides from animal tissues, J Biol Chem, vol.226, pp.497-509, 1957.

S. Krähenbühl, C. Talos, S. Fischer, and J. Reichen, Toxicity of bile acids on the electron transport chain of isolated rat liver mitochondria*1, Hepatology, vol.19, issue.2, pp.471-480, 1994.
DOI : 10.1016/0270-9139(94)90027-2

J. Madrazo and D. Kelly, The PPAR trio: Regulators of myocardial energy metabolism in health and disease, Journal of Molecular and Cellular Cardiology, vol.44, issue.6, pp.968-75, 2008.
DOI : 10.1016/j.yjmcc.2008.03.021

M. Stavinoha, J. Rayspellicy, M. Essop, C. Graveleau, and E. Abel, Evidence for mitochondrial thioesterase 1 as a peroxisome proliferator-activated receptor-alpha-regulated gene in cardiac and skeletal muscle

T. Aoyama, J. Peters, N. Iritani, T. Nakajima, and K. Furihata, Altered Constitutive Expression of Fatty Acid-metabolizing Enzymes in Mice Lacking the Peroxisome Proliferator-activated Receptor ?? (PPAR??), Journal of Biological Chemistry, vol.83, issue.10, pp.5678-84, 1998.
DOI : 10.1016/0002-9343(87)90873-4

N. Van-vlies, S. Ferdinandusse, M. Turkenburg, R. Wanders, and F. Vaz, PPAR??-activation results in enhanced carnitine biosynthesis and OCTN2-mediated hepatic carnitine accumulation, Biochimica et Biophysica Acta (BBA) - Bioenergetics, vol.1767, issue.9, pp.1134-1176, 2007.
DOI : 10.1016/j.bbabio.2007.07.001

C. Schachtrup, T. Emmler, B. Bleck, A. Sandqvist, and F. Spener, Functional analysis of peroxisome-proliferator-responsive element motifs in genes of fatty acid-binding proteins, Biochemical Journal, vol.382, issue.1, pp.239-284, 2004.
DOI : 10.1042/BJ20031340

K. Kawabe, H. Saegusa, K. Seto, H. Urabe, and K. Motojima, Peroxisome proliferator-activated receptor ?? and its response element are required but not sufficient for transcriptional activation of the mouse heart-type fatty acid binding protein gene, The International Journal of Biochemistry & Cell Biology, vol.37, issue.7, pp.1534-1580, 2005.
DOI : 10.1016/j.biocel.2005.02.011

L. Mikael, J. Genest, . Jr, and R. Rozen, Elevated Homocysteine Reduces Apolipoprotein A-I Expression in Hyperhomocysteinemic Mice and in Males With Coronary Artery Disease, Circulation Research, vol.98, issue.4, pp.564-71, 2006.
DOI : 10.1161/01.RES.0000204825.66410.0b

J. Yideng, L. Zhihong, X. Jiantuan, C. Jun, and L. Guizhong, Homocysteine-Mediated PPAR??,?? DNA Methylation and Its Potential Pathogenic Mechanism in Monocytes, DNA and Cell Biology, vol.27, issue.3, pp.143-50, 2008.
DOI : 10.1089/dna.2007.0658

M. Grube, M. Zu-schwabedissen, H. Präger, D. Haney, J. Möritz et al., Uptake of Cardiovascular Drugs Into the Human Heart: Expression, Regulation, and Function of the Carnitine Transporter OCTN2 (SLC22A5), Circulation, vol.113, issue.8, p.2
DOI : 10.1161/CIRCULATIONAHA.105.586107

P. Puigserver and B. Spiegelman, Peroxisome Proliferator-Activated Receptor-?? Coactivator 1?? (PGC-1??): Transcriptional Coactivator and Metabolic Regulator, Endocrine Reviews, vol.24, issue.1, pp.78-90, 2003.
DOI : 10.1210/er.2002-0012

URL : https://academic.oup.com/edrv/article-pdf/24/1/78/8861037/edrv0078.pdf

P. Puigserver, Z. Wu, C. Park, R. Graves, and M. Wright, A Cold-Inducible Coactivator of Nuclear Receptors Linked to Adaptive Thermogenesis, Cell, vol.92, issue.6, pp.829-868, 1998.
DOI : 10.1016/S0092-8674(00)81410-5

D. Larrouy, H. Vidal, F. Andreelli, M. Laville, and D. Langin, Cloning and mRNA tissue distribution of human PPAR?? coactivator-1, International Journal of Obesity, vol.23, issue.12, pp.1327-1359, 1999.
DOI : 10.1038/sj.ijo.0801106

J. Yoon, P. Puigserver, G. Chen, J. Donovan, and Z. Wu, Control of hepatic gluconeogenesis through the transcriptional coactivator PGC-1, Nature, vol.106, issue.6852, pp.131-139, 2001.
DOI : 10.1172/JCI10268

J. Yoon, G. Xu, J. Deeney, S. Yang, and J. Rhee, Suppression of ?? Cell Energy Metabolism and Insulin Release by PGC-1??, Developmental Cell, vol.5, issue.1, pp.73-83, 2003.
DOI : 10.1016/S1534-5807(03)00170-9

D. Souza, C. Gasparetti, A. Pereira-da-silva, M. Araújo, E. Carvalheira et al., Peroxisome proliferator-activated receptor ????coactivator-1-dependent uncoupling protein-2 expression in pancreatic islets of rats: a novel pathway for neural control of insulin secretion, Diabetologia, vol.46, issue.11, pp.1522-1553, 2003.
DOI : 10.1007/s00125-003-1222-5

S. Schreiber, R. Emter, M. Hock, D. Knutti, and J. Cardenas, The estrogen-related receptor ?? (ERR??) functions in PPAR?? coactivator 1?? (PGC-1??)-induced mitochondrial biogenesis, Proceedings of the National Academy of Sciences, vol.100, issue.21, pp.6472-6479, 2004.
DOI : 10.1073/pnas.2135217100

A. Russell, J. Feilchenfeldt, S. Schreiber, M. Praz, and A. Crettenand, Endurance Training in Humans Leads to Fiber Type-Specific Increases in Levels of Peroxisome Proliferator-Activated Receptor-?? Coactivator-1 and Peroxisome Proliferator-Activated Receptor-?? in Skeletal Muscle, Diabetes, vol.52, issue.12, pp.2874-81, 2003.
DOI : 10.2337/diabetes.52.12.2874

J. Lehman, P. Barger, A. Kovacs, J. Saffitz, and D. Medeiros, Peroxisome proliferator???activated receptor ?? coactivator-1 promotes cardiac mitochondrial biogenesis, Journal of Clinical Investigation, vol.106, issue.7, pp.847-56, 2000.
DOI : 10.1172/JCI10268

Z. Wu, P. Puigserver, U. Andersson, C. Zhang, and G. Adelmant, Mechanisms Controlling Mitochondrial Biogenesis and Respiration through the Thermogenic Coactivator PGC-1, Cell, vol.98, issue.1, pp.115-139, 1999.
DOI : 10.1016/S0092-8674(00)80611-X

C. Tiraby, G. Tavernier, C. Lefort, D. Larrouy, and F. Bouillaud, Acquirement of Brown Fat Cell Features by Human White Adipocytes, Journal of Biological Chemistry, vol.14, issue.35, pp.33370-33376, 2003.
DOI : 10.1042/cs0900243

R. Vega, J. Huss, and D. Kelly, The Coactivator PGC-1 Cooperates with Peroxisome Proliferator-Activated Receptor alpha in Transcriptional Control of Nuclear Genes Encoding Mitochondrial Fatty Acid Oxidation Enzymes, Molecular and Cellular Biology, vol.20, issue.5
DOI : 10.1128/MCB.20.5.1868-1876.2000

D. Iwata, Y. Kato, T. Wakayama, Y. Sai, and Y. Kubo, Involvement of Carnitine/organic Cation Transporter OCTN2 (SLC22A5) in Distribution of its Substrate Carnitine to the Heart, Drug Metabolism and Pharmacokinetics, vol.23, issue.3, pp.207-222, 2008.
DOI : 10.2133/dmpk.23.207

I. Tzoulaki, M. Molokhia, V. Curcin, M. Little, and C. Millett, Risk of cardiovascular disease and all cause mortality among patients with type 2 diabetes prescribed oral antidiabetes drugs: retrospective cohort study using UK general practice research database Homocysteine lowering and cardiovascular events after acute myocardial infarction, BMJ N Engl J Med, vol.339, issue.354, pp.1578-88, 2006.

E. Lonn, S. Yusuf, M. Arnold, P. Sheridan, and J. Pogue, Homocysteine lowering with folic acid and B vitamins in vascular disease, N Engl J Med, vol.354, pp.1567-77, 2006.

W. Rosamond, K. Flegal, K. Furie, A. Go, and K. Greenlund, Heart disease and stroke statistics-2008 update: a report from the American Heart Association Statistics Committee and Stroke Statistics Subcommittee, Circulation, vol.117, pp.25-146, 2008.

M. Rayner, S. Allender, and P. Scarborough, Cardiovascular disease in Europe, European Journal of Cardiovascular Prevention & Rehabilitation, vol.353, issue.2_suppl, pp.43-50, 2009.
DOI : 10.1016/S0140-6736(99)04021-0

F. Fowkes, E. Housley, E. Cawood, C. Macintyre, and C. Ruckley, Edinburgh Artery Study: Prevalence of Asymptomatic and Symptomatic Peripheral Arterial Disease in the General Population, International Journal of Epidemiology, vol.20, issue.2, pp.384-92, 1991.
DOI : 10.1093/ije/20.2.384

A. Hirsch, Z. Haskal, N. Hertzer, C. Bakal, and M. Creager, AHA Practice Guidelines for the management of patients with peripheral arterial disease (lower extremity, renal, mesenteric, and abdominal aortic)

F. Khawaja, K. Bailey, S. Turner, S. Kardia, and T. Mosley, Association of Novel Risk Factors With the Ankle Brachial Index in African American and Non-Hispanic White Populations, Mayo Clinic Proceedings, vol.82, issue.6, pp.709-725, 2007.
DOI : 10.1016/S0025-6196(11)61191-9

M. Mcdermott, L. Ferrucci, J. Guralnik, L. Tian, and D. Green, Elevated Levels of Inflammation, D-Dimer, and Homocysteine Are Associated With Adverse Calf Muscle Characteristics and Reduced Calf Strength in Peripheral Arterial Disease, Journal of the American College of Cardiology, vol.50, issue.9, pp.897-905, 2007.
DOI : 10.1016/j.jacc.2007.05.017

H. Vidula, L. Tian, K. Liu, M. Criqui, and L. Ferrucci, Biomarkers of Inflammation and Thrombosis as Predictors of Near-Term Mortality in Patients with Peripheral Arterial Disease: A Cohort Study, Annals of Internal Medicine, vol.148, issue.2, pp.85-93, 2008.
DOI : 10.7326/0003-4819-148-2-200801150-00003

M. Sentí, X. Nogués, J. Pedro-botet, J. Rubiés-prat, and F. Vidal-barraquer, Lipoprotein profile in men with peripheral vascular disease. Role of intermediate density lipoproteins and apoprotein E phenotypes, Circulation, vol.85, issue.1, pp.30-36, 1992.
DOI : 10.1161/01.CIR.85.1.30

P. Ridker, M. Stampfer, and N. Rifai, Novel Risk Factors for Systemic Atherosclerosis, JAMA, vol.285, issue.19, pp.2481-2486, 2001.
DOI : 10.1001/jama.285.19.2481

D. Wald, M. Law, and M. J. , Serum homocysteine and the severity of coronary artery disease, Thrombosis Research, vol.111, issue.1-2, pp.55-62, 2003.
DOI : 10.1016/j.thromres.2003.08.015

A. Undas, J. Brozek, and A. Szczeklik, Homocysteine and thrombosis: from basic science to clinical evidence, Thrombosis and Haemostasis
DOI : 10.1160/TH05-05-0313

N. Khandanpour, Y. Loke, F. Meyer, B. Jennings, and M. Armon, Homocysteine and Peripheral Arterial Disease: Systematic Review and Meta-analysis, European Journal of Vascular and Endovascular Surgery, vol.38, issue.3, pp.316-338, 2009.
DOI : 10.1016/j.ejvs.2009.05.007

K. Woo, J. Sanderson, Y. Sun, P. Chook, and A. Cheung, Hyperhomocyst(e)inemia is a risk factor for arterial endothelial dysfunction in humans Hyperhomocysteinemia decreases circulating high-density lipoprotein by inhibiting apolipoprotein A-I Protein synthesis and enhancing HDL cholesterol clearance, Circulation Circ Res, vol.101265, issue.99, pp.598-606, 2000.

M. Mcdermott, K. Liu, M. Criqui, K. Ruth, and D. Goff, Ankle-Brachial Index and Subclinical Cardiac and Carotid Disease, American Journal of Epidemiology, vol.162, issue.1, pp.33-41, 2005.
DOI : 10.1016/j.atherosclerosis.2003.09.015

S. Hsu, N. Akchiche, N. Noel, J. Alberto, and E. Jeannesson, Vitamin B12 deficiency reduces proliferation and promotes differentiation of neuroblastoma cells and upregulates PP2A, proNGF and TACE, Proc Natl Acad Sci U S A, vol.106, pp.21930-21935, 2009.

R. Guéant-rodriguez, Y. Juilliére, M. Candito, C. Adjalla, and P. Gibelin, Association of MTRRA66G polymorphism (but not of MTHFR C677T and A1298C, MTRA2756G, TCN C776G) with homocysteine and coronary artery disease in the French population, Thromb Haemost, vol.94, pp.510-515, 2005.

H. Sugiuchi, Y. Uji, H. Okabe, T. Irie, and K. Uekama, Direct measurement of high-density lipoprotein cholesterol in serum with polyethylene glycol-modified enzymes and sulfated alpha-cyclodextrin, Clin Chem, vol.41, pp.717-740, 1995.

W. Friedewald, R. Levy, and D. Fredrickson, Estimation of the concentration of low-density lipoprotein cholesterol in plasma, without use of the preparative ultracentrifuge, Clin Chem, vol.18, pp.499-502, 1972.

L. Lagrost, A. Athias, B. Herbeth, V. Guyard-dangremont, and Y. Artur, Opposite Effects of Cholesteryl Ester Transfer Protein and Phospholipid Transfer Protein on the Size Distribution of Plasma High Density Lipoproteins, Journal of Biological Chemistry, vol.111, issue.32, pp.19058-65, 1996.
DOI : 10.1016/0021-9150(85)90040-1

A. Newman, D. Siscovick, T. Manolio, J. Polak, and L. Fried, Ankle-arm index as a marker of atherosclerosis in the Cardiovascular Health Study. Cardiovascular Heart Study (CHS) Collaborative Research Group, Circulation, vol.88, issue.3, pp.837-882, 1993.
DOI : 10.1161/01.CIR.88.3.837

L. Norgren, W. Hiatt, J. Dormandy, M. Nehler, and K. Harris, Inter-Society Consensus for the Management of Peripheral Arterial Disease (TASC II), European Journal of Vascular and Endovascular Surgery, vol.33, issue.1, pp.1-75, 2007.
DOI : 10.1016/j.ejvs.2006.09.024

A. Pradhan, S. Shrivastava, N. Cook, N. Rifai, and M. Creager, Symptomatic Peripheral Arterial Disease in Women: Nontraditional Biomarkers of Elevated Risk, Circulation, vol.117, issue.6, pp.823-854, 2008.
DOI : 10.1161/CIRCULATIONAHA.107.719369

C. Antoniades, A. Antonopoulos, D. Tousoulis, K. Marinou, and C. Stefanadis, Homocysteine and coronary atherosclerosis: from folate fortification to the recent clinical trials, European Heart Journal, vol.26, issue.2-3, pp.6-15, 2009.
DOI : 10.1016/j.jacc.2004.03.039

J. Carrero, E. López-huertas, L. Salmerón, L. Baró, and R. E. , Daily Supplementation with (n-3) PUFAs, Oleic Acid, Folic Acid, and Vitamins B-6 and E Increases Pain-Free Walking Distance and Improves Risk Factors in Men with Peripheral Vascular Disease, The Journal of Nutrition, vol.109, issue.6, pp.1393-1402, 2005.
DOI : 10.1161/01.CIR.0000129138.08493.4D

D. Masson, M. Koseki, M. Ishibashi, C. Larson, and S. Miller, Increased HDL Cholesterol and ApoA-I in Humans and Mice Treated With a Novel SR-BI Inhibitor, Arteriosclerosis, Thrombosis, and Vascular Biology, vol.29, issue.12, pp.2054-60, 2009.
DOI : 10.1161/ATVBAHA.109.191320

A. Beers, M. Haas, N. Wong, and A. Mooradian, Inhibition of Apolipoprotein AI Gene Expression by Tumor Necrosis Factor ??:?? Roles for MEK/ERK and JNK Signaling, Biochemistry, vol.45, issue.7, pp.2408-2421, 2006.
DOI : 10.1021/bi0518040

M. Taskinen, D. Sullivan, C. Ehnholm, M. Whiting, and D. Zannino, Relationships of HDL Cholesterol, ApoA-I, and ApoA-II With Homocysteine and Creatinine in Patients With Type 2 Diabetes Treated With Fenofibrate, Arteriosclerosis, Thrombosis, and Vascular Biology, vol.29, issue.6
DOI : 10.1161/ATVBAHA.108.178228

A. De-bree, L. Mennen, M. Zureik, V. Ducros, and J. Guilland, Homocysteine is not associated with arterial thickness and stiffness in healthy middle-aged French volunteers, International Journal of Cardiology, vol.113, issue.3, pp.332-372, 2006.
DOI : 10.1016/j.ijcard.2005.11.045

H. Nakhai-pour, D. Grobbee, M. Bots, M. Muller, and Y. Van-der-schouw, Circulating homocysteine and large arterial stiffness and thickness in a population-based sample of middle-aged and elderly men, Journal of Human Hypertension, vol.75, issue.12, pp.942-950, 2007.
DOI : 10.1016/j.atherosclerosis.2004.12.043

D. Rader, Molecular regulation of HDL metabolism and function: implications for novel therapies, Journal of Clinical Investigation, vol.116, issue.12, pp.3090-100, 2006.
DOI : 10.1172/JCI30163