M. Benzinou, J. Creemers, H. Choquet, S. Lobbens, C. Dina et al., Common nonsynonymous variants in PCSK1 confer risk of obesity, Nature Genetics, vol.269, issue.8, pp.943-948, 2008.
DOI : 10.1093/hmg/ddl111

G. Med, REFERENCES BIBLIOGRAPHIQUES 1 The epidemiology of obesity: the size of the problem, James, W.P. J Intern Med, vol.2, issue.263, pp.36-336, 2008.

T. J. Cole, M. C. Bellizzi, K. M. Flegal, and W. H. Dietz, Establishing a standard definition for child overweight and obesity worldwide: international survey, BMJ, vol.320, issue.7244, pp.1240-1243, 2000.
DOI : 10.1136/bmj.320.7244.1240

S. Chinn, Definitions of childhood obesity: current practice, European Journal of Clinical Nutrition, vol.105, issue.10, pp.1189-94, 2006.
DOI : 10.1038/sj.ejcn.1602436

E. M. Poskitt, Defining childhood obesity: the relative body mass index (BMI), Acta Paediatrica, vol.39, issue.8, 1995.
DOI : 10.1016/0026-0495(77)90082-8

D. D. Hensrud and S. Klein, Extreme Obesity: A New Medical Crisis in the United States, Mayo Clinic Proceedings, vol.81, issue.10, pp.5-10, 2006.
DOI : 10.1016/S0025-6196(11)61175-0

R. R. Kipping, R. Jago, D. A. Lawlor, C. J. Bakker, and K. Van-der-kooy, Obesity in children Part 1: Epidemiology, measurement, risk factors, and screening Imaging techniques for measuring adipose-tissue distribution--a comparison between computed tomography and 1.5- T magnetic resonance, Bmj Am J Clin Nutr, vol.337, issue.51, pp.953-960, 1824.

D. H. Bessesen, Update on Obesity, The Journal of Clinical Endocrinology & Metabolism, vol.93, issue.6, pp.2027-2061, 2008.
DOI : 10.1210/jc.2008-0520

L. Lissner, A. Sohlstrom, E. Sundblom, A. Sjoberg, T. S. Olds et al., Trends in overweight and obesity in Swedish schoolchildren 1999-2005: has the epidemic reached a plateau Trends in the prevalence of childhood overweight and obesity in Australia between Obesity prevalence from a European perspective: a systematic review American Heart Association Childhood Obesity Research Summit: executive summary, Obes Rev Int J Obes (Lond) BMC Public Health Circulation, vol.11, issue.119, pp.553-562, 1985.

R. C. Whitaker, J. A. Wright, M. S. Pepe, K. D. Seidel, and W. H. Dietz, Predicting Obesity in Young Adulthood from Childhood and Parental Obesity, New England Journal of Medicine, vol.337, issue.13, pp.869-73, 1997.
DOI : 10.1056/NEJM199709253371301

K. F. Adams, A. Schatzkin, T. B. Harris, V. Kipnis, T. Mouw et al., A potential decline in life expectancy in the United States in the 21st century Diabetes mellitus and its chronic complications Obesity and dyslipidemia The disease burden associated with overweight and obesity Obesity: prevalence, theories, medical consequences, management, and research directions Extreme childhood obesity is associated with increased risk for gastroesophageal reflux disease in a large population-based study Obesity and urinary incontinence: epidemiology and clinical research update The prevalence and etiology of nongenetic obesity and associated disorders Psychiatric symptoms among prospective bariatric surgery patients: rates of prevalence and their relation to social desirability, pursuit of surgery, and follow-up attendance Social and economic consequences of overweight in adolescence and young adulthood Position of the American Dietetic Association: weight management How effective are traditional dietary and exercise interventions for weight loss? Management of obesity as a chronic disease: nonpharmacologic, pharmacologic, and surgical options Long term pharmacotherapy for obesity and overweight: updated meta-analysis Long-term persistence with orlistat and sibutramine in a population-based cohort, viii. 18. Must, pp.763-78, 1993.

D. S. Weigle, L. Sjostrom, A. K. Lindroos, M. Peltonen, J. Torgerson et al., The obesity epidemic: pharmacological challenges Lifestyle, diabetes, and cardiovascular risk factors 10 years after bariatric surgery Effects of bariatric surgery on mortality in Swedish obese subjects A decade of change in obesity surgery Bariatric surgery trends: an 18-year report from the International Bariatric Surgery Registry Trends in bariatric surgical procedures Long-term mortality after gastric bypass surgery Obesity and its surgical management Medical and surgical options in the treatment of severe obesity Clinical update: bariatric surgery, Mol Interv N Engl J Med N Engl J Med National Bariatric Surgery Registry (NBSR) Contributors. Obes Surg Am J Surg Jama N Engl J Med Am J Surg Am J Surg Lancet, vol.8, issue.370, pp.82-98, 1997.

O. 'brien, P. E. Dixon, J. B. Laurie, C. Skinner, S. Proietto et al., Treatment of Mild to Moderate Obesity with Laparoscopic Adjustable Gastric Banding or an Intensive Medical Program, Annals of Internal Medicine, vol.144, issue.9, pp.625-658, 2006.
DOI : 10.7326/0003-4819-144-9-200605020-00005

T. H. Inge, S. A. Xanthakos, and M. H. Zeller, Bariatric surgery for pediatric extreme obesity: now or later?, International Journal of Obesity, vol.38, issue.1, pp.1-14, 2007.
DOI : 10.1038/sj.ijo.0803525

D. Dan, D. Harnanan, S. Seetahal, V. Naraynsingh, S. Teelucksingh et al., Bariatric surgery in the management of childhood obesity: should there be an age limit? Obes Surg Lipectomy as a new approach to secondary procedure superficialization of direct autogenous forearm radial-cephalic arteriovenous accesses for hemodialysis Prevention of pediatric overweight and obesity An update in prevention and treatment of pediatric obesity Downward trends in the prevalence of childhood overweight in the setting of 12-year school-and community-based programmes Preventing childhood obesity: what works? Ten putative contributors to the obesity epidemic Alterations of lipid metabolism and gene expression in rat adipocytes during chronic olanzapine treatment, J Vasc Surg Pediatrics World J Pediatr Public Health Nutr Int J Obes (Lond) Crit Rev Food Sci Nutr Mol Psychiatry, vol.20, issue.12, pp.114-121, 2003.

H. S. Vestri, L. Maianu, D. R. Moellering, and W. T. Garvey, Atypical Antipsychotic Drugs Directly Impair Insulin Action in Adipocytes: Effects on Glucose Transport, Lipogenesis, and Antilipolysis, Neuropsychopharmacology, vol.184, issue.4, pp.765-72, 2007.
DOI : 10.1038/sj.npp.1301142

D. B. Allison, J. L. Mentore, M. Heo, L. P. Chandler, J. C. Cappelleri et al., Antipsychotic-induced weight gain: a comprehensive research synthesis Concentrations of urinary phthalate metabolites are associated with increased waist circumference and insulin resistance in adult U.S. males Infectobesity: obesity of infectious origin, Am J Psychiatry Environ Health Perspect, vol.156, issue.115, pp.1686-96, 1999.

B. R. Bistrian, L. J. Khaodhiar, M. B. Mattock, G. D. Chusney, D. F. Burt et al., Chronic systemic inflammation in overweight and obese adults NIDDM as a disease of the innate immune system: association of acute-phase reactants and interleukin-6 with metabolic syndrome X. Diabetologia The gut microbiota as an environmental factor that regulates fat storage An obesity-associated gut microbiome with increased capacity for energy harvest Quantitative differences in intestinal Faecalibacterium prausnitzii in obese Indian children, Adv Food Nutr Res Proc Natl Acad Sci U S A Nature Br J Nutr, vol.52, issue.103, pp.61-102, 1997.

M. Kalliomaki, M. C. Collado, S. Salminen, E. Isolauri, R. E. Ley et al., Microbial ecology: human gut microbes associated with obesity A human gut microbial gene catalogue established by metagenomic sequencing Obesity in childhood: A community study in Newcastle upon Tyne Risk factors for obesity in 7- year-old European children: the Auckland Birthweight Collaborative Study Timing of nutrient restriction and programming of fetal adipose tissue development Sleep deprivation in the rat: X. Integration and discussion of the findings Biological, physiological, pathophysiological, and pharmacological aspects of ghrelin Leptin levels are dependent on sleep duration: relationships with sympathovagal balance, carbohydrate regulation, cortisol, and thyrotropin, Am J Clin Nutr Nature Nature Lancet Arch Dis Child Proc Nutr Soc Sleep Endocr Rev J Clin Endocrinol Metab, vol.87, issue.89, pp.534-542, 1977.

S. Taheri, L. Lin, D. Austin, T. Young, E. J. Mignot et al., Short sleep duration is associated with reduced leptin, elevated ghrelin, and increased body mass index Energy Balance and Obesity in Man Effect of ambience on food intake and food choice (1962) Diabetes mellitus: a "thrifty" genotype rendered detrimental by "progress"? A nonadaptive scenario explaining the genetic predisposition to obesity: the "predation release" hypothesis, e62. 70. Garrow, pp.821-859, 1978.

J. C. Jimenez-chillaron, E. Isganaitis, M. Charalambous, S. Gesta, T. Pentinat-pelegrin et al., Intergenerational Transmission of Glucose Intolerance and Obesity by In Utero Undernutrition in Mice, Diabetes, vol.58, issue.2, pp.460-468, 2009.
DOI : 10.2337/db08-0490

D. Suner, J. C. Cigudosa, M. Urioste, and J. Benitez, Epigenetic differences arise during the lifetime of monozygotic twins The epigenetic basis of twin discordance in age-related diseases Hypothalamic proopiomelanocortin promoter methylation becomes altered by early overfeeding: an epigenetic model of obesity and the metabolic syndrome, Proc Natl Acad Sci U S A Pediatr Res J Physiol, vol.102, issue.587, pp.10604-10613, 2005.

F. I. Milagro, J. Campion, D. F. Garcia-diaz, E. Goyenechea, L. Paternain et al., High fat diet-induced obesity modifies the methylation pattern of leptin promoter in rats Metabolic plasticity during mammalian development is directionally dependent on early nutritional status Gene-nutrient interactions and DNA methylation, 81. Friso, S. and Choi, S.W. (2005) Gene-nutrient interactions in one-carbon metabolism, pp.1-9, 2002.

E. L. Sullivan, K. L. Grove, L. Bouchard, R. Rabasa-lhoret, M. Faraj et al., Metabolic imprinting in obesity Differential epigenomic and transcriptomic responses in subcutaneous adipose tissue between low and high responders to caloric restriction Heritability in the genomics era-concepts and misconceptions Genetics of body-weight regulation, Curr Drug Metab Forum Nutr Am J Clin Nutr Nat Rev Genet Nature, vol.6, issue.404, pp.37-46, 2000.

J. Wardle, S. Carnell, C. M. Haworth, and R. Plomin, Evidence for a strong genetic influence on childhood adiposity despite the force of the obesogenic environment, Am J Clin Nutr, vol.87, pp.398-404, 2008.

I. S. Farooqi, O. Rahilly, S. Farooqi, I. S. Keogh, J. M. Yeo et al., Mutations in ligands and receptors of the leptin???melanocortin pathway that lead to obesity, Nature Clinical Practice Endocrinology & Metabolism, vol.7, issue.10, pp.569-77, 2003.
DOI : 10.1038/ncpendmet0966

I. S. Farooqi, K. Volders, R. Stanhope, R. Heuschkel, A. White et al., Hyperphagia and early-onset obesity due to a novel homozygous missense mutation in prohormone convertase 1/3 Clinical and molecular genetic spectrum of congenital deficiency of the leptin receptor Functional characterization of human NTRK2 mutations identified in patients with severe early-onset obesity, ) Hyperphagia, severe obesity, impaired cognitive function, and hyperactivity associated with functional loss of one copy of the brainderived neurotrophic factor (BDNF) gene. Diabetes, pp.3369-73, 2000.

R. S. Jackson, J. W. Creemers, I. S. Farooqi, M. L. Raffin-sanson, A. Varro et al., Small-intestinal dysfunction accompanies the complex endocrinopathy of human proprotein convertase 1 deficiency, Journal of Clinical Investigation, vol.112, issue.10, pp.1550-60, 1997.
DOI : 10.1172/JCI200318784

H. Krude, H. Biebermann, W. Luck, R. Horn, G. Brabant et al., Severe early-onset obesity, adrenal insufficiency and red hair pigmentation caused by POMC mutations in humans, Nature Genetics, vol.19, issue.2, pp.155-162, 1998.
DOI : 10.1038/509

C. T. Montague, I. S. Farooqi, J. P. Whitehead, M. A. Soos, H. Rau et al., Congenital leptin deficiency is associated with severe early-onset obesity in humans (1998) A leptin missense mutation associated with hypogonadism and morbid obesity A frameshift mutation in human MC4R is associated with a dominant form of obesity, Nature Nat Genet Nat Genet, vol.387, issue.20, pp.903-911, 1997.

G. S. Yeo, C. Hung, C. C. Rochford, J. Keogh, J. Gray et al., A de novo mutation affecting human TrkB associated with severe obesity and developmental delay, Nature Neuroscience, vol.127, issue.11, pp.1187-1196, 2004.
DOI : 10.1038/ng0896-485

G. S. Yeo, I. S. Farooqi, S. Aminian, D. J. Halsall, R. G. Stanhope et al., A frameshift mutation in MC4R associated with dominantly inherited human obesity, Nature Genetics, vol.20, issue.2, pp.111-113, 1998.
DOI : 10.1038/2404

G. S. Yeo, E. J. Lank, I. S. Farooqi, J. Keogh, B. G. Challis et al., Mutations in the human melanocortin-4 receptor gene associated with severe familial obesity disrupts receptor function through multiple molecular mechanisms, Human Molecular Genetics, vol.12, issue.5, pp.561-74, 2003.
DOI : 10.1093/hmg/ddg057

I. S. Farooqi, G. Matarese, G. M. Lord, J. M. Keogh, E. Lawrence et al., Beneficial effects of leptin on obesity, T cell hyporesponsiveness, and neuroendocrine/metabolic dysfunction of human congenital leptin deficiency, Journal of Clinical Investigation, vol.110, issue.8, pp.1093-103, 2002.
DOI : 10.1172/JCI0215693

M. Ozata, G. Uckaya, Z. Beyhan, and I. C. Ozdemir, Plasma leptin levels in male patients with idiopathic central diabetes insipidus, Journal of Endocrinological Investigation, vol.82, issue.part2, pp.451-455, 1999.
DOI : 10.1007/BF03343589

J. C. Han, Q. R. Liu, M. Jones, R. L. Levinn, C. M. Menzie et al., Brain-Derived Neurotrophic Factor and Obesity in the WAGR Syndrome, New England Journal of Medicine, vol.359, issue.9, pp.918-945, 2008.
DOI : 10.1056/NEJMoa0801119

I. S. Farooqi, S. A. Jebb, G. Langmack, E. Lawrence, C. H. Cheetham et al., Effects of Recombinant Leptin Therapy in a Child with Congenital Leptin Deficiency, New England Journal of Medicine, vol.341, issue.12, pp.879-84, 1999.
DOI : 10.1056/NEJM199909163411204

M. A. Delrue and J. L. Michaud, Fat chance: genetic syndromes with obesity, Clinical Genetics, vol.8, issue.2, pp.83-93, 2004.
DOI : 10.1111/j.0009-9163.2004.00300.x

T. Sahoo, D. Del-gaudio, J. R. German, M. Shinawi, S. U. Peters et al., Prader-Willi phenotype caused by paternal deficiency for the HBII-85 C/D box small nucleolar RNA cluster, Nature Genetics, vol.40, issue.6, pp.719-740, 2008.
DOI : 10.1371/journal.pgen.0030235

A. J. De-smith, C. Purmann, R. G. Walters, R. J. Ellis, S. E. Holder et al., A deletion of the HBII-85 class of small nucleolar RNAs (snoRNAs) is associated with hyperphagia, obesity and hypogonadism, Human Molecular Genetics, vol.18, issue.17, pp.3257-65, 2009.
DOI : 10.1093/hmg/ddp263

A. L. Duker, B. C. Ballif, E. V. Bawle, R. E. Person, S. Mahadevan et al., Paternally inherited microdeletion at 15q11.2 confirms a significant role for the SNORD116 C/D box snoRNA cluster in Prader???Willi syndrome, European Journal of Human Genetics, vol.18, issue.11, 2010.
DOI : 10.1038/ejhg.2010.102

S. Kishore and S. Stamm, The snoRNA HBII-52 Regulates Alternative Splicing of the Serotonin Receptor 2C, Science, vol.311, issue.5758, pp.230-232, 2006.
DOI : 10.1126/science.1118265

J. Muller, C. Stoetzel, M. C. Vincent, C. C. Leitch, V. Laurier et al., Identification of 28 novel mutations in the Bardet???Biedl syndrome genes: the burden of private mutations in an extensively heterogeneous disease, Human Genetics, vol.101, issue.1, pp.583-93, 2010.
DOI : 10.1007/s00439-010-0804-9

URL : https://hal.archives-ouvertes.fr/inserm-00462147

K. Mykytyn and V. C. Sheffield, Establishing a connection between cilia and Bardet???Biedl Syndrome, Trends in Molecular Medicine, vol.10, issue.3, pp.106-115, 2004.
DOI : 10.1016/j.molmed.2004.01.003

J. R. Davenport, A. J. Watts, V. C. Roper, M. J. Croyle, T. Van-groen et al., Disruption of Intraflagellar Transport in Adult Mice Leads to Obesity and Slow-Onset Cystic Kidney Disease, Current Biology, vol.17, issue.18, pp.1586-94, 2007.
DOI : 10.1016/j.cub.2007.08.034

S. Seo, D. F. Guo, K. Bugge, D. A. Morgan, K. Rahmouni et al., Requirement of Bardet-Biedl syndrome proteins for leptin receptor signaling, Human Molecular Genetics, vol.18, issue.7, pp.1323-1354, 2009.
DOI : 10.1093/hmg/ddp031

J. D. Marshall, R. T. Bronson, G. B. Collin, A. D. Nordstrom, P. Maffei et al., New Alstr??m Syndrome Phenotypes Based on the Evaluation of 182 Cases, Archives of Internal Medicine, vol.165, issue.6, pp.675-83, 2005.
DOI : 10.1001/archinte.165.6.675

W. Seifert, M. Holder-espinasse, S. Spranger, M. Hoeltzenbein, E. Rossier et al., Mutational spectrum of COH1 and clinical heterogeneity in Cohen syndrome, Journal of Medical Genetics, vol.43, issue.5, p.22, 2006.
DOI : 10.1136/jmg.2005.039867

A. M. Spiegel, Albright's Hereditary Osteodystrophy and Defective G Proteins, New England Journal of Medicine, vol.322, issue.20, pp.1461-1463, 1990.
DOI : 10.1056/NEJM199005173222010

L. S. Weinstein, P. V. Gejman, E. Friedman, T. Kadowaki, R. M. Collins et al., Mutations of the Gs alpha-subunit gene in Albright hereditary osteodystrophy detected by denaturing gradient gel electrophoresis., Proceedings of the National Academy of Sciences, vol.87, issue.21, pp.8287-90, 1990.
DOI : 10.1073/pnas.87.21.8287

L. S. Weinstein, T. Xie, A. Qasem, J. Wang, and M. Chen, The role of GNAS and other imprinted genes in the development of obesity, International Journal of Obesity, vol.22, issue.1, pp.6-17, 2010.
DOI : 10.1038/ijo.2009.222

T. Rankinen, A. Zuberi, Y. C. Chagnon, S. J. Weisnagel, G. Argyropoulos et al., The Human Obesity Gene Map: The 2005 Update, Obesity, vol.33, issue.4, pp.529-644, 2006.
DOI : 10.1038/oby.2006.71

S. Stone, V. Abkevich, S. C. Hunt, A. Gutin, D. L. Russell et al., A major predisposition locus for severe obesity, Am J Hum Genet, vol.70, pp.4-15, 2002.

M. Ohman, L. Oksanen, J. Kaprio, M. Koskenvuo, P. Mustajoki et al., Genome-Wide Scan of Obesity in Finnish Sibpairs Reveals Linkage to Chromosome Xq24, Journal of Clinical Endocrinology & Metabolism, vol.85, issue.9, pp.3183-90, 2000.
DOI : 10.1210/jc.85.9.3183

S. Stone, V. Abkevich, D. L. Russell, R. Riley, K. Timms et al., TBC1D1 is a candidate for a severe obesity gene and evidence for a gene/gene interaction in obesity predisposition, Human Molecular Genetics, vol.15, issue.18, pp.2709-2729, 2006.
DOI : 10.1093/hmg/ddl204

E. Suviolahti, L. J. Oksanen, M. Ohman, R. M. Cantor, M. Ridderstrale et al., The SLC6A14 gene shows evidence of association with obesity, Journal of Clinical Investigation, vol.112, issue.11, pp.1762-72, 2003.
DOI : 10.1172/JCI200317491

D. Meyre, M. Farge, C. Lecoeur, C. Proenca, E. Durand et al., R125W coding variant in TBC1D1 confers risk for familial obesity and contributes to linkage on chromosome 4p14 in the French population, Human Molecular Genetics, vol.17, issue.12, pp.1798-802, 2008.
DOI : 10.1093/hmg/ddn070

A. Chadt, K. Leicht, A. Deshmukh, L. Q. Jiang, S. Scherneck et al., Tbc1d1 mutation in lean mouse strain confers leanness and protects from diet-induced obesity, Nature Genetics, vol.279, issue.11, pp.1354-1363, 2008.
DOI : 10.1038/ng.244

J. L. Sloan and S. Mager, Cloning and Functional Expression of a Human Na+and Cl--dependent Neutral and Cationic Amino Acid Transporter B0+, Journal of Biological Chemistry, vol.274, issue.34, pp.23740-23745, 1999.
DOI : 10.1074/jbc.274.34.23740

E. Durand, P. Boutin, D. Meyre, M. A. Charles, K. Clement et al., Polymorphisms in the Amino Acid Transporter Solute Carrier Family 6 (Neurotransmitter Transporter) Member 14 Gene Contribute to Polygenic Obesity in French Caucasians, Diabetes, vol.53, issue.9, pp.2483-2489, 2004.
DOI : 10.2337/diabetes.53.9.2483

URL : https://hal.archives-ouvertes.fr/hal-00094121

M. Benzinou, J. C. Chevre, K. J. Ward, C. Lecoeur, C. Dina et al., Endocannabinoid receptor 1 gene variations increase risk for obesity and modulate body mass index in European populations, Human Molecular Genetics, vol.17, issue.13, pp.1916-1937, 2008.
DOI : 10.1093/hmg/ddn089

D. Meyre, N. Bouatia-naji, A. Tounian, C. Samson, C. Lecoeur et al., Variants of ENPP1 are associated with childhood and adult obesity and increase the risk of glucose intolerance and type 2 diabetes, Nature Genetics, vol.68, issue.8, pp.863-870, 2005.
DOI : 10.1038/ng1604

URL : https://hal.archives-ouvertes.fr/hal-00174474

P. Gazzerro, M. G. Caruso, M. Notarnicola, G. Misciagna, V. Guerra et al., Association between cannabinoid type-1 receptor polymorphism and body mass index in a southern Italian population, International Journal of Obesity, vol.19, issue.6, pp.908-920, 2007.
DOI : 10.1038/sj.ijo.0803510

J. P. Jaeger, V. S. Mattevi, S. M. Callegari-jacques, and M. H. Hutz, Cannabinoid Type-1 Receptor Gene Polymorphisms Are Associated with Central Obesity in a Southern Brazilian Population, Disease Markers, vol.25, issue.1, pp.67-74, 2008.
DOI : 10.1155/2008/841490

A. Peeters, S. Beckers, I. Mertens, W. Van-hul, V. Gaal et al., The G1422A variant of the cannabinoid receptor gene (CNR1) is associated with abdominal adiposity in obese men, Endocrine, vol.49, issue.Suppl 4, pp.138-179, 2007.
DOI : 10.1007/s12020-007-0022-y

P. Russo, P. Strazzullo, F. P. Cappuccio, D. A. Tregouet, F. Lauria et al., ) Are Associated with Obesity Phenotypes in Men, The Journal of Clinical Endocrinology & Metabolism, vol.92, issue.6, pp.2382-2388, 2007.
DOI : 10.1210/jc.2006-2523

Y. Bottcher, A. Korner, T. Reinehr, B. Enigk, W. Kiess et al., Variants and Haplotypes Predispose to Early Onset Obesity and Impaired Glucose and Insulin Metabolism in German Obese Children, The Journal of Clinical Endocrinology & Metabolism, vol.91, issue.12, pp.4948-52, 2006.
DOI : 10.1210/jc.2006-0540

E. Achhab, Y. Meyre, D. Bouatia-naji, N. Berraho, M. Deweirder et al., Association of the ENPP1 K121Q polymorphism with type 2 diabetes and obesity in the Moroccan population, Diabetes & Metabolism, vol.35, issue.1, pp.37-42, 2009.
DOI : 10.1016/j.diabet.2008.06.005

C. P. Jenkinson, D. K. Coletta, M. Flechtner-mors, S. L. Hu, M. J. Fourcaudot et al., With Obesity-related Phenotypes, Obesity, vol.28, issue.Suppl 1, pp.1708-1721, 2008.
DOI : 10.1038/oby.2008.262

A. Peeters, S. Beckers, A. Verrijken, I. Mertens, L. Van-gaal et al., Possible role for ENPP1 polymorphism in obesity but not for INSIG2 and PLIN variants, Endocrine, vol.57, issue.Suppl 1, pp.103-112, 2009.
DOI : 10.1007/s12020-009-9194-y

K. Valli-jaakola, E. Suviolahti, C. Schalin-jantti, S. Ripatti, K. Silander et al., in Obesity: Association With Morbid Obesity in Finns, Obesity, vol.20, issue.9, pp.2113-2122, 2008.
DOI : 10.1038/oby.2008.313

F. Stutzmann, V. Vatin, S. Cauchi, A. Morandi, B. Jouret et al., Non-synonymous polymorphisms in melanocortin-4 receptor protect against obesity: the two facets of a Janus obesity gene, Human Molecular Genetics, vol.16, issue.15, pp.1837-1881, 2007.
DOI : 10.1093/hmg/ddm132

URL : https://hal.archives-ouvertes.fr/hal-00172976

M. Traurig, J. Mack, R. L. Hanson, M. Ghoussaini, D. Meyre et al., Common Variation in SIM1 Is Reproducibly Associated With BMI in Pi ma Indians, Diabetes, vol.58, issue.7, pp.1682-1691, 2009.
DOI : 10.2337/db09-0028

A. Hinney, T. T. Nguyen, A. Scherag, S. Friedel, G. Bronner et al., Genome Wide Association (GWA) Study for Early Onset Extreme Obesity Supports the Role of Fat Mass and Obesity Associated Gene (FTO) Variants, PLoS ONE, vol.43, issue.3, p.1361, 2007.
DOI : 10.1371/journal.pone.0001361.s003

D. Meyre, J. Delplanque, J. C. Chevre, C. Lecoeur, S. Lobbens et al., Genome-wide association study for early-onset and morbid adult obesity identifies three new risk loci in European populations, Nature Genetics, vol.276, issue.2, pp.157-166, 2009.
DOI : 10.1016/S0006-291X(02)00379-0

A. Scherag, C. Dina, A. Hinney, V. Vatin, S. Scherag et al., Two New Loci for Body-Weight Regulation Identified in a Joint Analysis of Genome-Wide Association Studies for Early-Onset Extreme Obesity in French and German Study Groups, PLoS Genetics, vol.19, issue.4, p.1000916, 2010.
DOI : 10.1371/journal.pgen.1000916.s012

C. J. Willer, E. K. Speliotes, R. J. Loos, S. Li, C. M. Lindgren et al., Six new loci associated with body mass index highlight a neuronal influence on body weight regulation, Nature Genetics, vol.294, issue.1, pp.25-34, 2009.
DOI : 10.1086/521580

D. F. Conrad, D. Pinto, R. Redon, L. Feuk, O. Gokcumen et al., Origins and functional impact of copy number variation in the human genome, Nature, vol.36, issue.7289, pp.704-716, 2010.
DOI : 10.1038/nature08516

J. Zhao, J. P. Bradfield, M. Li, K. Wang, H. Zhang et al., The Role of Obesity-associated Loci Identified in Genome-wide Association Studies in the Determination of Pediatric BMI, Obesity, vol.17, issue.12, pp.2254-2261, 2009.
DOI : 10.1086/519795

E. G. Bochukova, N. Huang, J. Keogh, E. Henning, C. Purmann et al., Large, rare chromosomal deletions associated with severe early-onset obesity, Nature, vol.40, issue.7281, pp.666-70, 2010.
DOI : 10.1038/nature08689

R. G. Walters, S. Jacquemont, A. Valsesia, A. J. De-smith, D. Martinet et al., A new highly penetrant form of obesity due to deletions on chromosome 16p11.2, Nature, vol.30, issue.7281, pp.671-676, 2010.
DOI : 10.1038/nature08727

D. F. Steiner, D. Cunningham, L. Spigelman, and B. Aten, Insulin Biosynthesis: Evidence for a Precursor, Science, vol.157, issue.3789, pp.697-700, 1967.
DOI : 10.1126/science.157.3789.697

M. Chretien and C. H. Li, ISOLATION, PURIFICATION, AND CHARACTERIZATION OF ??-LIPOTROPIC HORMONE FROM SHEEP PITUITARY GLANDS, Canadian Journal of Biochemistry, vol.45, issue.7, pp.1163-74, 1967.
DOI : 10.1139/o67-133

N. G. Seidah and A. Prat, Precursor convertases in the secretory pathway, cytosol and extracellular milieu, Essays In Biochemistry, vol.38, pp.79-94, 2002.
DOI : 10.1042/bse0380079

J. W. Creemers and A. M. Khatib, Knock-out mouse models of proprotein convertases: unique functions or redundancy?, Frontiers in Bioscience, vol.Volume, issue.13, pp.4960-71, 2008.
DOI : 10.2741/3055

G. Thomas, Furin at the cutting edge: From protein traffic to embryogenesis and disease, Nature Reviews Molecular Cell Biology, vol.2, issue.10, pp.753-66, 2002.
DOI : 10.1038/nrm934

S. Benjannet, D. Rhainds, R. Essalmani, J. Mayne, L. Wickham et al., NARC-1/PCSK9 and Its Natural Mutants: ZYMOGEN CLEAVAGE AND EFFECTS ON THE LOW DENSITY LIPOPROTEIN (LDL) RECEPTOR AND LDL CHOLESTEROL, Journal of Biological Chemistry, vol.279, issue.47, pp.48865-75, 2004.
DOI : 10.1074/jbc.M409699200

A. Zhou, S. Martin, G. Lipkind, J. Lamendola, and D. F. Steiner, Regulatory Roles of the P Domain of the Subtilisin-like Prohormone Convertases, Journal of Biological Chemistry, vol.273, issue.18, pp.11107-11121, 1998.
DOI : 10.1074/jbc.273.18.11107

J. W. Creemers, M. Vey, W. Schafer, T. A. Ayoubi, A. J. Roebroek et al., Endoproteolytic Cleavage of Its Propeptide Is a Prerequisite for Efficient Transport of Furin Out of the Endoplasmic Reticulum, Journal of Biological Chemistry, vol.270, issue.6, pp.2695-702, 1995.
DOI : 10.1074/jbc.270.6.2695

A. Zhou, L. Paquet, and R. E. Mains, Structural Elements That Direct Specific Processing of Different Mammalian Subtilisin-like Prohormone Convertases, Journal of Biological Chemistry, vol.270, issue.37, pp.21509-21525, 1995.
DOI : 10.1074/jbc.270.37.21509

N. A. Taylor, W. J. Van-de-ven, and J. W. Creemers, Curbing activation: proprotein convertases in homeostasis and pathology, The FASEB Journal, vol.17, issue.10, pp.1215-1242, 2003.
DOI : 10.1096/fj.02-0831rev

F. Bergeron, R. Leduc, and R. Day, Subtilase-like pro-protein convertases: from molecular specificity to therapeutic applications, Journal of Molecular Endocrinology, vol.24, issue.1, pp.1-22, 2000.
DOI : 10.1677/jme.0.0240001

X. Zhu, Y. Cao, K. Voogd, and D. F. Steiner, On the Processing of Proghrelin to Ghrelin, Journal of Biological Chemistry, vol.281, issue.50, pp.38867-70, 2006.
DOI : 10.1074/jbc.M607955200

R. Ugleholdt, X. Zhu, C. F. Deacon, C. Orskov, D. F. Steiner et al., Impaired Intestinal Proglucagon Processing in Mice Lacking Prohormone Convertase 1, Endocrinology, vol.145, issue.3, pp.1349-55, 2004.
DOI : 10.1210/en.2003-0801

J. W. Creemers, L. E. Pritchard, A. Gyte, L. Rouzic, P. Meulemans et al., and Melanocortin Receptors Cannot Be Influenced by Syndecan-3, Endocrinology, vol.147, issue.4, pp.1621-1652, 2006.
DOI : 10.1210/en.2005-1373

J. Yoon and M. C. Beinfeld, Prohormone convertase 1 is necessary for the formation of cholecystokinin 8 in Rin5F and STC-1 cells, J Biol Chem, vol.272, pp.9450-9456, 1997.

R. Ugleholdt, M. L. Poulsen, P. J. Holst, J. C. Irminger, C. Orskov et al., Prohormone Convertase 1/3 Is Essential for Processing of the Glucose-dependent Insulinotropic Polypeptide Precursor, Journal of Biological Chemistry, vol.281, issue.16, pp.11050-11057, 2006.
DOI : 10.1074/jbc.M601203200

A. Dey, X. Xhu, R. Carroll, C. W. Turck, J. Stein et al., Biological Processing of the Cocaine and Amphetamine-regulated Transcript Precursors by Prohormone Convertases, PC2 and PC1/3, Journal of Biological Chemistry, vol.278, issue.17, pp.15007-15021, 2003.
DOI : 10.1074/jbc.M212128200

V. Hook, L. Funkelstein, T. Toneff, C. Mosier, and S. R. Hwang, Human pituitary contains dual cathepsin L and prohormone convertase processing pathway components involved in converting POMC into the peptide hormones ACTH, ??-MSH, and ??-endorphin, Endocrine, vol.89, issue.3, pp.429-466, 2009.
DOI : 10.1007/s12020-009-9163-5

J. H. Wardman, X. Zhang, S. Gagnon, L. M. Castro, X. Zhu et al., Analysis of peptides in prohormone convertase 1/3 null mouse brain using quantitative peptidomics, Journal of Neurochemistry, vol.99, pp.215-240, 2010.
DOI : 10.1111/j.1471-4159.2010.06760.x

E. Apletalina, J. Appel, N. S. Lamango, R. A. Houghten, and I. Lindberg, Identification of Inhibitors of Prohormone Convertases 1 and 2 Using a Peptide Combinatorial Library, Journal of Biological Chemistry, vol.273, issue.41, pp.26589-95, 1998.
DOI : 10.1074/jbc.273.41.26589

L. D. Fricker, A. A. Mckinzie, J. Sun, E. Curran, Y. Qian et al., Identification and characterization of proSAAS, a granin-like neuroendocrine peptide precursor that inhibits prohormone processing, J Neurosci, vol.20, pp.639-687, 2000.

D. J. Morgan, S. Wei, I. Gomes, T. Czyzyk, N. Mzhavia et al., The propeptide precursor proSAAS is involved in fetal neuropeptide processing and body weight regulation, Journal of Neurochemistry, vol.99, pp.1275-84, 2010.
DOI : 10.1111/j.1471-4159.2010.06706.x

D. L. Fox and D. J. Good, Nescient Helix-Loop-Helix 2 Interacts with Signal Transducer and Activator of Transcription 3 to Regulate Transcription of Prohormone Convertase 1/3, Molecular Endocrinology, vol.22, issue.6, pp.1438-1486, 2008.
DOI : 10.1210/me.2008-0010

J. H. Wen, Y. Y. Chen, S. J. Song, J. Ding, Y. Gao et al., Paired box 6 (PAX6) regulates glucose metabolism via proinsulin processing mediated by prohormone convertase 1/3 (PC1/3), Diabetologia, vol.89, issue.Suppl 1, pp.504-517, 2009.
DOI : 10.1007/s00125-008-1210-x

Y. Berman, N. Mzhavia, A. Polonskaia, and L. A. Devi, Impaired Prohormone Convertases inCpe fat/Cpe fat Mice, Journal of Biological Chemistry, vol.276, issue.2, pp.1466-73, 2001.
DOI : 10.1074/jbc.M008499200

M. Chretien, N. G. Seidah, A. Basak, and M. Mbikay, Proprotein convertases as therapeutic targets, Expert Opinion on Therapeutic Targets, vol.48, issue.10, pp.1289-300, 2008.
DOI : 10.1128/IAI.72.1.602-605.2004

R. Essalmani, J. Hamelin, J. Marcinkiewicz, A. Chamberland, M. Mbikay et al., Deletion of the Gene Encoding Proprotein Convertase 5/6 Causes Early Embryonic Lethality in the Mouse, Molecular and Cellular Biology, vol.26, issue.1, pp.354-61, 2006.
DOI : 10.1128/MCB.26.1.354-361.2006

N. G. Seidah, M. Chretien, and R. Day, The family of subtilisin/kexin like pro-protein and pro-hormone convertases: Divergent or shared functions, Biochimie, vol.76, issue.3-4, pp.197-209, 1994.
DOI : 10.1016/0300-9084(94)90147-3

N. G. Seidah, S. J. Mowla, J. Hamelin, A. M. Mamarbachi, S. Benjannet et al., Mammalian subtilisin/kexin isozyme SKI-1: A widely expressed proprotein convertase with a unique cleavage specificity and cellular localization, Proceedings of the National Academy of Sciences, vol.96, issue.4, pp.1321-1327, 1999.
DOI : 10.1073/pnas.96.4.1321

URL : https://hal.archives-ouvertes.fr/hal-00126413

C. Freyer, L. M. Kilpatrick, L. A. Salamonsen, and G. Nie, Pro-protein convertases (PCs) other than PC6 are not tightly regulated for implantation in the human endometrium, Reproduction, vol.133, issue.6, pp.1189-97, 2007.
DOI : 10.1530/REP-06-0285

J. Gagnon, J. Mayne, M. Mbikay, J. Woulfe, and M. Chretien, Expression of PCSK1 (PC1/3), PCSK2 (PC2) and PCSK3 (furin) in mouse small intestine, PCSK2 (PC2) and PCSK3 (furin) in mouse small intestine, pp.54-60, 2009.
DOI : 10.1016/j.regpep.2008.07.006

D. Szumska, G. Pieles, R. Essalmani, M. Bilski, D. Mesnard et al., VACTERL/caudal regression/Currarino syndrome-like malformations in mice with mutation in the proprotein convertase Pcsk5, Genes & Development, vol.22, issue.11, pp.1465-77, 2008.
DOI : 10.1101/gad.479408

M. S. Sarac, A. Cameron, and I. Lindberg, The Furin Inhibitor Hexa-D-Arginine Blocks the Activation of Pseudomonas aeruginosa Exotoxin A In Vivo, Infection and Immunity, vol.70, issue.12, pp.7136-7145, 2002.
DOI : 10.1128/IAI.70.12.7136-7139.2002

K. V. Kibler, A. Miyazato, V. S. Yedavalli, A. I. Dayton, B. L. Jacobs et al., Polyarginine Inhibits gp160 Processing by Furin and Suppresses Productive Human Immunodeficiency Virus Type 1 Infection, Journal of Biological Chemistry, vol.279, issue.47, pp.49055-63, 2004.
DOI : 10.1074/jbc.M403394200

G. Bolt and I. R. Pedersen, The Role of Subtilisin-like Proprotein Convertases for Cleavage of the Measles Virus Fusion Glycoprotein in Different Cell Types, Virology, vol.252, issue.2, pp.387-98, 1998.
DOI : 10.1006/viro.1998.9464

P. Konig, K. Giesow, K. Schuldt, U. J. Buchholz, and G. M. Keil, A novel protein expression strategy using recombinant bovine respiratory syncytial virus (BRSV): modifications of the peptide sequence between the two furin cleavage sites of the BRSV fusion protein yield secreted proteins, but affect processing and function of the BRSV fusion protein, Journal of General Virology, vol.85, issue.7, pp.1815-1839, 2004.
DOI : 10.1099/vir.0.80010-0

G. Neumann, H. Feldmann, S. Watanabe, I. Lukashevich, and Y. Kawaoka, Reverse Genetics Demonstrates that Proteolytic Processing of the Ebola Virus Glycoprotein Is Not Essential for Replication in Cell Culture, Journal of Virology, vol.76, issue.1, pp.406-416, 2002.
DOI : 10.1128/JVI.76.1.406-410.2002

V. E. Volchkov, H. Feldmann, V. A. Volchkova, and H. D. Klenk, Processing of the Ebola virus glycoprotein by the proprotein convertase furin, Proceedings of the National Academy of Sciences, vol.95, issue.10, pp.5762-5769, 1998.
DOI : 10.1073/pnas.95.10.5762

D. E. Bassi, R. Lopez-de-cicco, J. Cenna, S. Litwin, E. Cukierman et al., PACE4 Expression in Mouse Basal Keratinocytes Results in Basement Membrane Disruption and Acceleration of Tumor Progression, Cancer Research, vol.65, issue.16, pp.7310-7319, 2005.
DOI : 10.1158/0008-5472.CAN-05-1213

A. M. Khatib, G. Siegfried, M. Chretien, P. Metrakos, and N. G. Seidah, Proprotein Convertases in Tumor Progression and Malignancy, The American Journal of Pathology, vol.160, issue.6, 2002.
DOI : 10.1016/S0002-9440(10)61140-6

G. Siegfried, A. Basak, J. A. Cromlish, S. Benjannet, J. Marcinkiewicz et al., The secretory proprotein convertases furin, PC5, and PC7 activate VEGF-C to induce tumorigenesis, Journal of Clinical Investigation, vol.111, issue.11, pp.1723-1755, 2003.
DOI : 10.1172/JCI200317220

D. E. Bassi, H. Mahloogi, and A. J. Klein-szanto, The Proprotein Convertases Furin and PACE4 Play a Significant Role in Tumor Progression, Molecular Carcinogenesis, vol.28, issue.2, pp.63-72, 2000.
DOI : 10.1002/1098-2744(200006)28:2<63::AID-MC1>3.3.CO;2-3

M. Cheng, P. H. Watson, J. A. Paterson, N. Seidah, M. Chretien et al., Pro-protein convertase gene expression in human breast cancer, International Journal of Cancer, vol.267, issue.6, pp.966-71, 1997.
DOI : 10.1002/(SICI)1097-0215(19970611)71:6<966::AID-IJC10>3.0.CO;2-4

F. C. Hubbard, T. L. Goodrow, S. C. Liu, M. H. Brilliant, P. Basset et al., Expression of PACE4 in chemically induced carcinomas is associated with spindle cell tumor conversion and increased invasive ability, Cancer Res, vol.57, pp.5226-5257, 1997.

D. Vos, L. Declercq, J. Rosas, G. G. Van-damme, B. Roebroek et al., MMTV-cre-mediated fur inactivation concomitant with PLAG1 proto-oncogene activation delays salivary gland tumorigenesis in mice, Int J Oncol, vol.32, pp.1073-83, 2008.

X. Sun, R. Essalmani, N. G. Seidah, and A. Prat, The proprotein convertase PC5/6 is protective against intestinal tumorigenesis: in vivo mouse model, Molecular Cancer, vol.8, issue.1, p.73, 2009.
DOI : 10.1186/1476-4598-8-73

J. W. Creemers, A. J. Roebroek, and W. J. Van-de-ven, Expression in human lung tumor cells of the proprotein processing enzyme PC1/PC3 Cloning and primary sequence of a 5 kb cDNA, FEBS Letters, vol.81, issue.1, pp.82-90, 1992.
DOI : 10.1016/0014-5793(92)80169-H

J. A. Schalken, A. J. Roebroek, P. P. Oomen, S. S. Wagenaar, F. M. Debruyne et al., fur gene expression as a discriminating marker for small cell and nonsmall cell lung carcinomas., Journal of Clinical Investigation, vol.80, issue.6, pp.1545-1554, 1987.
DOI : 10.1172/JCI113240

A. C. Moss, G. M. Jacobson, L. E. Walker, N. W. Blake, E. Marshall et al., SCG3 Transcript in Peripheral Blood Is a Prognostic Biomarker for REST-Deficient Small Cell Lung Cancer, Clinical Cancer Research, vol.15, issue.1, pp.274-83, 2009.
DOI : 10.1158/1078-0432.CCR-08-1163

L. H. Stockwin, D. T. Vistica, S. Kenney, D. S. Schrump, D. O. Butcher et al., Gene expression profiling of alveolar soft-part sarcoma (ASPS), BMC Cancer, vol.25, issue.9, 2009.
DOI : 10.1097/00000478-200109000-00005

A. Blanchard, B. Iwasiow, A. Yarmill, A. Fresnosa, J. Silha et al., Targeted production of proprotein convertase PC1 enhances mammary development and tumorigenesis in transgenic miceThis article is one of a selection of papers published in a special issue celebrating the 125th anniversary of the Faculty of Medicine at the University of Manitoba., Canadian Journal of Physiology and Pharmacology, vol.87, issue.10, pp.831-839, 2009.
DOI : 10.1139/Y09-073

S. Ohta, S. Nishizawa, Y. Oki, T. Yokoyama, and H. Namba, Significance of absent prohormone convertase 1/3 in inducing clinically silent corticotroph pituitary adenoma of subtype I--immunohistochemical study, Pituitary, vol.5, issue.4, pp.221-224, 2002.
DOI : 10.1023/A:1025321731790

T. Tateno, H. Izumiyama, M. Doi, T. Akashi, K. Ohno et al., Defective Expression of Prohormone Convertase 1/3 in Silent Corticotroph Adenoma, Endocrine Journal, vol.54, issue.5, pp.777-82, 2007.
DOI : 10.1507/endocrj.K07-059

K. I. Tatsumi, S. Tanaka, T. Takano, S. Tahara, Y. Murakami et al., Frequent Appearance of Autoantibodies Against Prohormone Convertase 1/3 and Neuroendocrine Protein 7B2 in Patients with Nonfunctioning Pituitary Macroadenoma, Endocrine, vol.22, issue.3, pp.335-375, 2003.
DOI : 10.1385/ENDO:22:3:335

B. D. Bennett, P. Denis, M. Haniu, D. B. Teplow, S. Kahn et al., A Furin-like Convertase Mediates Propeptide Cleavage of BACE, the Alzheimer's beta -Secretase, Journal of Biological Chemistry, vol.275, issue.48, pp.37712-37719, 2000.
DOI : 10.1074/jbc.M005339200

I. Pinnix, J. E. Council, B. Roseberry, L. Onstead, W. Mallender et al., Convertases other than furin cleave beta-secretase to its mature form, Faseb J, vol.15, pp.1810-1812, 2001.

E. Lopez-perez, N. G. Seidah, and F. Checler, Proprotein convertase activity contributes to the processing of the Alzheimer's beta-amyloid precursor protein in human cells: evidence for a role of the prohormone convertase PC7 in the constitutive alphasecretase pathway, J Neurochem, vol.73, pp.2056-62, 1999.

S. H. Kim, J. W. Creemers, S. Chu, G. Thinakaran, and S. S. Sisodia, Proteolytic Processing of Familial British Dementia-associated BRI Variants, Journal of Biological Chemistry, vol.277, issue.3, pp.1872-1879, 2002.
DOI : 10.1074/jbc.M108739200

S. H. Kim, R. Wang, D. J. Gordon, J. Bass, D. F. Steiner et al., Furin mediates enhanced production of fibrillogenic ABri peptides in familial British dementia, Nat Neurosci, vol.2, pp.984-992, 1999.

K. N. Maxwell, E. A. Fisher, and J. L. Breslow, Overexpression of PCSK9 accelerates the degradation of the LDLR in a post-endoplasmic reticulum compartment, Proceedings of the National Academy of Sciences, vol.102, issue.6, pp.2069-74, 2005.
DOI : 10.1073/pnas.0409736102

M. Abifadel, M. Varret, J. P. Rabes, D. Allard, K. Ouguerram et al., Mutations in PCSK9 cause autosomal dominant hypercholesterolemia, Nature Genetics, vol.34, issue.2, pp.154-160, 2003.
DOI : 10.1038/ng1161

J. Cohen, A. Pertsemlidis, I. K. Kotowski, R. Graham, C. K. Garcia et al., Low LDL cholesterol in individuals of African descent resulting from frequent nonsense mutations in PCSK9, Nature Genetics, vol.41, issue.2, pp.161-166, 2005.
DOI : 10.1086/344207

I. K. Kotowski, A. Pertsemlidis, A. Luke, R. S. Cooper, G. L. Vega et al., A Spectrum of PCSK9 Alleles Contributes to Plasma Levels of Low-Density Lipoprotein Cholesterol, The American Journal of Human Genetics, vol.78, issue.3, pp.410-432, 2006.
DOI : 10.1086/500615

T. P. Leren, Mutations in the PCSK9 gene in Norwegian subjects with autosomal dominant hypercholesterolemia, Clinical Genetics, vol.277, issue.5, pp.419-441, 2004.
DOI : 10.1111/j.0009-9163.2004.0238.x

K. M. Timms, S. Wagner, M. E. Samuels, K. Forbey, H. Goldfine et al., A mutation in PCSK9 causing autosomal-dominant hypercholesterolemia in a Utah pedigree, Human Genetics, vol.114, issue.4, pp.349-53, 2004.
DOI : 10.1007/s00439-003-1071-9

S. G. Lakoski, T. A. Lagace, J. C. Cohen, J. D. Horton, and H. H. Hobbs, Genetic and Metabolic Determinants of Plasma PCSK9 Levels, The Journal of Clinical Endocrinology & Metabolism, vol.94, issue.7, pp.2537-2580, 2009.
DOI : 10.1210/jc.2009-0141

M. R. Law, N. J. Wald, and A. R. Rudnicka, Quantifying effect of statins on low density lipoprotein cholesterol, ischaemic heart disease, and stroke: systematic review and meta-analysis, BMJ, vol.326, issue.7404, p.1423, 2003.
DOI : 10.1136/bmj.326.7404.1423

J. Stamler, M. L. Daviglus, D. B. Garside, A. R. Dyer, P. Greenland et al., Relationship of Baseline Serum Cholesterol Levels in 3 Large Cohorts of Younger Men to Long-term Coronary, Cardiovascular, and All-Cause Mortality and to Longevity, JAMA, vol.284, issue.3, pp.311-319, 2000.
DOI : 10.1001/jama.284.3.311

J. C. Cohen, E. Boerwinkle, T. H. Mosley, . Jr, and H. H. Hobbs, Low LDL, and Protection against Coronary Heart Disease, New England Journal of Medicine, vol.354, issue.12, pp.1264-72, 2006.
DOI : 10.1056/NEJMoa054013

J. Sakai, R. B. Rawson, P. J. Espenshade, D. Cheng, A. C. Seegmiller et al., Molecular Identification of the Sterol-Regulated Luminal Protease that Cleaves SREBPs and Controls Lipid Composition of Animal Cells, Molecular Cell, vol.2, issue.4, pp.505-519, 1998.
DOI : 10.1016/S1097-2765(00)80150-1

W. Jin, I. V. Fuki, N. G. Seidah, S. Benjannet, J. M. Glick et al., Proprotein Covertases Are Responsible for Proteolysis and Inactivation of Endothelial Lipase, Journal of Biological Chemistry, vol.280, issue.44, pp.36551-36560, 2005.
DOI : 10.1074/jbc.M502264200

N. Scamuffa, F. Calvo, M. Chretien, N. G. Seidah, and A. M. Khatib, Proprotein convertases: lessons from knockouts, The FASEB Journal, vol.20, issue.12, pp.1954-63, 2006.
DOI : 10.1096/fj.05-5491rev

A. J. Roebroek, L. Umans, I. G. Pauli, E. J. Robertson, F. Van-leuven et al., Failure of ventral closure and axial rotation in embryos lacking the proprotein convertase Furin, Development, vol.125, pp.4863-76, 1998.

M. Furuta, H. Yano, A. Zhou, Y. Rouille, J. J. Holst et al., Defective prohormone processing and altered pancreatic islet morphology in mice lacking active SPC2, Proceedings of the National Academy of Sciences, vol.94, issue.13, pp.6646-51, 1997.
DOI : 10.1073/pnas.94.13.6646

X. Zhu, A. Zhou, A. Dey, C. Norrbom, R. Carroll et al., Disruption of PC1/3 expression in mice causes dwarfism and multiple neuroendocrine peptide processing defects, Proceedings of the National Academy of Sciences, vol.99, issue.16, pp.10293-10301, 2002.
DOI : 10.1073/pnas.162352599

M. Mbikay, H. Tadros, N. Ishida, C. P. Lerner, E. De-lamirande et al., Impaired fertility in mice deficient for the testicular germ-cell protease PC4, Proceedings of the National Academy of Sciences, vol.94, issue.13, pp.6842-6848, 1997.
DOI : 10.1073/pnas.94.13.6842

D. B. Constam and E. J. Robertson, SPC4/PACE4 regulates a TGFbeta signaling network during axis formation, Genes Dev, vol.14, pp.1146-55, 2000.

B. D. Gaylinn, V. I. Dealmeida, C. E. Lyons, . Jr, K. C. Wu et al., The Mutant Growth Hormone-Releasing Hormone (GHRH) Receptor of the Little Mouse Does Not Bind GHRH, Endocrinology, vol.140, issue.11, pp.5066-74, 1999.
DOI : 10.1210/en.140.11.5066

D. J. Lloyd, S. Bohan, and N. Gekakis, Obesity, hyperphagia and increased metabolic efficiency in Pc1 mutant mice, Human Molecular Genetics, vol.15, issue.11, pp.1884-93, 2006.
DOI : 10.1093/hmg/ddl111

M. Mbikay, G. Croissandeau, F. Sirois, Y. Anini, J. Mayne et al., A targeted deletion/insertion in the mouse Pcsk1 locus is associated with homozygous embryo preimplantation lethality, mutant allele preferential transmission and heterozygous female susceptibility to dietary fat, Developmental Biology, vol.306, issue.2, pp.584-98, 2007.
DOI : 10.1016/j.ydbio.2007.03.523

O. 'rahilly, S. Gray, H. Humphreys, P. J. Krook, A. Polonsky et al., Impaired Processing of Prohormones Associated with Abnormalities of Glucose Homeostasis and Adrenal Function, New England Journal of Medicine, vol.333, issue.21, pp.1386-90, 1995.
DOI : 10.1056/NEJM199511233332104

O. 'rahilly, S. Farooqi, and I. S. , Genetics of obesity, Philosophical Transactions of the Royal Society B: Biological Sciences, vol.372, issue.6505, pp.1095-105, 2006.
DOI : 10.1038/372425a0

C. Dina, D. Meyre, S. Gallina, E. Durand, A. Korner et al., Variation in FTO contributes to childhood obesity and severe adult obesity, Nature Genetics, vol.165, issue.6, pp.724-730, 2007.
DOI : 10.1073/pnas.0400782101

URL : https://hal.archives-ouvertes.fr/hal-00173651

T. M. Frayling, N. J. Timpson, M. N. Weedon, E. Zeggini, R. M. Freathy et al., A Common Variant in the FTO Gene Is Associated with Body Mass Index and Predisposes to Childhood and Adult Obesity, Science, vol.316, issue.5826, pp.889-94, 2007.
DOI : 10.1126/science.1141634

C. G. Bell, M. Benzinou, A. Siddiq, C. Lecoeur, C. Dina et al., Genome-wide Linkage Analysis for Severe Obesity in French Caucasians Finds Significant Susceptibility Locus on Chromosome 19q, Diabetes, vol.53, issue.7, pp.1857-65, 2004.
DOI : 10.2337/diabetes.53.7.1857

URL : https://hal.archives-ouvertes.fr/hal-00174527

Y. C. Chagnon, T. Rice, L. Perusse, I. B. Borecki, M. A. Ho-kim et al., Genomic scan for genes affecting body composition before and after training in Caucasians from HERITAGE, J Appl Physiol, vol.90, pp.1777-87, 2001.

G. Chen, A. A. Adeyemo, T. Johnson, J. Zhou, A. Amoah et al., A genome-wide scan for quantitative trait loci linked to obesity phenotypes among West Africans, International Journal of Obesity, vol.55, issue.3, pp.255-264, 2005.
DOI : 10.1038/sj.ijo.0802873

J. Hager, C. Dina, S. Francke, S. Dubois, M. Houari et al., A genome-wide scan for human obesity genes reveals a major susceptibility locus on chromosome 10, Nat Genet, vol.20, pp.304-312, 1998.

M. Benzinou, J. W. Creemers, H. Choquet, S. Lobbens, C. Dina et al., Common nonsynonymous variants in PCSK1 confer risk of obesity, Nature Genetics, vol.269, issue.8, pp.943-948, 2008.
DOI : 10.1093/hmg/ddl111

T. O. Kilpelainen, S. A. Bingham, K. T. Khaw, N. J. Wareham, and R. J. Loos, Association of variants in the PCSK1 gene with obesity in the EPIC-Norfolk study, Human Molecular Genetics, vol.18, issue.18, pp.3496-501, 2009.
DOI : 10.1093/hmg/ddp280

C. H. Sandholt, T. Sparso, N. Grarup, A. Albrechtsen, K. Almind et al., Combined Analyses of 20 Common Obesity Susceptibility Variants, Diabetes, vol.59, issue.7, pp.1667-73, 2010.
DOI : 10.2337/db09-1042

Q. Qi, H. Li, R. J. Loos, C. Liu, F. B. Hu et al., Association of PCSK1 rs6234 with Obesity and Related Traits in a Chinese Han Population, PLoS ONE, vol.41, issue.5, p.10590, 2010.
DOI : 10.1371/journal.pone.0010590.t003

F. Renstrom, F. Payne, A. Nordstrom, E. C. Brito, O. Rolandsson et al., Replication and extension of genome-wide association study results for obesity in 4923 adults from northern Sweden, Human Molecular Genetics, vol.18, issue.8, pp.1489-96, 2009.
DOI : 10.1093/hmg/ddp041

M. Heni, A. Haupt, S. A. Schafer, C. Ketterer, C. Thamer et al., Association of obesity risk SNPs in PCSK1with insulin sensitivity and proinsulin conversion, BMC Medical Genetics, vol.46, issue.1, p.86, 2010.
DOI : 10.2337/diab.46.12.1990

G. H. Goossens, L. Petersen, E. E. Blaak, G. Hul, P. Arner et al., Several obesity- and nutrient-related gene polymorphisms but not FTO and UCP variants modulate postabsorptive resting energy expenditure and fat-induced thermogenesis in obese individuals: the NUGENOB Study, International Journal of Obesity, vol.17, issue.6, pp.669-79, 2009.
DOI : 10.1017/BJN20061901

E. Corpeleijn, L. Petersen, C. Holst, W. H. Saris, A. Astrup et al., Obesity-related Polymorphisms and Their Associations With the Ability to Regulate Fat Oxidation in Obese Europeans: The NUGENOB Study, Obesity, vol.9, issue.7, pp.1369-77, 2010.
DOI : 10.1017/BJN20061901

URL : https://hal.archives-ouvertes.fr/inserm-00505128

B. G. Challis, L. E. Pritchard, J. W. Creemers, J. Delplanque, J. M. Keogh et al., A missense mutation disrupting a dibasic prohormone processing site in pro-opiomelanocortin (POMC) increases susceptibility to early-onset obesity through a novel molecular mechanism, Human Molecular Genetics, vol.11, issue.17, 1997.
DOI : 10.1093/hmg/11.17.1997

URL : https://hal.archives-ouvertes.fr/hal-00174706

J. N. Hirschhorn, Genomewide Association Studies ??? Illuminating Biologic Pathways, New England Journal of Medicine, vol.360, issue.17, pp.1699-701, 2009.
DOI : 10.1056/NEJMp0808934

M. Baker, N. Gaukrodger, B. M. Mayosi, H. Imrie, M. Farrall et al., Association Between Common Polymorphisms of the Proopiomelanocortin Gene and Body Fat Distribution: A Family Study, Diabetes, vol.54, issue.8, pp.2492-2498, 2005.
DOI : 10.2337/diabetes.54.8.2492

B. S. Sutton, C. D. Langefeld, A. H. Williams, J. M. Norris, M. F. Saad et al., Association of Proopiomelanocortin Gene Polymorphisms with Obesity in the IRAS Family Study, Obesity Research, vol.25, issue.9, pp.1491-1499, 2005.
DOI : 10.1038/oby.2005.180

E. T. Cirulli and D. B. Goldstein, Uncovering the roles of rare variants in common disease through whole-genome sequencing, Nature Reviews Genetics, vol.39, issue.6, pp.415-440, 2010.
DOI : 10.1038/nrg2779

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

F. Stutzmann, K. Tan, V. Vatin, C. Dina, B. Jouret et al., Prevalence of Melanocortin-4 Receptor Deficiency in Europeans and Their Age-Dependent Penetrance in Multigenerational Pedigrees, Diabetes, vol.57, issue.9, pp.2511-2519, 2008.
DOI : 10.2337/db08-0153

R. Redon, S. Ishikawa, K. R. Fitch, L. Feuk, G. H. Perry et al., Global variation in copy number in the human genome, Nature, vol.38, issue.7118, pp.444-54, 2006.
DOI : 10.1126/science.1117196

L. J. Coin, J. E. Asher, R. G. Walters, J. S. Moustafa, A. J. De-smith et al., cnvHap: an integrative population and haplotype???based multiplatform model of SNPs and CNVs, Nature Methods, vol.8, issue.7, pp.541-547, 2010.
DOI : 10.1038/nmeth.1466

L. Ma, P. A. Tataranni, C. Bogardus, and L. J. Baier, Melanocortin 4 Receptor Gene Variation Is Associated With Severe Obesity in Pima Indians, Diabetes, vol.53, issue.10, pp.2696-2705, 2004.
DOI : 10.2337/diabetes.53.10.2696

Y. C. Chang, Y. F. Chiu, K. C. Shih, M. W. Lin, W. H. Sheu et al., Common PCSK1 Haplotypes Are Associated With Obesity in the Chinese Population, Obesity, vol.79, issue.7, pp.1404-1413, 2010.
DOI : 10.1038/ng.156

B. J. Hennig, A. J. Fulford, G. Sirugo, P. Rayco-solon, A. T. Hattersley et al., FTOgene variation and measures of body mass in an African population, BMC Medical Genetics, vol.73, issue.1, p.21, 2009.
DOI : 10.1136/adc.73.1.25

S. Chiu, K. Kim, K. A. Haus, G. M. Espinal, L. V. Millon et al., Identification of positional candidate genes for body weight and adiposity in subcongenic mice, Physiological Genomics, vol.31, issue.1, pp.75-85, 2007.
DOI : 10.1152/physiolgenomics.00267.2006

Y. Anini, J. Mayne, J. Gagnon, J. Sherbafi, A. Chen et al., Genetic deficiency for proprotein convertase subtilisin/kexin type 2 in mice is associated with decreased adiposity and protection from dietary fat-induced body weight gain, International Journal of Obesity, vol.275, issue.11, 2010.
DOI : 10.1210/en.2003-0829

M. E. Rac, K. Safranow, and W. Poncyljusz, Molecular basis of human CD36 gene mutations, Mol Med, vol.13, pp.288-96, 2007.

F. Lepretre, F. Vasseur, M. Vaxillaire, P. E. Scherer, S. Ali et al., A CD36 nonsense mutation associated with insulin resistance and familial type 2 diabetes, Human Mutation, vol.24, issue.1, p.104, 2004.
DOI : 10.1002/humu.9256

URL : https://hal.archives-ouvertes.fr/hal-00174525

F. Laugerette, P. Passilly-degrace, B. Patris, I. Niot, M. Febbraio et al., CD36 involvement in orosensory detection of dietary lipids, spontaneous fat preference, and digestive secretions, Journal of Clinical Investigation, vol.115, issue.11, pp.3177-84, 2005.
DOI : 10.1172/JCI25299

S. Bokor, V. Legry, A. Meirhaeghe, J. R. Ruiz, B. Mauro et al., Single-nucleotide Polymorphism of CD36 Locus and Obesity in European Adolescents, Obesity, vol.2008, issue.7, pp.1398-403, 2010.
DOI : 10.1016/0959-437X(95)80025-5

H. Choquet, Y. Labrune, F. De-graeve, A. Hinney, J. Hebebrand et al., Lack of Association of CD36 SNPs With Early Onset Obesity: A Meta-Analysis in 9,973 European Subjects, Obesity, vol.315, issue.4, 2010.
DOI : 10.1038/ng.301

A. Hirasawa, K. Tsumaya, T. Awaji, S. Katsuma, T. Adachi et al., Free fatty acids regulate gut incretin glucagon-like peptide-1 secretion through GPR120, Nature Medicine, vol.52, issue.1, pp.90-94, 2005.
DOI : 10.1074/jbc.M201624200

H. E. Parker, A. M. Habib, G. J. Rogers, F. M. Gribble, and F. Reimann, Nutrient-dependent secretion of glucose-dependent insulinotropic polypeptide from primary murine K cells, Diabetologia, vol.447, issue.Suppl 1, pp.289-98, 2009.
DOI : 10.1007/s00125-008-1202-x

C. Gotoh, Y. H. Hong, T. Iga, D. Hishikawa, Y. Suzuki et al., The regulation of adipogenesis through GPR120, Biochemical and Biophysical Research Communications, vol.354, issue.2, pp.591-598, 2007.
DOI : 10.1016/j.bbrc.2007.01.028

I. Pereiro, D. Valverde, T. Pineiro-gallego, M. Baiget, S. Borrego et al., New mutations in BBS genes in small consanguineous families with Bardet-Biedl syndrome: detection of candidate regions by homozygosity mapping, Mol Vis, vol.16, pp.137-180, 2010.

D. Filippo, C. Pini-prato, A. Mattioli, G. Avanzini, S. Rapuzzi et al., Genomics approach to the analysis of bacterial communities dynamics in Hirschsprung???s disease-associated enterocolitis: a pilot study, Pediatric Surgery International, vol.44, issue.1, pp.465-71, 2010.
DOI : 10.1007/s00383-010-2586-5

H. Choquet and D. Meyre, Genomic insights into early-onset obesity, Genome Medicine, vol.2, issue.6, p.36, 2010.
DOI : 10.1186/gm157

URL : http://doi.org/10.1186/gm157

Z. Wei, K. Wang, H. Q. Qu, H. Zhang, J. Bradfield et al., From Disease Association to Risk Assessment: An Optimistic View from Genome-Wide Association Studies on Type 1 Diabetes, PLoS Genetics, vol.38, issue.10, p.1000678, 2009.
DOI : 10.1371/journal.pgen.1000678.s006

O. 'rahilly, S. Farooqi, and I. S. , Human obesity as a heritable disorder of the central control of energy balance, International Journal of Obesity, vol.14, issue.7, pp.55-61, 2008.
DOI : 10.1016/j.cmet.2007.06.004

E. Sonestedt, C. Roos, B. Gullberg, U. Ericson, E. Wirfalt et al., Fat and carbohydrate intake modify the association between genetic variation in the FTO genotype and obesity, American Journal of Clinical Nutrition, vol.90, issue.5, pp.1418-1443, 2009.
DOI : 10.3945/ajcn.2009.27958

D. Corella, G. Peloso, D. K. Arnett, S. Demissie, L. A. Cupples et al., APOA2, Dietary Fat, and Body Mass Index, Archives of Internal Medicine, vol.169, issue.20, pp.1897-906, 2009.
DOI : 10.1001/archinternmed.2009.343

S. Cauchi, F. Stutzmann, C. Cavalcanti-proenca, E. Durand, A. Pouta et al., Combined effects of MC4R and FTO common genetic variants on obesity in European general populations, Journal of Molecular Medicine, vol.25, issue.5, pp.537-583, 2009.
DOI : 10.1007/s00109-009-0451-6

J. A. Jacobsson, U. Riserus, T. Axelsson, L. Lannfelt, H. B. Schioth et al., The common FTOvariant rs9939609 is not associated with BMI in a longitudinal study on a cohort of Swedish men born 1920-1924, BMC Medical Genetics, vol.458, issue.Suppl 3, p.131, 2009.
DOI : 10.1038/nature07848

S. Karasawa, M. Daimon, S. Sasaki, S. Toriyama, T. Oizumi et al., Association of the Common Fat Mass and Obesity Associated (FTO) Gene Polymorphism with Obesity in a Japanese Population, Endocrine Journal, vol.57, issue.4, 2010.
DOI : 10.1507/endocrj.K09E-305

J. A. Mitchell, T. S. Church, T. Rankinen, C. P. Earnest, X. Sui et al., FTO Genotype and the Weight Loss Benefits of Moderate Intensity Exercise, Obesity, vol.276, issue.3, pp.641-644, 2010.
DOI : 10.1371/journal.pone.0004428

E. Rampersaud, B. D. Mitchell, T. I. Pollin, M. Fu, H. Shen et al., Physical Activity and the Association of Common FTO Gene Variants With Body Mass Index and Obesity, Archives of Internal Medicine, vol.168, issue.16, pp.1791-1798, 2008.
DOI : 10.1001/archinte.168.16.1791

J. R. Ruiz, I. Labayen, F. B. Ortega, V. Legry, L. A. Moreno et al., Attenuation of the Effect of the FTO rs9939609 Polymorphism on Total and Central Body Fat by Physical Activity in Adolescents, Archives of Pediatrics & Adolescent Medicine, vol.164, issue.4, pp.328-361, 2010.
DOI : 10.1001/archpediatrics.2010.29

K. S. Vimaleswaran, S. Li, J. H. Zhao, J. Luan, S. A. Bingham et al., Physical activity attenuates the body mass index-increasing influence of genetic variation in the FTO gene, American Journal of Clinical Nutrition, vol.90, issue.2, pp.425-433, 2009.
DOI : 10.3945/ajcn.2009.27652

H. Hauner, M. Meier, K. H. Jockel, U. H. Frey, and W. Siffert, Prediction of successful weight reduction under sibutramine therapy through genotyping of the G-protein ??3 subunit gene (GNB3) C825T polymorphism, Pharmacogenetics, vol.13, issue.8, pp.453-462, 2003.
DOI : 10.1097/00008571-200308000-00003

D. J. Hsiao, L. S. Wu, S. Y. Huang, and E. Lin, Weight loss and body fat reduction under sibutramine therapy in obesity with the C825T polymorphism in the GNB3 gene, Pharmacogenetics and Genomics, vol.19, issue.9, pp.730-733, 2009.
DOI : 10.1097/FPC.0b013e3283307cf1

T. J. Hsiao, L. S. Wu, Y. Hwang, S. Y. Huang, and E. Lin, Effect of the Common -866G/A Polymorphism of the Uncoupling Protein 2 Gene on Weight Loss and Body Composition under Sibutramine Therapy in an Obese Taiwanese Population, Molecular Diagnosis & Therapy, vol.7, issue.1, pp.101-107, 2010.
DOI : 10.1007/BF03256359

H. H. Chen, W. J. Lee, W. Wang, M. T. Huang, Y. C. Lee et al., Ala55Val Polymorphism on UCP2 Gene Predicts Greater Weight Loss in Morbidly Obese Patients Undergoing Gastric Banding, Obesity Surgery, vol.55, issue.6B, pp.926-959, 2007.
DOI : 10.1007/s11695-007-9171-6

F. H. Luyckx, A. J. Scheen, A. M. Proenza, A. D. Strosberg, P. J. Lefebvre et al., Influence of the A???G (-3826) uncoupling protein-1 gene (UCP1) variant on the dynamics of body weight before and after gastroplasty in morbidly obese subjects, International Journal of Obesity, vol.22, issue.12, pp.1244-1249, 1998.
DOI : 10.1038/sj.ijo.0800775

C. Peraglie, Mini-Gastric Bypass in a Patient Homozygous for Factor V Leiden, Obesity Surgery, vol.38, issue.1, pp.104-111, 2007.
DOI : 10.1007/s11695-007-9014-5

R. Peterli, A. Donadini, T. Peters, C. Ackermann, and P. Tondelli, Re-operations Following Laparoscopic Adjustable Gastric Banding, Obesity Surgery, vol.12, issue.6, pp.851-857, 2002.
DOI : 10.1381/096089202320995691

C. Poitou, J. M. Lacorte, M. Coupaye, S. Bertrais, J. F. Bedel et al., Relationship between Single Nucleotide Polymorphisms in Leptin, IL6 and Adiponectin Genes and their Circulating Product in Morbidly Obese Subjects before and after Gastric Banding Surgery, Obesity Surgery, vol.15, issue.1, pp.11-23, 2005.
DOI : 10.1381/0960892052993431

G. Sesti, L. Perego, M. Cardellini, F. Andreozzi, C. Ricasoli et al., Impact of Common Polymorphisms in Candidate Genes for Insulin Resistance and Obesity on Weight Loss of Morbidly Obese Subjects after Laparoscopic Adjustable Gastric Banding and Hypocaloric Diet, The Journal of Clinical Endocrinology & Metabolism, vol.90, issue.9, pp.5064-5073, 2005.
DOI : 10.1210/jc.2005-0404

L. B. Daniels and A. S. Maisel, Natriuretic Peptides, Journal of the American College of Cardiology, vol.50, issue.25, pp.2357-68, 2007.
DOI : 10.1016/j.jacc.2007.09.021

A. Meirhaeghe, M. S. Sandhu, M. I. Mccarthy, P. De-groote, D. Cottel et al., Association between the T-381C polymorphism of the brain natriuretic peptide gene and risk of type 2 diabetes in human populations, Human Molecular Genetics, vol.16, issue.11, pp.1343-50, 2007.
DOI : 10.1093/hmg/ddm084

H. Choquet, C. Cavalcanti-proenca, C. Lecoeur, C. Dina, S. Cauchi et al., The T-381C SNP in BNP gene may be modestly associated with type 2 diabetes: an updated meta-analysis in 49 279 subjects, Human Molecular Genetics, vol.18, issue.13, pp.2495-501, 2009.
DOI : 10.1093/hmg/ddp169