Y. Wang and T. Lobstein, Worldwide trends in childhood overweight and obesity, International Journal of Pediatric Obesity, vol.1, issue.1, pp.11-25, 2006.
DOI : 10.1080/17477160600586747

M. Park, C. Falconer, R. Viner, and S. Kinra, The impact of childhood obesity on morbidity and mortality in adulthood: a systematic review, Obesity Reviews, vol.111, issue.2, 2012.
DOI : 10.1161/01.CIR.0000161798.45728.4D

J. Reilly and J. Kelly, Long-term impact of overweight and obesity in childhood and adolescence on morbidity and premature mortality in adulthood: systematic review, International Journal of Obesity, vol.91, issue.7, 2011.
DOI : 10.3945/ajcn.2010.28701D

H. Arima, F. Barzi, and C. J. , Mortality patterns in hypertension, Journal of Hypertension, vol.29, p.29, 2011.
DOI : 10.1097/01.hjh.0000410246.59221.b1

S. Najjar, A. Scuteri, E. Lakatta, E. Ford, U. Ajani et al., Arterial aging: is it an immutable cardiovascular risk factor? Explaining the decrease in U.S. deaths from coronary disease Endogenous vascular hydrogen peroxide regulates arteriolar tension in vivo Reactive oxygen species as cardiovascular mediators :lessons from endothelial-specific protein overexpression models, Hypertension N Engl J Med Circulation Biochim Biophys Acta, vol.465, issue.1787, pp.454-462, 1980.

M. Freidja, B. Toutain, and A. Caillon, Heme Oxygenase 1 Is Differentially Involved in Blood Flow-Dependent Arterial Remodeling: Role of Inflammation, Oxidative Stress, and Nitric Oxide, Hypertension, vol.58, issue.2, pp.225-231, 2011.
DOI : 10.1161/HYPERTENSIONAHA.111.170266

J. Deverse, K. Bailey, K. Jackson, and A. Passerini, Shear stress modulates RAGE-mediated inflammation in a model of diabetes-induced metabolic stress, American Journal of Physiology-Heart and Circulatory Physiology, vol.55, issue.12, 2012.
DOI : 10.2337/db09-0801

A. Collins, C. Lyon, and X. Xia, Age-Accelerated Atherosclerosis Correlates With Failure to Upregulate Antioxidant Genes, Circulation Research, vol.104, issue.6, pp.42-54, 2009.
DOI : 10.1161/CIRCRESAHA.108.188771

H. Yang, L. Roberts, and M. Shi, Retardation of Atherosclerosis by Overexpression of Catalase or Both Cu/Zn-Superoxide Dismutase and Catalase in Mice Lacking Apolipoprotein E, Circulation Research, vol.95, issue.11, pp.1075-1081, 2004.
DOI : 10.1161/01.RES.0000149564.49410.0d

H. Yang, L. Zhou, and Z. Wang, Overexpression of antioxidant enzymes in ApoE-deficient mice suppresses Benzo(a)pyrene-accelerated atherosclerosis, Atherosclerosis, vol.207, issue.1, pp.51-58, 2009.
DOI : 10.1016/j.atherosclerosis.2009.03.052

S. Schriner, N. Linford, and G. Martin, Extension of Murine Life Span by Overexpression of Catalase Targeted to Mitochondria, Science, vol.308, issue.5730, pp.1909-1911, 2005.
DOI : 10.1126/science.1106653

M. Hebert-schuster, E. Fabre, and V. Nivet-antoine, Catalase polymorphisms and metabolic diseases, Current Opinion in Clinical Nutrition and Metabolic Care, vol.15, issue.4, pp.397-402, 2012.
DOI : 10.1097/MCO.0b013e328354a326

M. Hebert-schuster, C. Cottart, and C. Laguillier-morizot, Catalase rs769214 SNP in elderly malnutrition and during renutrition: Is glucagon to blame?, Free Radical Biology and Medicine, vol.51, issue.8, pp.1583-1588, 2011.
DOI : 10.1016/j.freeradbiomed.2011.07.016

E. Fabre, A. Raynaud-simon, and J. Golmard, Gene polymorphisms of oxidative stress enzymes: prediction of elderly renutrition, The American Journal of Clinical Nutrition, vol.84, issue.5, pp.1504-1512, 2008.
DOI : 10.1007/s11060-007-9347-x

Y. Watanabe, H. Metoki, and T. Ohkubo, Accumulation of common polymorphisms is associated with development of hypertension: a 12-year follow-up from the Ohasama study, Hypertension Research, vol.101, issue.2
DOI : 10.1161/01.CIR.101.17.2060

Z. Jiang, J. Akey, and J. Shi, A polymorphism in the promoter region of catalase is associated with blood pressure levels, Human Genetics, vol.109, issue.1, pp.95-103, 2001.
DOI : 10.1007/s004390100553

X. Zhou, J. Cui, and A. Destefano, Polymorphisms in the Promoter Region of Catalase Gene and Essential Hypertension, Disease Markers, vol.21, issue.1, pp.3-7, 2005.
DOI : 10.1155/2005/487014

M. Mansego, S. Gde, M. Alonso, and M. , Polymorphisms of antioxidant enzymes, blood pressure and risk of hypertension, Journal of Hypertension, vol.29, issue.3, pp.492-500, 2011.
DOI : 10.1097/HJH.0b013e328341f1b2

Z. Wang, Y. Li, and B. Wang, A haplotype of the catalase gene confers an increased risk of essential hypertension in Chinese Han, Human Mutation, vol.297, issue.3, pp.272-278, 2010.
DOI : 10.1038/emm.1999.9

A. Benetos, C. Adamopoulos, and J. Bureau, Determinants of Accelerated Progression of Arterial Stiffness in Normotensive Subjects and in Treated Hypertensive Subjects Over a 6-Year Period, Circulation, vol.105, issue.10, pp.1202-1207, 2002.
DOI : 10.1161/hc1002.105135

S. Visvikis-siest and G. Siest, The STANISLAS Cohort:a 10-year follow-up of supposed healthy families. Gene-environment interactions, reference values and evaluation of biomarkers in prevention of cardiovascular diseases, Clin Chem Lab Med, vol.20, issue.46, pp.733-747, 2008.

M. Zureik, M. Temmar, and C. Adamopoulos, Carotid plaques, but not common carotid intimamedia thickness, are independently associated with aortic stiffness HAS (Haute Autorité de Santé) Clinical practice guidelines. Prise en charge des patients atteints d'hypertension artérielle essentielle. Saint-Denis, France: HAS, Carotid-artery intima and media thickness as a risk factor for myocardial infarction and stroke in older adults, pp.85-93, 2002.

J. Rybka, D. Kupczyk, and K. K?dziora-kornatowska, Age-related changes in an antioxidant defense system in elderly patients with essential hypertension compared with healthy controls, Redox Report, vol.29, issue.3
DOI : 10.1067/mlc.2002.129504

M. Bhattacharya, J. Wang, and F. Ribeiro, Analysis of a missense variant of the human N-formyl peptide receptor that is associated with agonist-independent ??-arrestin association and indices of inflammation, The Pharmacogenomics Journal, vol.280, issue.3, pp.190-199, 2007.
DOI : 10.1073/pnas.78.12.7540

H. Benachour, M. Zaiou, and B. Herbeth, ) c.32C>T SNP is associated with decreased soluble E-selectin levels, Pharmacogenomics, vol.15, issue.6, pp.951-960, 2009.
DOI : 10.1097/gme.0b013e31816d8171

D. Arking, A. Krebsova, M. Macek, and . Sr, Association of human aging with a functional variant of klotho, Proceedings of the National Academy of Sciences, vol.37, issue.3, pp.856-61, 2002.
DOI : 10.1016/S0008-6363(97)00233-2

D. Arking, D. Becker, and L. Yanek, KLOTHO Allele Status and the Risk of Early-Onset Occult Coronary Artery Disease, The American Journal of Human Genetics, vol.72, issue.5, pp.1154-61, 2003.
DOI : 10.1086/375035

D. Arking, G. Atzmon, A. Arking, N. Barzilai, and H. Dietz, Association Between a Functional Variant of the KLOTHO Gene and High-Density Lipoprotein Cholesterol, Blood Pressure, Stroke, and Longevity, Circulation Research, vol.96, issue.4, pp.412-420, 2005.
DOI : 10.1161/01.RES.0000157171.04054.30

Z. Jiang, J. Akey, and J. Shi, A polymorphism in the promoter region of catalase is associated with blood pressure levels, Human Genetics, vol.109, issue.1, pp.95-103, 2001.
DOI : 10.1007/s004390100553

X. Zhou, J. Cui, and A. Destefano, Polymorphisms in the Promoter Region of Catalase Gene and Essential Hypertension, Disease Markers, vol.21, issue.1, pp.3-7, 2005.
DOI : 10.1155/2005/487014

M. Hebert-schuster, C. Cottart, and C. Laguillier-morizot, Catalase rs769214 SNP in elderly malnutrition and during renutrition: Is glucagon to blame?, Free Radical Biology and Medicine, vol.51, issue.8, pp.1583-1591, 2011.
DOI : 10.1016/j.freeradbiomed.2011.07.016

F. Boretti, P. Buehler, D. Agnillo, and F. , Sequestration of extracellular hemoglobin within a haptoglobin complex decreases its hypertensive and oxidative effects in dogs and guinea pigs, Journal of Clinical Investigation, vol.119, issue.10, pp.2271-80, 2009.
DOI : 10.1172/JCI39115DS1

P. Kearney, M. Whelton, K. Reynolds, P. Muntner, P. Whelton et al., Global burden of hypertension: analysis of worldwide data, The Lancet, vol.365, issue.9455, pp.217-240, 2005.
DOI : 10.1016/S0140-6736(05)70151-3

K. Anderson, A Nonproportional Hazards Weibull Accelerated Failure Time Regression Model, Biometrics, vol.47, issue.1, pp.281-289, 1991.
DOI : 10.2307/2532512

A. Cowley and J. , The genetic dissection of essential hypertension, Nature Reviews Genetics, vol.290, issue.11, pp.829-869, 2006.
DOI : 10.1152/physiolgenomics.00089.2002

P. Plouin, L. Batide, A. Fiquet-kempf, B. Rossignol, P. Launay-mignot et al., Arterial hypertension secondary to curable causes in adults], Presse Med, vol.31, pp.371-379, 2002.

K. Rahmouni, M. Correia, W. Haynes, and A. Mark, Obesity-Associated Hypertension: New Insights Into Mechanisms, Hypertension, vol.45, issue.1, pp.9-14, 2005.
DOI : 10.1161/01.HYP.0000151325.83008.b4

A. Sironi, A. Gastaldelli, and A. Mari, Visceral Fat in Hypertension: Influence on Insulin Resistance and ??-Cell Function, Hypertension, vol.44, issue.2, pp.127-160, 2004.
DOI : 10.1161/01.HYP.0000137982.10191.0a

L. Lopez, E. Cook, M. Horng, and L. Hicks, Lifestyle Modification Counseling for Hypertensive Patients: Results From the National Health and Nutrition Examination Survey 1999-2004, American Journal of Hypertension, vol.279, issue.11, pp.325-356, 2009.
DOI : 10.1001/jama.279.11.839

V. Kotsis, S. Stabouli, S. Papakatsika, Z. Rizos, and G. Parati, Mechanisms of obesity-induced hypertension, Hypertension Research, vol.8, issue.5, pp.386-93, 2010.
DOI : 10.1001/jama.290.21.2805

P. Mathieu, P. Pibarot, and J. Despres, Metabolic syndrome: the danger signal in atherosclerosis, Vascular Health and Risk Management, vol.2, issue.3, pp.285-302, 2006.
DOI : 10.2147/vhrm.2006.2.3.285

P. Pauletto and M. Rattazzi, Inflammation and hypertension: the search for a link, Nephrology Dialysis Transplantation, vol.21, issue.4, pp.850-853, 2006.
DOI : 10.1093/ndt/gfl019

C. Chrysohoou, C. Pitsavos, D. Panagiotakos, J. Skoumas, and C. Stefanadis, Association between prehypertension status and inflammatory markers related to atherosclerotic disease*1The ATTICA Study, American Journal of Hypertension, vol.17, issue.7, pp.568-73, 2004.
DOI : 10.1016/j.amjhyper.2004.03.675

Q. Capers, R. Alexander, and P. Lou, Monocyte Chemoattractant Protein-1 Expression in Aortic Tissues of Hypertensive Rats, Hypertension, vol.30, issue.6, pp.1397-402, 1997.
DOI : 10.1161/01.HYP.30.6.1397

G. Luvara, M. Pueyo, and M. Philippe, Chronic Blockade of NO Synthase Activity Induces a Proinflammatory Phenotype in the Arterial Wall : Prevention by Angiotensin II Antagonism, Arteriosclerosis, Thrombosis, and Vascular Biology, vol.18, issue.9, pp.1408-1424, 1998.
DOI : 10.1161/01.ATV.18.9.1408

D. Sanz-rosa, M. Oubina, and E. Cediel, receptor antagonism on vascular and circulating inflammatory mediators in SHR: role of NF-??B/I??B system, American Journal of Physiology-Heart and Circulatory Physiology, vol.288, issue.1, pp.111-115, 2005.
DOI : 10.1161/hy1201.099611

J. Marteau, M. Zaiou, and G. Siest, Genetic determinants of blood pressure regulation, Journal of Hypertension, vol.23, issue.12, pp.2127-2170, 2005.
DOI : 10.1097/01.hjh.0000186024.12364.2e

A. Warnholtz, H. Mollnau, and T. Heitzer, Adverse effects of nitroglycerin treatment on endothelial function, vascular nitrotyrosine levels and cGMP-dependent protein kinase activity in hyperlipidemic Watanabe rabbits, Journal of the American College of Cardiology, vol.40, issue.7, pp.1356-63, 2002.
DOI : 10.1016/S0735-1097(02)02133-2

D. Harrison, T. Guzik, and H. Lob, Inflammation, Immunity, and Hypertension, Hypertension, vol.57, issue.2, pp.132-172, 2011.
DOI : 10.1161/HYPERTENSIONAHA.110.163576

L. Leborgne, J. Maziere, C. Maziere, and M. Andrejak, [Oxidative stress, atherogenesis and cardiovascular risk factors], Arch Mal Coeur Vaiss, vol.95, pp.805-819, 2002.

K. Andrews, C. Triggle, and A. Ellis, NO and the Vasculature: Where Does It Come from and What Does It Do?, Heart Fail Rev, vol.7, pp.423-468, 2002.
DOI : 10.1007/1-4020-7960-5_2

J. Fennell, M. Brosnan, and A. Frater, Adenovirus-mediated overexpression of extracellular superoxide dismutase improves endothelial dysfunction in a rat model of hypertension, Gene Therapy, vol.101, issue.13, pp.110-117, 2002.
DOI : 10.1172/JCI1584

C. Schnackenberg, W. Welch, and C. Wilcox, Normalization of Blood Pressure and Renal Vascular Resistance in SHR With a Membrane-Permeable Superoxide Dismutase Mimetic : Role of Nitric Oxide, Hypertension, vol.32, issue.1, pp.59-64, 1998.
DOI : 10.1161/01.HYP.32.1.59

M. Tschudi, S. Mesaros, T. Luscher, and T. Malinski, Direct In Situ Measurement of Nitric Oxide in Mesenteric Resistance Arteries : Increased Decomposition by Superoxide in Hypertension, Hypertension, vol.27, issue.1, pp.32-37, 1996.
DOI : 10.1161/01.HYP.27.1.32

S. Kerr, M. Brosnan, M. Mcintyre, J. Reid, A. Dominiczak et al., Superoxide Anion Production Is Increased in a Model of Genetic Hypertension : Role of the Endothelium, Hypertension, vol.33, issue.6, pp.1353-1361, 1999.
DOI : 10.1161/01.HYP.33.6.1353

M. Mcintyre, D. Bohr, and A. Dominiczak, Endothelial Function in Hypertension : The Role of Superoxide Anion, Hypertension, vol.34, issue.4, pp.539-584, 1999.
DOI : 10.1161/01.HYP.34.4.539

G. Ehret, Genome-Wide Association Studies: Contribution of Genomics to Understanding Blood Pressure and Essential Hypertension, Current Hypertension Reports, vol.316, issue.5829, pp.17-25, 2010.
DOI : 10.1161/01.CIR.22.5.857

R. Lifton, A. Gharavi, and D. Geller, Molecular Mechanisms of Human Hypertension, Cell, vol.104, issue.4, pp.545-56, 2001.
DOI : 10.1016/S0092-8674(01)00241-0

S. Harrap, Where are all the blood-pressure genes?, The Lancet, vol.361, issue.9375, pp.2149-51, 2003.
DOI : 10.1016/S0140-6736(03)13694-X

S. Sherry, M. Ward, and M. Kholodov, dbSNP: the NCBI database of genetic variation, Nucleic Acids Research, vol.29, issue.1, pp.308-319, 2001.
DOI : 10.1093/nar/29.1.308

E. Morris, Analysis of Complex Disease Association Studies, 2011.

C. Delles, M. Mcbride, D. Graham, S. Padmanabhan, and A. Dominiczak, Genetics of hypertension: From experimental animals to humans, Biochimica et Biophysica Acta (BBA) - Molecular Basis of Disease, vol.1802, issue.12, 2009.
DOI : 10.1016/j.bbadis.2009.12.006

URL : https://hal.archives-ouvertes.fr/hal-00636201

D. Levy, G. Ehret, and K. Rice, Genome-wide association study of blood pressure and hypertension, Nature Genetics, vol.79, issue.6, pp.677-87, 2009.
DOI : 10.1214/07-AOAS102

B. Rigat, C. Hubert, F. Alhenc-gelas, F. Cambien, P. Corvol et al., An insertion/deletion polymorphism in the angiotensin I-converting enzyme gene accounting for half the variance of serum enzyme levels., Journal of Clinical Investigation, vol.86, issue.4, pp.1343-1349, 1990.
DOI : 10.1172/JCI114844

M. Caulfield, P. Munroe, and P. J. , Genome-wide mapping of human loci for essential hypertension, The Lancet, vol.361, issue.9375, pp.2118-2141, 2003.
DOI : 10.1016/S0140-6736(03)13722-1

K. Edwards, C. Hutter, J. Wan, H. Kim, and S. Monks, Genome-wide Linkage Scan for the Metabolic Syndrome: The GENNID Study, Obesity, vol.161, issue.Suppl 1, pp.1596-601, 2008.
DOI : 10.1093/aje/kwi154

W. Hsueh, B. Mitchell, and J. Schneider, QTL Influencing Blood Pressure Maps to the Region of PPH1 on Chromosome 2q31-34 in Old Order Amish, Circulation, vol.101, issue.24, pp.2810-2816, 2000.
DOI : 10.1161/01.CIR.101.24.2810

L. Koivukoski, S. Fisher, and T. Kanninen, Meta-analysis of genome-wide scans for hypertension and blood pressure in Caucasians shows evidence of susceptibility regions on chromosomes 2 and 3, Human Molecular Genetics, vol.13, issue.19, pp.2325-2357, 2004.
DOI : 10.1093/hmg/ddh237

P. Mcardle, H. Dytch, O. Connell, J. Shuldiner, A. Mitchell et al., Homozygosity by descent mapping of blood pressure in the Old Order Amish: evidence for sex specific genetic architecture, BMC Genetics, vol.8, issue.1, p.66, 2007.
DOI : 10.1186/1471-2156-8-66

E. Mocci, M. Concas, and M. Fanciulli, Microsatellites and SNPs linkage analysis in a Sardinian genetic isolate confirms several essential hypertension loci previously identified in different populations, BMC Medical Genetics, vol.16, issue.2, p.81, 2009.
DOI : 10.1016/S0895-7061(02)03247-8

E. Murray, Chapter 8 Contributions of the amygdalar complex to behavior in macaque monkeys, Prog Brain Res, vol.87, pp.167-80, 1991.
DOI : 10.1016/S0079-6123(08)63051-4

M. Perola, K. Kainulainen, and P. Pajukanta, Genome-wide scan of predisposing loci for increased diastolic blood pressure in Finnish siblings, Journal of Hypertension, vol.18, issue.11, pp.1579-85, 2000.
DOI : 10.1097/00004872-200018110-00008

T. Rice, T. Rankinen, and M. Province, Genome-Wide Linkage Analysis of Systolic and Diastolic Blood Pressure : The Quebec Family Study, Circulation, vol.102, issue.16, pp.1956-63, 2000.
DOI : 10.1161/01.CIR.102.16.1956

A. Morrison, A. Brown, S. Kardia, S. Turner, and E. Boerwinkle, Evaluating the Context-Dependent Effect of Family History of Stroke in a Genome Scan for Hypertension, Stroke, vol.34, issue.5, pp.1170-1175, 2003.
DOI : 10.1161/01.STR.0000068780.47411.16

D. Rao, M. Province, and M. Leppert, A genome-wide affected sibpair linkage analysis of hypertension: the HyperGEN network, American Journal of Hypertension, vol.16, issue.2, pp.148-50, 2003.
DOI : 10.1016/S0895-7061(02)03247-8

D. Ge, Y. W. Huang, and J. , Linkage analysis of 2q14-q23 and 5q32 with blood pressure quantitative traits in Chinese sib pairs, Journal of Hypertension, vol.21, issue.2, pp.305-315, 2003.
DOI : 10.1097/00004872-200302000-00021

D. Zhu, H. Wang, and M. Xiong, Linkage of hypertension to chromosome 2q14-q23 in Chinese families, Journal of Hypertension, vol.19, issue.1, pp.55-61, 2001.
DOI : 10.1097/00004872-200101000-00008

K. Aberg, F. Dai, and S. Viali, Suggestive linkage detected for blood pressure related traits on 2q and 22q in the population on the Samoan islands, BMC Medical Genetics, vol.4, issue.5, p.107, 2009.
DOI : 10.1371/journal.pone.0005459

L. Hindorff, P. Sethupathy, and H. Junkins, Potential etiologic and functional implications of genome-wide association loci for human diseases and traits, Proceedings of the National Academy of Sciences, vol.40, issue.3, pp.9362-9369, 2009.
DOI : 10.1038/ng.91

C. Newton-cheh, T. Johnson, and V. Gateva, Genome-wide association study identifies eight loci associated with blood pressure, Nature Genetics, vol.79, issue.6, 2009.
DOI : 10.1126/science.1117196

G. Ehret, P. Munroe, and K. Rice, Genetic variants in novel pathways influence blood pressure and cardiovascular disease risk, Nature, vol.43, issue.7367, pp.103-112, 2011.
DOI : 10.1038/ng.834

G. Perry, N. Dominy, and K. Claw, Diet and the evolution of human amylase gene copy number variation, Nature Genetics, vol.16, issue.10, pp.1256-60, 2007.
DOI : 10.1128/MCB.10.6.2513

P. Smith and N. Day, The Design of Case-Control Studies: The Influence of Confounding and Interaction Effects, International Journal of Epidemiology, vol.13, issue.3, pp.356-65, 1984.
DOI : 10.1093/ije/13.3.356

R. Nzietchueng, S. El, and H. Benachour, Klotho KL-VS genotype is involved in blood pressure regulation, Clinica Chimica Acta, vol.412, issue.19-20, pp.1773-1780, 2011.
DOI : 10.1016/j.cca.2011.05.032

A. Feinberg, Epigenetics at the Epicenter of Modern Medicine, JAMA, vol.299, issue.11, pp.1345-50, 2008.
DOI : 10.1001/jama.299.11.1345

A. Baccarelli, M. Rienstra, and E. Benjamin, Cardiovascular Epigenetics: Basic Concepts and Results From Animal and Human Studies, Circulation: Cardiovascular Genetics, vol.3, issue.6, pp.567-73, 2010.
DOI : 10.1161/CIRCGENETICS.110.958744

URL : http://circgenetics.ahajournals.org/content/circcvg/3/6/567.full.pdf

V. Rakyan, T. Down, D. Balding, and S. Beck, Epigenome-wide association studies for common human diseases, Nature Reviews Genetics, vol.14, issue.8, pp.529-570, 2011.
DOI : 10.1038/sj.ejhg.5201538

K. Makar and C. Wilson, DNA Methylation Is a Nonredundant Repressor of the Th2 Effector Program, The Journal of Immunology, vol.173, issue.7, pp.4402-4408, 2004.
DOI : 10.4049/jimmunol.173.7.4402

Z. Chen, A. Karaplis, and S. Ackerman, Mice deficient in methylenetetrahydrofolate reductase exhibit hyperhomocysteinemia and decreased methylation capacity, with neuropathology and aortic lipid deposition, Human Molecular Genetics, vol.10, issue.5, pp.433-476, 2001.
DOI : 10.1093/hmg/10.5.433

I. Smolarek, E. Wyszko, and A. Barciszewska, Global DNA methylation changes in blood of patients with essential hypertension, Med Sci Monit, vol.16, pp.149-155, 2010.

S. Friso, F. Pizzolo, and S. Choi, Epigenetic control of 11 beta-hydroxysteroid dehydrogenase 2 gene promoter is related to human hypertension, Atherosclerosis, vol.199, issue.2, pp.323-330, 2008.
DOI : 10.1016/j.atherosclerosis.2007.11.029

V. Bollati, J. Schwartz, and R. Wright, Decline in genomic DNA methylation through aging in a cohort of elderly subjects, Mechanisms of Ageing and Development, vol.130, issue.4, pp.234-243, 2009.
DOI : 10.1016/j.mad.2008.12.003

M. Jeffrey, N. C. Craig, and . Wong, Epigenetics: A Reference Manual, 2011.

J. Jason, L. Archibald, and O. Nwanyanwu, Cytokines and Malaria Parasitemia, Clinical Immunology, vol.100, issue.2, pp.208-226, 2001.
DOI : 10.1006/clim.2001.5057

A. Zanchetti, Impact of hypertension and antihypertensive treatment on organ damage, The American Journal of Cardiology, vol.84, issue.2, pp.18-24, 1999.
DOI : 10.1016/S0002-9149(99)00364-1

S. Visvikis-siest, J. Marteau, A. Samara, H. Berrahmoune, M. B. Pfister et al., Peripheral blood mononuclear cells (PBMCs): a possible model for studying cardiovascular biology systems, Clinical Chemical Laboratory Medicine, vol.32, issue.9, pp.1154-68, 2007.
DOI : 10.2337/diabetes.53.7.1857

J. Kontaraki, M. Marketou, E. Zacharis, F. Parthenakis, and P. Vardas, Early cardiac gene transcript levels in peripheral blood mononuclear cells in patients with untreated essential hypertension, Journal of Hypertension, vol.29, issue.4, pp.791-798, 2011.
DOI : 10.1097/HJH.0b013e3283424bc4

S. Pendergrass, E. Hayes, and G. Farina, Limited Systemic Sclerosis Patients with Pulmonary Arterial Hypertension Show Biomarkers of Inflammation and Vascular Injury, PLoS ONE, vol.25, issue.8, p.12106, 2010.
DOI : 10.1371/journal.pone.0012106.s008

C. Duffaut, A. Zakaroff-girard, and V. Bourlier, Interplay Between Human Adipocytes and T Lymphocytes in Obesity: CCL20 as an Adipochemokine and T Lymphocytes as Lipogenic Modulators, Arteriosclerosis, Thrombosis, and Vascular Biology, vol.29, issue.10, pp.1608-1622, 2009.
DOI : 10.1161/ATVBAHA.109.192583

URL : https://hal.archives-ouvertes.fr/inserm-00410481

P. Jeemon, K. Pettigrew, C. Sainsbury, D. Prabhakaran, and S. Padmanabhan, Implications of discoveries from genome-wide association studies in current cardiovascular practice, World Journal of Cardiologyy, vol.3, issue.7, pp.230-277, 2011.
DOI : 10.4330/wjc.v3.i7.230

O. Zuk, E. Hechter, S. Sunyaev, and E. Lander, The mystery of missing heritability: Genetic interactions create phantom heritability, Proceedings of the National Academy of Sciences, vol.43, issue.11, pp.1193-1201, 2012.
DOI : 10.1038/ng.952

J. Bell, P. Tsai, and T. Yang, Epigenome-Wide Scans Identify Differentially Methylated Regions for Age and Age-Related Phenotypes in a Healthy Ageing Population, PLoS Genetics, vol.196, issue.4, p.1002629, 2012.
DOI : 10.1371/journal.pgen.1002629.s012

R. Lister, M. Pelizzola, and R. Dowen, Human DNA methylomes at base resolution show widespread epigenomic differences, Nature, vol.10, issue.7271, pp.315-337, 2009.
DOI : 10.1038/nature08514

M. Tomaszewski, F. Charchar, and C. Nelson, Pathway Analysis Shows Association between FGFBP1 and Hypertension, Journal of the American Society of Nephrology, vol.22, issue.5, pp.947-55, 2011.
DOI : 10.1681/ASN.2010080829

S. Ye, B. Simon, and J. Maloney, Pre???B-Cell Colony-enhancing Factor as a Potential Novel Biomarker in Acute Lung Injury, American Journal of Respiratory and Critical Care Medicine, vol.171, issue.4, pp.361-70, 2005.
DOI : 10.1152/ajpcell.00388.2001

A. Moschen, A. Kaser, and B. Enrich, Visfatin, an Adipocytokine with Proinflammatory and Immunomodulating Properties, The Journal of Immunology, vol.178, issue.3, pp.1748-58, 2007.
DOI : 10.4049/jimmunol.178.3.1748

C. Bernard, R. Merval, B. Esposito, and A. Tedgui, Resistance to Endotoxin Shock in Spontaneously Hypertensive Rats, Hypertension, vol.31, issue.6, pp.1350-1356, 1998.
DOI : 10.1161/01.HYP.31.6.1350

D. Dzielak, The immune system and hypertension., Hypertension, vol.19, issue.1_Suppl, pp.36-44, 1992.
DOI : 10.1161/01.HYP.19.1_Suppl.I36

A. Khraibi, Association Between Disturbances in the Immune System and Hypertension, American Journal of Hypertension, vol.4, issue.7_Pt_1, pp.635-676, 1991.
DOI : 10.1093/ajh/4.7.635

I. Berkestedt, A. Nelson, and M. Bodelsson, Endogenous antimicrobial peptide LL-37 induces human vasodilatation ??? ???A preliminary account of these results has been presented to the 29th Congress of the Scandinavian Society of Anaesthesiology and Intensive Care, Gothenburg, September 5???8, 2007., British Journal of Anaesthesia, vol.100, issue.6, pp.803-812, 2008.
DOI : 10.1093/bja/aen074

M. Saraheimo, C. Forsblom, K. Pettersson-fernholm, A. Flyvbjerg, P. Groop et al., Increased levels of ??-defensin (-1, -2 and -3) in type 1 diabetic patients with nephropathy, Nephrology Dialysis Transplantation, vol.23, issue.3, pp.914-922, 2008.
DOI : 10.1093/ndt/gfm711

B. Chowdhury, G. Mantile-selvaggi, L. Miele, E. Cordella-miele, Z. Zhang et al., Lys 43 and Asp 46 in ??-helix 3 of uteroglobin are essential for its phospholipase A2 inhibitory activity, Biochemical and Biophysical Research Communications, vol.295, issue.4, pp.877-83, 2002.
DOI : 10.1016/S0006-291X(02)00767-2

U. Tietge, C. Maugeais, S. Lund-katz, D. Grass, F. Debeer et al., Human Secretory Phospholipase A2 Mediates Decreased Plasma Levels of HDL Cholesterol and ApoA-I in Response to Inflammation in Human ApoA-I Transgenic Mice, Arteriosclerosis, Thrombosis, and Vascular Biology, vol.22, issue.7, pp.1213-1221, 2002.
DOI : 10.1161/01.ATV.0000023228.90866.29

N. Kanda, C. Hau, Y. Tada, A. Tatsuta, S. Sato et al., Visfatin Enhances CXCL8, CXCL10, and CCL20 Production in Human Keratinocytes, Endocrinology, vol.152, issue.8, pp.3155-64, 2011.
DOI : 10.1210/en.2010-1481

S. Bode, M. Kuhn, and U. Forstermann, Chemotactic peptide FMLP contracts human coronary arteries via cyclooxygenase products, American Journal of Physiology-Heart and Circulatory Physiology, vol.258, issue.3, pp.848-853, 1990.
DOI : 10.1152/ajpheart.1990.258.3.H848

M. Keitoku, M. Kohzuki, and H. Katoh, FMLP Actions and its Binding Sites in Isolated Human Coronary Arteries, Journal of Molecular and Cellular Cardiology, vol.29, issue.3, pp.881-94, 1997.
DOI : 10.1006/jmcc.1996.0291

H. Benachour, M. Zaiou, and A. Samara, Association of human cathelicidin (hCAP-18/LL-37) gene expression with cardiovascular disease risk factors, Nutrition, Metabolism and Cardiovascular Diseases, vol.19, issue.10, pp.720-728, 2009.
DOI : 10.1016/j.numecd.2009.01.001

S. Nadar, A. Blann, and G. Lip, Endothelial Dysfunction: Methods of Assessment and Application to Hypertension, Current Pharmaceutical Design, vol.10, issue.29, pp.3591-605, 2004.
DOI : 10.2174/1381612043382765

Y. Saito, T. Nakamura, and Y. Ohyama, In Vivo klotho Gene Delivery Protects against Endothelial Dysfunction in Multiple Risk Factor Syndrome, Biochemical and Biophysical Research Communications, vol.276, issue.2, pp.767-72, 2000.
DOI : 10.1006/bbrc.2000.3470

G. Dedoussis, A. Kapiri, and A. Samara, Visfatin: The Link Between Inflammation and Childhood Obesity, Diabetes Care, vol.32, issue.6, p.71, 2009.
DOI : 10.2337/dc08-2304

R. Dorschner, V. Pestonjamasp, and S. Tamakuwala, Cutaneous Injury Induces the Release of Cathelicidin Anti-Microbial Peptides Active Against Group A Streptococcus, Journal of Investigative Dermatology, vol.117, issue.1, pp.91-98, 2001.
DOI : 10.1046/j.1523-1747.2001.01340.x

J. Malm, O. Sorensen, and T. Persson, The Human Cationic Antimicrobial Protein (hCAP-18) Is Expressed in the Epithelium of Human Epididymis, Is Present in Seminal Plasma at High Concentrations, and Is Attached to Spermatozoa, Infection and Immunity, vol.68, issue.7, pp.4297-302, 2000.
DOI : 10.1128/IAI.68.7.4297-4302.2000

H. Taal, G. Verwoert, and A. Demirkan, Genome-Wide Profiling of Blood Pressure in Adults and Children, Hypertension, vol.59, issue.2, 2011.
DOI : 10.1161/HYPERTENSIONAHA.111.179481

G. Gibson, Rare and common variants: twenty arguments, Nature Reviews Genetics, vol.27, issue.2, pp.135-180, 2011.
DOI : 10.1186/gm131

P. Tarpey, R. Smith, and E. Pleasance, A systematic, large-scale resequencing screen of X-chromosome coding exons in mental retardation, Nature Genetics, vol.16, issue.5, pp.535-578, 2009.
DOI : 10.1126/science.185.4154.862

S. Dickson, K. Wang, I. Krantz, H. Hakonarson, and D. Goldstein, Rare Variants Create Synthetic Genome-Wide Associations, PLoS Biology, vol.81, issue.1, p.1000294, 2010.
DOI : 10.1371/journal.pbio.1000294.t001

URL : https://doi.org/10.1371/journal.pbio.1000294

R. 1. Carmeliet, P. , and R. K. Jain, Molecular mechanisms and clinical applications of angiogenesis, Nature, vol.464, issue.7347, pp.298-307, 2011.
DOI : 10.1038/nature08889

S. Yla-herttuala, T. T. Rissanen, I. Vajanto, and J. Hartikainen, Vascular Endothelial Growth Factors, Journal of the American College of Cardiology, vol.49, issue.10, pp.1015-1026, 2007.
DOI : 10.1016/j.jacc.2006.09.053

C. Vaklavas, D. Lenihan, R. Kurzrock, and A. M. Tsimberidou, Anti-Vascular Endothelial Growth Factor Therapies and Cardiovascular Toxicity: What Are the Important Clinical Markers to Target?, The Oncologist, vol.15, issue.2, pp.130-141, 2010.
DOI : 10.1634/theoncologist.2009-0252

. Lip, Vascular endothelial growth factor and its receptor, Flt-1, in the plasma of patients with coronary or peripheral atherosclerosis, or Type II diabetes, Clin Sci (Lond), vol.102, pp.187-194, 2002.

. Simoons, Prognostic significance of angiogenic growth factor serum levels in patients with acute coronary syndromes, Circulation, vol.107, pp.524-530, 2003.

K. Katsuki and . Shimada, Expression of vascular endothelial growth factor in patients with acute myocardial infarction, J Am Coll Cardiol, vol.35, pp.968-973, 2000.

K. , S. Tabata, and T. Yamashita, Plasma levels of vascular endothelial growth factor and its soluble receptor (SFlt-1) in essential hypertension, Am J Cardiol, vol.87, pp.805-807

K. Yonemura, M. Higashi, H. Ayaori, F. Nakamura, P. M. Ohsuzu et al., Plasma vascular endothelial growth factor level is elevated in patients with multivessel coronary artery disease, Clin Cardiol, vol.27, pp.281-286, 2004.

D. Dake, Vascular endothelial growth factor enhances atherosclerotic plaque progression, Nat Med, vol.7, pp.425-429, 2001.

K. Becker, R. Nakao-khurana, Z. Zhuang, S. Bhardwaj, M. Murakami et al., Vascular endothelial growth factor (VEGF) expression in human coronary atherosclerotic lesions: possible pathophysiological significance of VEGF in progression of atherosclerosis Angiogenesisdependent and independent phases of intimal hyperplasia, Circulation Circulation, vol.98, issue.110, pp.2108-2116, 1998.

K. S. Moulton, K. Vakili, D. Zurakowski, M. Soliman, C. Butterfield et al., Inhibition of plaque neovascularization reduces macrophage accumulation and progression of advanced atherosclerosis, Proceedings of the National Academy of Sciences, vol.91, issue.2, pp.4736-4741, 2003.
DOI : 10.1161/01.RES.0000028149.15986.4C

M. Vajanto, T. Niemi, K. Hakkinen, S. A. Karkola, M. G. Stacker et al., Vascular endothelial growth factor-D expression in human atherosclerotic lesions, Cardiovasc Res, vol.59, issue.18, pp.971-979, 2003.

W. Abedi, M. Risau, M. Soma, J. F. Laakso, S. Martin et al., VEGF gene transfer reduces intimal thickening via increased production of nitric oxide in carotid arteries, Hum Gene Ther, vol.8, issue.19, pp.1737-1744, 1997.

S. Alitalo and . Yla-herttuala, Gene transfers of vascular endothelial growth factor- A, vascular endothelial growth factor-B, vascular endothelial growth factor-C, and vascular endothelial growth factor-D have no effects on atherosclerosis in hypercholesterolemic low-density lipoprotein-receptor/apolipoprotein B48-deficient mice, Circulation, vol.112, pp.1347-1352, 2005.

B. J. Arsenault, S. M. Boekholdt, J. J. Kastelein-blann, A. D. , F. M. Belgore et al., Lipid parameters for measuring risk of cardiovascular disease, Nature Reviews Cardiology, vol.25, issue.4, pp.197-206, 2000.
DOI : 10.1016/S0828-282X(09)70715-9

I. Setoyama, M. Maruyama, K. Osame, and . Arimura, Serum VEGF--as a prognostic factor of atherosclerosis, Atherosclerosis, vol.194, issue.24, pp.182-188, 2007.

P. Patsch and . Schratzberger, Are plasma VEGF and its soluble receptor sFlt-1 atherogenic risk factors? Cross-sectional data from the SAPHIR study, Atherosclerosis, vol.206, pp.265-269, 2009.

R. S. Ingelsson, S. Vasan, and . Seshadri, Identification of cis-and trans-acting genetic variants explaining up to half the variation in circulating vascular endothelial growth factor levels, Circ Res, vol.109, issue.26, pp.554-563, 2011.

E. Beaud, J. Lecomte, J. Steinmetz, P. Locuty, S. Chevrier-visvikis-siest et al., Objectives, design and recruitment of a familial and longitudinal cohort for studying gene-environment interactions in the field of cardiovascular risk: the Stanislas cohort The STANISLAS Cohort: a 10-year followup of supposed healthy families. Gene-environment interactions, reference values and evaluation of biomarkers in prevention of cardiovascular diseases Estimation of the concentration of low-density lipoprotein cholesterol in plasma, without use of the preparative ultracentrifuge Multiplexed discovery of sequence polymorphisms using base-specific cleavage and MALDI-TOF MS, Clin Chem Lab Med Clin Chem Lab Med Clin Chem Nucleic Acids Res, vol.36, issue.33, pp.35-42, 1972.

P. Maller, P. I. Sklar, M. J. De-bakker, P. C. Daly, W. Yu et al., PLINK: a tool set for whole-genome association and population-based linkage analyses A navigator for human genome epidemiology, Am J Hum Genet Nat Genet, vol.81, issue.32, pp.559-575, 2007.

T. A. Collins and . Manolio, Potential etiologic and functional implications of genome-wide association loci for human diseases and traits, Proc Natl Acad Sci, vol.106, pp.9362-9367, 2009.

M. Van-duijn and L. Peltonen, Loci influencing lipid levels and coronary heart disease risk in 16 European population cohorts, Nat Genet, vol.41, pp.47-55, 2009.

T. Luchner, H. E. Meitinger, F. Wichmann, and . Kronenberg, Genome-wide association analysis of high-density lipoprotein cholesterol in the population-based KORA study sheds new light on intergenic regions, Circ Cardiovasc Genet, vol.1, pp.10-20, 2008.

E. S. Corella, J. M. Tai, G. Ordovas, E. Berglund, P. Vartiainen et al., Six new loci associated with blood low-density lipoprotein cholesterol, high-density lipoprotein cholesterol or triglycerides in humans, Nat Genet, vol.40, pp.189-197, 2008.

G. R. Boehnke, K. L. Abecasis, L. A. Mohlke, A. T. Kraja, D. Vaidya et al., Common variants at 30 loci contribute to polygenic dyslipidemia, Nat Genet, vol.41, issue.37, pp.56-65, 2009.

. Borecki, A bivariate genome-wide approach to metabolic syndrome: STAMPEED consortium, Diabetes, vol.60, pp.1329-1339, 2011.

M. J. Mccarthy, M. R. Daly, N. B. Jarvelin, L. Freimer, and . Peltonen, Genomewide association analysis of metabolic traits in a birth cohort from a founder population, 2009.

V. Mcpherson, M. S. Mooser, and . Sandhu, Genetic variants influencing circulating lipid levels and risk of coronary artery disease, Arterioscler Thromb Vasc Biol, vol.30, pp.2264-2276, 2010.

D. Boehnke, K. L. Schlessinger, G. R. Mohlke, and . Abecasis, Newly identified loci that influence lipid concentrations and risk of coronary artery disease, Nat Genet, vol.40, pp.161-169, 2008.

S. Hunter and D. J. , Biological determinants of and reference values for plasma interleukin-8, monocyte chemoattractant protein-1, epidermal growth factor, and vascular endothelial growth factor: Results from the STANISLAS cohort Gene-environment interactions in human diseases, Clin Chem Nat Rev Genet, vol.52, issue.6, pp.504-510, 2005.

T. A. Manolio, Genomewide Association Studies and Assessment of the Risk of Disease, New England Journal of Medicine, vol.363, issue.2, pp.166-176, 2010.
DOI : 10.1056/NEJMra0905980

. Visvikis-siest, Cardiovascular diseases and genome-wide association studies Third Report of the National Cholesterol Education Program (NCEP) Expert Panel on Detection, Evaluation, and Treatment of High Blood Cholesterol in Adults (Adult Treatment Panel III) final report, Clin Chim Acta Circulation, vol.412, issue.106, pp.1697-1701, 2002.

C. Lawes, S. Vander-hoorn, and A. Rodgers, Global burden of blood-pressure-related disease, 2001, The Lancet, vol.371, issue.9623, pp.1513-1521, 2001.
DOI : 10.1016/S0140-6736(08)60655-8

G. Lip, D. Felmeden, and G. Dwivedi, Antiplatelet agents and anticoagulants for hypertension, Cochrane Database Syst Rev, p.3186, 2001.

T. Heitzer, V. Rudolph, E. Schwedhelm, M. Karstens, K. Sydow et al., Clopidogrel Improves Systemic Endothelial Nitric Oxide Bioavailability in Patients With Coronary Artery Disease: Evidence for Antioxidant and Antiinflammatory Effects, Arteriosclerosis, Thrombosis, and Vascular Biology, vol.26, issue.7, pp.1648-52, 2006.
DOI : 10.1161/01.ATV.0000225288.74170.dc

M. Kazui, Y. Nishiya, T. Ishizuka, K. Hagihara, N. Farid et al., Identification of the Human Cytochrome P450 Enzymes Involved in the Two Oxidative Steps in the Bioactivation of Clopidogrel to Its Pharmacologically Active Metabolite, Drug Metabolism and Disposition, vol.38, issue.1, pp.92-101, 2010.
DOI : 10.1124/dmd.109.029132

A. Shuldiner, O. Connell, J. Bliden, K. Gandhi, A. Ryan et al., Association of Cytochrome P450 2C19 Genotype With the Antiplatelet Effect and Clinical Efficacy of Clopidogrel Therapy, JAMA, vol.302, issue.8, pp.849-57, 2009.
DOI : 10.1001/jama.2009.1232

M. Holmes, P. Perel, T. Shah, A. Hingorani, and J. Casas, CYP2C19 Genotype, Clopidogrel Metabolism, Platelet Function, and Cardiovascular Events, JAMA, vol.306, issue.24, pp.2704-2718, 2011.
DOI : 10.1001/jama.2011.1880

G. Pare, S. Mehta, S. Yusuf, S. Anand, S. Connolly et al., Genotype on Outcomes of Clopidogrel Treatment, New England Journal of Medicine, vol.363, issue.18, pp.1704-1718, 2010.
DOI : 10.1056/NEJMoa1008410

I. Mackenzie, M. Coughtrie, T. Macdonald, and L. Wei, Antiplatelet drug interactions, Journal of Internal Medicine, vol.38, issue.6, pp.516-545, 2010.
DOI : 10.1136/bmj.319.7217.1106

M. Katoh, M. Nakajima, N. Shimada, H. Yamazaki, and T. Yokoi, Inhibition of human cytochrome P450 enzymes by 1,4-dihydropyridine calcium antagonists: prediction of in vivo drug-drug interactions, European Journal of Clinical Pharmacology, vol.55, issue.11-12, pp.843-52, 2000.
DOI : 10.1007/s002280050706

T. Gremmel, S. Steiner, D. Seidinger, R. Koppensteiner, S. Panzer et al., Calcium-channel blockers decrease clopidogrel-mediated platelet inhibition, Heart, vol.96, issue.3, pp.186-195, 2010.
DOI : 10.1136/hrt.2009.171488

J. Siller-matula, I. Lang, G. Christ, and B. Jilma, Calcium-Channel Blockers Reduce the Antiplatelet Effect of Clopidogrel, Journal of the American College of Cardiology, vol.52, issue.19, pp.1557-63, 2008.
DOI : 10.1016/j.jacc.2008.07.055

S. Lee, J. Bae, K. Park, S. Rha, J. Bae et al., Inhibitory Interaction Between Calcium Channel Blocker and Clopidogrel, Circulation Journal, vol.75, issue.11, pp.2581-2590, 2011.
DOI : 10.1253/circj.CJ-11-0113

C. Good, S. Steinhubl, D. Brennan, A. Lincoff, E. Topol et al., Is There a Clinically Significant Interaction Between Calcium Channel Antagonists and Clopidogrel?: Results From the Clopidogrel for the Reduction of Events During Observation (CREDO) Trial, Circulation: Cardiovascular Interventions, vol.5, issue.1, pp.77-81, 2012.
DOI : 10.1161/CIRCINTERVENTIONS.111.963405

J. Olesen, G. Gislason, M. Charlot, E. Fosbol, C. Andersson et al., Calcium-Channel Blockers Do Not Alter the Clinical Efficacy of Clopidogrel After Myocardial Infarction, Journal of the American College of Cardiology, vol.57, issue.4, pp.409-426, 2011.
DOI : 10.1016/j.jacc.2010.08.640

K. Thygesen, J. Alpert, H. White, E. Joint, and . Aha, WHF Task Force for the Redifinition of Myocardial Infarction. Universal definition of myocardial infarction, Eur Heart J, vol.28, pp.2525-2563, 2007.

J. Anderson, C. Adams, E. Antman, C. Bridges, R. Califf et al., ACC/AHA 2007 Guidelines for the Management of Patients With Unstable Angina/Non???ST-Elevation Myocardial Infarction, Journal of the American College of Cardiology, vol.50, issue.7, 2002.
DOI : 10.1016/j.jacc.2007.02.013

S. Purcell, N. B. Todd-brown, K. Thomas, L. Ferreira, M. Bender et al., PLINK: A Tool Set for Whole-Genome Association and Population-Based Linkage Analyses, The American Journal of Human Genetics, vol.81, issue.3, pp.559-75, 2007.
DOI : 10.1086/519795

Z. Ding, S. Kim, R. Dorsam, J. J. Kunapuli, and S. , Inactivation of the human P2Y12 receptor by thiol reagents requires interaction with both extracellular cysteine residues, Cys17 and Cys270, Blood, vol.101, issue.10, pp.3908-3922, 2003.
DOI : 10.1182/blood-2002-10-3027

A. Wihlborg, L. Wang, O. Braun, A. Eyjolfsson, R. Gustafsson et al., ADP Receptor P2Y12 Is Expressed in Vascular Smooth Muscle Cells and Stimulates Contraction in Human Blood Vessels, Arteriosclerosis, Thrombosis, and Vascular Biology, vol.24, issue.10, pp.1810-1815, 2004.
DOI : 10.1161/01.ATV.0000142376.30582.ed

F. Giachini, D. Osmond, S. Zhang, F. Carneiro, V. Lima et al., Clopidogrel, independent of vascular P2Y12 receptor, improves the arterial function in small mesenteric arteries from Ang II-hypertensive rats, Clin Sci, 2009.

E. Bates, W. Lau, and D. Angiolillo, Clopidogrel???Drug Interactions, Journal of the American College of Cardiology, vol.57, issue.11, pp.1251-63, 2011.
DOI : 10.1016/j.jacc.2010.11.024

T. Ma, Y. Lam, V. Tan, T. Kiernan, and B. Yan, Impact of genetic and acquired alteration in cytochrome P450 system on pharmacologic and clinical response to clopidogrel, Pharmacology & Therapeutics, vol.125, issue.2
DOI : 10.1016/j.pharmthera.2009.10.008

M. Graciano, A. Nishiyama, K. Jackson, D. Seth, R. Ortiz et al., Purinergic receptors contribute to early mesangial cell transformation and renal vessel hypertrophy during angiotensin II-induced hypertension, American Journal of Physiology-Renal Physiology, vol.294, issue.1, pp.161-170, 2008.
DOI : 10.1161/01.HYP.27.3.658

M. Abbracchio, G. Burnstock, J. Boeynaems, E. Barnard, J. Boyer et al., International Union of Pharmacology LVIII: Update on the P2Y G Protein-Coupled Nucleotide Receptors: From Molecular Mechanisms and Pathophysiology to Therapy, Pharmacological Reviews, vol.58, issue.3, pp.281-341, 2006.
DOI : 10.1124/pr.58.3.3

M. Schmidt, M. Johansen, D. Robertson, M. Maeng, A. Kaltoft et al., Use of clopidogrel and calcium channel blockers and risk of major adverse cardiovascular events, European Journal of Clinical Investigation, vol.56, issue.3, pp.266-74, 2012.
DOI : 10.1016/S0895-4356(02)00591-7

S. De-morais, G. Wilkinson, J. Blaisdell, K. Nakamura, U. Meyer et al., The major genetic defect responsible for the polymorphism of S-mephenytoin metabolism in humans, J Biol Chem, vol.269, pp.15419-15441, 1994.

Y. Ma, W. Ni, W. Zhu, Y. Xiong, and X. Deng, Association of genetic polymorphisms of CYP 2C19 with hypertension in a Chinese Han population, Blood Pressure, vol.11, issue.3, pp.166-70, 2011.
DOI : 10.1161/01.RES.87.11.992

H. Yamazaki and T. Shimada, Progesterone and Testosterone Hydroxylation by Cytochromes P450 2C19, 2C9, and 3A4 in Human Liver Microsomes, Archives of Biochemistry and Biophysics, vol.346, issue.1, pp.161-170, 1997.
DOI : 10.1006/abbi.1997.0302

L. Gomes, N. Huang, V. Agrawal, B. Mendonca, T. Bachega et al., Extraadrenal 21-Hydroxylation by CYP2C19 and CYP3A4: Effect on 21-Hydroxylase Deficiency, The Journal of Clinical Endocrinology & Metabolism, vol.94, issue.1, pp.89-95, 2009.
DOI : 10.1210/jc.2008-1174