T. Niccoli and L. Partridge, Ageing as a risk factor for disease, Curr Biol, vol.22, issue.17, pp.741-52, 2012.

K. Barnett, S. W. Mercer, M. Norbury, G. Watt, S. Wyke et al., Epidemiology of multimorbidity and implications for health care, research, and medical education: a cross-sectional study, Lancet, vol.380, issue.9836, pp.37-43, 2012.

M. Helfand, D. Buckley, C. Fleming, R. Fu, M. Freeman et al., Screening for intermediate risk factors for coronary heart disease, 2009.

D. Goldman and F. Ducci, Deconstruction of vulnerability to complex diseases: enhanced effect sizes and power of intermediate phenotypes, ScientificWorldJournal, vol.7, pp.124-154, 2007.

E. K. Silverman and L. J. Palmer, Case-control association studies for the genetics of complex respiratory diseases, Am J Respir Cell Mol Biol, vol.22, issue.6, pp.645-653, 2000.

T. F. Mackay, Q&A: Genetic analysis of quantitative traits, J Biol, vol.8, issue.3, p.23, 2009.

H. Jammes, C. Junien, and P. Chavatte-palmer, Epigenetic control of development and expression of quantitative traits, Reprod Fertil Dev, vol.23, issue.1, pp.64-74, 2011.
URL : https://hal.archives-ouvertes.fr/hal-01018986

K. Marinou, D. Tousoulis, A. S. Antonopoulos, E. Stefanadi, and C. Stefanadis, Obesity and cardiovascular disease: from pathophysiology to risk stratification, Int J Cardiol, vol.138, issue.1, pp.3-8, 2010.

J. E. Hokanson and M. A. Austin, Plasma triglyceride level is a risk factor for cardiovascular disease independent of high-density lipoprotein cholesterol level: a meta-analysis of population-based prospective studies, J Cardiovasc Risk, vol.3, issue.2, pp.213-222, 1996.

R. M. Califf, Biomarker definitions and their applications, Exp Biol Med (Maywood), vol.243, issue.3, pp.213-234, 2018.

K. Strimbu and J. A. Tavel, What are biomarkers?, Curr Opin HIV AIDS, vol.5, issue.6, pp.463-469, 2010.

A. Torkamani, E. J. Topol, and N. J. Schork, Pathway analysis of seven common diseases assessed by genome-wide association, Genomics, vol.92, issue.5, pp.265-72, 2008.

T. K. Wojdacz, Methylation biomarker development in the context of the EU regulations for clinical use of in-vitro diagnostic devices. Expert review of molecular diagnostics, vol.19, pp.439-480, 2019.

P. Cordero, J. Li, and J. A. Oben, Epigenetics of obesity: beyond the genome sequence, Curr Opin Clin Nutr Metab Care, vol.18, issue.4, pp.361-367, 2015.

C. Mitchell, L. M. Schneper, and D. A. Notterman, DNA methylation, early life environment, and health outcomes, Pediatr Res, vol.79, p.212, 2015.

R. Feil and M. F. Fraga, Epigenetics and the environment: emerging patterns and implications, Nat Rev Genet, vol.13, issue.2, pp.97-109, 2012.
URL : https://hal.archives-ouvertes.fr/hal-02193314

P. A. Jones, H. Ohtani, A. Chakravarthy, D. Carvalho, and D. D. , Epigenetic therapy in immuneoncology, Nature Reviews Cancer, vol.19, issue.3, pp.151-61, 2019.

M. Fardi, S. Solali, F. Hagh, and M. , Epigenetic mechanisms as a new approach in cancer treatment: An updated review, Genes & diseases, vol.5, issue.4, pp.304-315, 2018.

P. Durrington and . Dyslipidaemia, Lancet, vol.362, issue.9385, pp.717-748, 2003.

S. Sayols-baixeras, M. R. Irvin, D. K. Arnett, R. Elosua, and S. W. Aslibekyan, Epigenetics of Lipid Phenotypes, Curr Cardiovasc Risk Rep, vol.10, issue.10, 2016.

K. Lokk, V. Modhukur, B. Rajashekar, K. Märtens, R. Mägi et al., DNA methylome profiling of human tissues identifies global and tissue-specific methylation patterns, Genome Biol, vol.15, issue.4, p.54, 2014.

B. C. Christensen, E. A. Houseman, C. J. Marsit, S. Zheng, M. R. Wrensch et al., Aging and environmental exposures alter tissue-specific DNA methylation dependent upon CpG island context, PLoS Genet, vol.5, issue.8, p.1000602, 2009.

M. M. Mendelson, R. E. Marioni, R. Joehanes, C. Liu, Å. Hedman et al., Association of Body Mass Index with DNA Methylation and Gene Expression in Blood Cells and Relations to Cardiometabolic Disease: A Mendelian Randomization Approach, PLoS Med, vol.14, issue.1, p.1002215, 2017.

Y. Huang, S. Chu, E. B. Loucks, C. Lin, C. B. Eaton et al., Gong Y, Dou LJ, Liang J. Link between obesity and cancer: role of triglyceride/free fatty acid cycling, Eur Rev Med Pharmacol Sci, vol.11, issue.3, pp.2808-2828, 2014.

K. J. Dick, C. P. Nelson, L. Tsaprouni, J. K. Sandling, D. Aissi et al., DNA methylation and bodymass index: a genome-wide analysis, Lancet, vol.383, issue.9933, pp.1990-1998, 2014.

E. W. Demerath, W. Guan, M. L. Grove, S. Aslibekyan, M. Mendelson et al., Epigenomewide association study (EWAS) of BMI, BMI change and waist circumference in African American adults identifies multiple replicated loci, Hum Mol Genet, vol.24, issue.15, pp.4464-79, 2015.

T. Kouzarides, Histone methylation in transcriptional control, Curr Opin Genet Dev, vol.12, issue.2, pp.198-209, 2002.

. Abcam, Histone modifications, 2019.

M. Budoff, Triglycerides and Triglyceride-Rich Lipoproteins in the Causal Pathway of Cardiovascular Disease, Am J Cardiol, vol.118, issue.1, pp.138-183, 2016.

Z. Reiner, Hypertriglyceridaemia and risk of coronary artery disease, Nat Rev Cardiol, vol.14, issue.7, pp.401-412, 2017.

Y. Handelsman, M. D. Shapiro, A. Triglycerides, A. Cardiovascular, and . Studies, FOCUS ON OMEGA-3 FATTY ACIDS, vol.23, pp.100-112, 2017.

M. Miller, N. J. Stone, C. Ballantyne, V. Bittner, M. H. Criqui et al., Triglycerides and cardiovascular disease: a scientific statement from the, American Heart Association. Circulation, vol.123, issue.20, pp.2292-333, 2011.

. Who and . Obesity, , 2019.

J. Després and I. Lemieux, Abdominal obesity and metabolic syndrome, Nature, vol.444, issue.7121, p.881, 2006.

K. Dhana, K. Braun, J. Nano, T. Voortman, E. W. Demerath et al., An Epigenome-Wide Association Study (EWAS) of Obesity-Related Traits, Am J Epidemiol, 2018.

B. B. Kahn and J. S. Flier, Obesity and insulin resistance, J Clin Invest, vol.106, issue.4, pp.473-81, 2000.

S. Wahl, A. Drong, B. Lehne, M. Loh, W. R. Scott et al., Epigenome-wide association study of body mass index, and the adverse outcomes of adiposity, Nature, vol.541, p.81, 2016.

C. L. Relton, D. Smith, and G. , Two-step epigenetic Mendelian randomization: a strategy for establishing the causal role of epigenetic processes in pathways to disease, Int J Epidemiol, vol.41, issue.1, pp.161-76, 2012.

S. I. Zittermann and A. C. Issekutz, Endothelial growth factors VEGF and bFGF differentially enhance monocyte and neutrophil recruitment to inflammation, J Leukoc Biol, vol.80, issue.2, pp.247-57, 2006.

A. K. Pandey, E. K. Singhi, J. P. Arroyo, T. A. Ikizler, E. R. Gould et al., Mechanisms of VEGF (Vascular Endothelial Growth Factor) inhibitor-associated hypertension and vascular disease, Hypertension, vol.71, issue.2, pp.1-8, 2018.

P. Carmeliet and R. K. Jain, Angiogenesis in cancer and other diseases, Nature, vol.407, issue.6801, p.249, 2000.

H. Berrahmoune, B. Herbeth, J. V. Lamont, C. Masson, P. S. Fitzgerald et al., Six Novel Loci Associated with Circulating VEGF Levels Identified by a Meta-analysis of Genome-Wide Association Studies, Ann Hum Genet, vol.71, issue.2, p.1005874, 2007.

S. Debette, S. Visvikis-siest, M. H. Chen, N. C. Ndiaye, C. Song et al., 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.5, pp.554-63, 2011.

C. Tomii, M. Inokuchi, Y. Takagi, T. Ishikawa, S. Otsuki et al., TPX2 expression is associated with poor survival in gastric cancer, World J Surg Oncol, p.15, 2017.

J. Jian, Y. Huang, L. S. Liu-l-z, and . Deng-f, TPX2 gene-silencing inhibits the proliferation and invasion of human colon cancer SW480 cells, TUMOR, vol.36, issue.6, pp.628-662, 2016.

G. Wang, W. Zhao, X. Gao, D. Zhang, Y. Li et al., HNF1AAS1 promotes growth and metastasis of esophageal squamous cell carcinoma by sponging miR214 to upregulate the expression of SOX-4, Int J Oncol, vol.51, issue.2, pp.657-67, 2017.

M. I. Costache, M. Ioana, S. Iordache, D. Ene, C. A. Costache et al., VEGF Expression in Pancreatic Cancer and Other Malignancies: A Review of the Literature, Rom J Intern Med, vol.53, issue.3, pp.199-208, 2015.

S. Visvikis-siest, J. B. Marteau, A. Samara, H. Berrahmoune, B. Marie et al., Peripheral blood mononuclear cells (PBMCs): a possible model for studying cardiovascular biology systems, Clin Chem Lab Med, vol.45, issue.9, pp.1154-68, 2007.

Y. B. Shaik-dasthagirisaheb, G. Varvara, G. Murmura, A. Saggini, G. Potalivo et al.,

, Mast Cells and Inflammation, Vascular Endothelial Growth Factor (VEGF), vol.26, issue.2, pp.327-362, 2013.

D. Jager, W. Velthuis, H. Prakken, B. J. Kuis, W. Rijkers et al., Simultaneous detection of 15 human cytokines in a single sample of stimulated peripheral blood mononuclear cells, Clin Diagn Lab Immunol, vol.10, issue.1, pp.133-142, 2003.

M. D. Turner, B. Nedjai, T. Hurst, and D. J. Pennington, Cytokines and chemokines: At the crossroads of cell signalling and inflammatory disease, Biochim Biophys Acta, vol.1843, issue.11, pp.2563-82, 2014.

N. Linde, W. Lederle, S. Depner, N. Van-rooijen, C. M. Gutschalk et al., Vascular endothelial growth factor-induced skin carcinogenesis depends on recruitment and alternative activation of macrophages, J Pathol, vol.227, issue.1, pp.17-28, 2012.

D. S. Faffe, L. Flynt, K. Bourgeois, R. A. Panettieri, and S. A. Shore,

, Induce Vascular Endothelial Growth Factor Release from Airway Smooth Muscle Cells: Role of Vascular Endothelial Growth Factor Genotype, Am J Respir Cell Mol Biol, vol.34, issue.2, pp.213-221, 2006.

K. H. Hong, J. Ryu, and K. H. Han, Monocyte chemoattractant protein-1-induced angiogenesis is mediated by vascular endothelial growth factor-A, Blood, vol.105, issue.4, pp.1405-1412, 2005.

F. Ciardiello, T. Troiani, R. Bianco, M. Orditura, F. Morgillo et al., Interaction between the epidermal growth factor receptor (EGFR) and the vascular endothelial growth factor (VEGF) pathways: a rational approach for multi-target anticancer therapy, Ann Oncol, vol.17, issue.7, pp.109-123, 2006.

G. Tortora, F. Ciardiello, and G. Gasparini, Combined targeting of EGFR-dependent and VEGFdependent pathways: rationale, preclinical studies and clinical applications, Nat Clin Pract Oncol, vol.5, issue.9, pp.521-551, 2008.

J. Tabernero, The role of VEGF and EGFR inhibition: implications for combining anti-VEGF and anti-EGFR agents, Mol Cancer Res, vol.5, issue.3, pp.203-223, 2007.

T. Winder and H. J. Lenz, Vascular endothelial growth factor and epidermal growth factor signaling pathways as therapeutic targets for colorectal cancer, Gastroenterology, vol.138, issue.6, pp.2163-76, 2010.

S. P. Huang, M. S. Wu, C. T. Shun, H. P. Wang, M. T. Lin et al., Interleukin-6 increases vascular endothelial growth factor and angiogenesis in gastric carcinoma, J Biomed Sci, vol.11, issue.4, pp.517-544, 2004.

Y. Adachi, C. Aoki, N. Yoshio-hoshino, K. Takayama, D. T. Curiel et al., Interleukin-6 induces both cell growth and VEGF production in malignant mesotheliomas, Int J Cancer, vol.119, issue.6, pp.1303-1314, 2006.

R. F. Tang, S. X. Wang, F. R. Zhang, L. Peng, S. X. Wang et al., Interleukin-1alpha, 6 regulate the secretion of vascular endothelial growth factor A, C in pancreatic cancer, Hepatobiliary Pancreat Dis Int, vol.4, issue.3, pp.460-463, 2005.

S. A. Borg, K. E. Kerry, J. A. Royds, R. D. Battersby, and T. H. Jones, Correlation of VEGF production with IL1 alpha and IL6 secretion by human pituitary adenoma cells, Eur J Endocrinol, vol.152, issue.2, pp.293-300, 2005.

V. Loizzi, D. Vecchio, V. Gargano, G. , D. Liso et al., Biological Pathways Involved in Tumor Angiogenesis and Bevacizumab Based Anti-Angiogenic Therapy with Special References to Ovarian Cancer, Int J Mol Sci, vol.18, issue.9, 2017.

S. S. Savant, S. Sriramkumar, and H. M. O'hagan, The Role of Inflammation and Inflammatory Mediators in the Development, Progression, Metastasis, and Chemoresistance of Epithelial Ovarian Cancer, Cancers (Basel), issue.8, p.10, 2018.

A. J. Lusis, Atherosclerosis. Nature, vol.407, issue.6801, pp.233-274, 2000.

I. Fernández-ruiz, Redefining leukocytes in atherosclerosis, Nature Reviews Cardiology, vol.15, issue.6, p.319, 2018.

T. Jiang, J. Yu, X. Zhu, and L. Tan, TREM2 in Alzheimer's disease, Mol Neurobiol, vol.48, issue.1, pp.180-185, 2013.

J. F. Polak, M. J. Pencina, K. M. Pencina, O. Donnell, C. J. Wolf et al., Carotid-wall intima-media thickness and cardiovascular events, N Engl J Med, vol.365, issue.3, pp.213-234, 2011.

A. D. Giannoukas, G. A. Antoniou, V. Saleptsis, C. Baros, M. Griffin et al., Common femoral artery intima-media thickness as marker for cardiovascular disease in asymptomatic adults, Vasa, vol.38, issue.2, pp.147-54, 2009.

P. J. Touboul, M. G. Hennerici, S. Meairs, H. Adams, P. Amarenco et al., Cerebrovasc Dis, vol.23, issue.1, pp.75-80, 2004.

K. Urschel and I. Cicha, TNF-? in the cardiovascular system: from physiology to therapy, Internat J Interferon Cytokine Med Res, vol.7, pp.9-25, 2015.

I. Petrache, A. Birukova, S. I. Ramirez, J. G. Garcia, and A. D. Verin, The role of the microtubules in tumor necrosis factor-alpha-induced endothelial cell permeability, Am J Respir Cell Mol Biol, vol.28, issue.5, pp.574-81, 2003.

C. M. Steyers, F. J. Miller, and J. , Endothelial dysfunction in chronic inflammatory diseases, Int J Mol Sci, vol.15, issue.7, pp.11324-11373, 2014.

L. Guo, Y. Du, S. Chang, K. Zhang, and J. Wang, rSNPBase: a database for curated regulatory SNPs, Nucleic Acids Res, vol.42, issue.D1, pp.1033-1042, 2013.

I. R. Turnbull, S. Gilfillan, M. Cella, T. Aoshi, M. Miller et al., Cutting edge: TREM-2 attenuates macrophage activation, J Immunol, vol.177, issue.6, pp.3520-3524, 2006.

D. Liu, Y. Dong, Z. Liu, B. Niu, Y. Wang et al., Impact of TREM-2 gene silencing on inflammatory response of endotoxin-induced acute lung injury in mice. Molecular and cellular biochemistry, vol.394, pp.155-61, 2014.

X. Gao, Y. Dong, Z. Liu, and B. Niu, Silencing of triggering receptor expressed on myeloid cells-2 enhances the inflammatory responses of alveolar macrophages to lipopolysaccharide, Molecular medicine reports, vol.7, issue.3, pp.921-927, 2013.

A. Benetos, S. Toupance, S. Gautier, C. Labat, M. Kimura et al., Short leukocyte telomere length precedes clinical expression of atherosclerosis: the blood-and-muscle model, Circ Res, vol.122, issue.4, pp.616-639, 2018.

S. Khan, A. A. Chuturgoon, and D. P. Naidoo, Telomeres and atherosclerosis, Cardiovasc J Afr, vol.23, issue.10, pp.563-71, 2012.

C. Camaré, M. Pucelle, A. Nègre-salvayre, and R. Salvayre, Angiogenesis in the atherosclerotic plaque. Redox biology, vol.12, pp.18-34, 2017.

M. G. Stathopoulou, A. Bonnefond, N. C. Ndiaye, M. Azimi-nezhad, E. Shamieh et al., A common variant highly associated with plasma VEGFA levels also contributes to the variation of both LDL-C and HDL-C, J Lipid Res, vol.54, issue.2, pp.535-576, 2013.
URL : https://hal.archives-ouvertes.fr/hal-01708241

D. R. Zerbino, P. Achuthan, W. Akanni, M. R. Amode, D. Barrell et al., Nucleic Acids Res, vol.46, issue.D1, pp.754-61, 2018.

C. Kut, M. Gabhann, F. Popel, and A. S. , Where is VEGF in the body? A meta-analysis of VEGF distribution in cancer, Br J Cancer, vol.97, p.978, 2007.

M. L. George, S. A. Eccles, M. G. Tutton, A. M. Abulafi, and R. I. Swift, Correlation of plasma and serum vascular endothelial growth factor levels with platelet count in colorectal cancer: clinical evidence of platelet scavenging?, Clin Cancer Res, vol.6, issue.8, pp.3147-52, 2000.

I. Benoy, R. Salgado, C. Colpaert, R. Weytjens, P. B. Vermeulen et al., Serum interleukin 6, plasma VEGF, serum VEGF, and VEGF platelet load in breast cancer patients, Clin Breast Cancer, vol.2, issue.4, pp.311-316, 2002.

N. Rufer, T. H. Brummendorf, S. Kolvraa, C. Bischoff, K. Christensen et al., Telomere fluorescence measurements in granulocytes and T lymphocyte subsets point to a high turnover of hematopoietic stem cells and memory T cells in early childhood, J Exp Med, vol.190, issue.2, pp.157-67, 1999.

K. Youngren, E. Jeanclos, H. Aviv, M. Kimura, J. Stock et al., Synchrony in telomere length of the human fetus, Hum Genet, vol.102, issue.6, pp.640-643, 1998.

J. M. Houben, H. J. Moonen, F. J. Van-schooten, and G. J. Hageman, Telomere length assessment: biomarker of chronic oxidative stress?, Free Radic Biol Med, vol.44, issue.3, pp.235-281, 2008.

A. T. Ludlow, J. B. Zimmerman, S. Witkowski, J. W. Hearn, B. D. Hatfield et al., Relationship between physical activity level, telomere length, and telomerase activity, Med Sci Sports Exerc, vol.40, issue.10, p.1764, 2008.

A. Aviv and J. W. Shay, Reflections on telomere dynamics and ageing-related diseases in humans

, Proc R Soc Lond B Biol Sci, vol.373, p.20160436, 1741.

S. Entringer, K. De-punder, C. Buss, and P. D. Wadhwa, The fetal programming of telomere biology hypothesis: an update, Proc R Soc Lond B Biol Sci, vol.373, p.20170151, 1741.

N. J. Samani, R. Boultby, R. Butler, J. R. Thompson, and A. H. Goodall, Telomere shortening in atherosclerosis, Lancet, vol.358, issue.9280, pp.472-475, 2001.

M. Yang, J. Prescott, E. M. Poole, M. S. Rice, L. D. Kubzansky et al., Prediagnosis Leukocyte Telomere Length and Risk of Ovarian Cancer, Biomarkers & Prevention: A Publication of the American Association for Cancer Research, vol.26, pp.339-384, 2017.

J. L. Sanders and A. B. Newman, Telomere length in epidemiology: a biomarker of aging, age-related disease, both, or neither?, Epidemiol Rev, vol.35, pp.112-143, 2013.

C. Falandry, B. Horard, A. Bruyas, E. Legouffe, J. Cretin et al., Telomere length is a prognostic biomarker in elderly advanced ovarian cancer patients: a multicenter GINECO study, Aging (Milano), vol.7, pp.1066-76, 2015.
URL : https://hal.archives-ouvertes.fr/hal-01850677

I. P. Tzanetakou, R. Nzietchueng, D. N. Perrea, and A. Benetos, Telomeres and their role in aging and longevity, Curr Vasc Pharmacol, vol.12, issue.5, pp.726-760, 2014.

V. Gorenjak, S. Akbar, and M. G. Stathopoulou, Visvikis-Siest S. The future of telomere length in personalized medicine, Front Biosci (Landmark Ed), vol.23, pp.1628-54, 2018.

D. Kuh and Y. B. Shlomo, A life course approach to chronic disease epidemiology, 2004.

C. M. Molster, F. L. Bowman, G. A. Bilkey, A. S. Cho, B. L. Burns et al., The Evolution of Public Health Genomics: Exploring Its Past, Present, and Future. Frontiers in public health, vol.6, p.247, 2018.

H. E. Whitson and C. M. Boyd, Managing multiple comorbidities

, WHO. Cardiovascular diseases (CVDs), p.2019

J. Scott, Pathophysiology and biochemistry of cardiovascular disease, Curr Opin Genet Dev, vol.14, issue.3, pp.271-280, 2004.

C. K. Glass and J. L. Witztum, Atherosclerosis: the road ahead, Cell, vol.104, issue.4, pp.503-519, 2001.

E. Falk, Pathogenesis of atherosclerosis, J Am Coll Cardiol, vol.47, issue.8, pp.7-12, 2006.

B. M. Leon and T. M. Maddox, Diabetes and cardiovascular disease: Epidemiology, biological mechanisms, treatment recommendations and future research, World J Diabetes, vol.6, issue.13, pp.1246-58, 2015.

, Diagnosis and classification of diabetes mellitus, Diabetes Care, vol.33, pp.62-71, 2010.

, WHO. WHO diabetes, 2019.

M. A. Atkinson, G. S. Eisenbarth, and A. W. Michels, Type 1 diabetes, Lancet, vol.383, issue.9911, pp.69-82, 2014.

E. Cersosimo, C. Triplitt, C. Solis-herrera, L. J. Mandarino, and R. A. Defronzo,

K. R. Feingold, B. Anawalt, A. Boyce, G. Chrousos, K. Dungan et al., Diabetes Mellitus, 2000.

M. Szabo, B. Máté, K. Csép, and T. Benedek, Genetic Approaches to the Study of Gene Variants and Their Impact on the Pathophysiology of Type 2 Diabetes, Biochem Genet, vol.56, issue.1, pp.22-55, 2018.

, Association AD. Gestational diabetes mellitus. Diabetes Care, vol.27, issue.1, pp.88-90, 2004.

J. Robitaille and A. M. Grant, The genetics of gestational diabetes mellitus: evidence for relationship with type 2 diabetes mellitus, Genet Med, vol.10, p.240, 2008.

. Who and . Cancer, , 2019.

. Uk-cr, Cancer statistics, 2019.

G. M. Cooper, Sinauer Associates, 2000.

C. Tomasetti, L. Li, and B. Vogelstein, Stem cell divisions, somatic mutations, cancer etiology, and cancer prevention, Science, vol.355, issue.6331, pp.1330-1334, 2017.

F. Islami, G. Sauer, A. Miller, K. D. Siegel, R. L. Fedewa et al., Proportion and number of cancer cases and deaths attributable to potentially modifiable risk factors in the United States, CA Cancer J Clin, vol.68, issue.1, pp.31-54, 2018.

C. Turnbull, A. Sud, and R. S. Houlston, Cancer genetics, precision prevention and a call to action, Nat Genet, p.1, 2018.

A. Nebbioso, F. P. Tambaro, C. Dell'aversana, and L. Altucci, Cancer epigenetics: moving forward, PLoS Genet, vol.14, issue.6, p.1007362, 2018.

L. The, Should we officially recognise obesity as a disease? The lancet Diabetes & endocrinology, vol.5, p.483, 2017.

M. Frellick and C. P. Vega, Is Obesity a Disease? The Health Professionals Point of View: Medscape, 2018.

Y. C. Chooi, C. Ding, and F. Magkos, The epidemiology of obesity, Metabolism, vol.92, pp.6-10, 2019.

F. B. Ortega, C. J. Lavie, and S. N. Blair, Obesity and cardiovascular disease, Circ Res, vol.118, issue.11, pp.1752-70, 2016.

E. E. Calle and R. Kaaks, Overweight, obesity and cancer: epidemiological evidence and proposed mechanisms, Nature Reviews Cancer, vol.4, issue.8, pp.579-91, 2004.

D. García-giustiniani and R. Stein, Genetics of Dyslipidemia, Arq Bras Cardiol, vol.106, issue.5, pp.434-442, 2016.

G. R. Thompson, Management of dyslipidaemia, Heart, vol.90, issue.8, pp.949-55, 2004.

H. Soran, S. Adam, J. B. Mohammad, J. H. Ho, J. D. Schofield et al., Hypercholesterolaemiapractical information for non-specialists, Arch Med Sci, vol.14, issue.1, pp.1-21, 2018.

F. R. Maxfield and I. Tabas, Role of cholesterol and lipid organization in disease, Nature, vol.438, issue.7068, p.612, 2005.

L. Badimon and G. Vilahur, LDL-cholesterol versus HDL-cholesterol in the atherosclerotic plaque: inflammatory resolution versus thrombotic chaos, Ann N Y Acad Sci, vol.1254, pp.18-32, 2012.

C. J. Lin, C. K. Lai, M. C. Kao, L. T. Wu, U. G. Lo et al., Impact of cholesterol on disease progression, Biomedicine (Taipei), vol.5, issue.2, p.7, 2015.

T. S. Wingo, D. J. Cutler, A. P. Wingo, N. Le, G. D. Rabinovici et al., Association of Early-Onset Alzheimer Disease With Elevated Low-Density Lipoprotein Cholesterol Levels and Rare Genetic Coding Variants of APOBAssociation of Early-Onset AD With Elevated LDL-C Levels and Rare APOB Coding VariantsAssociation of Early-Onset AD With Elevated LDL-C Levels and Rare APOB Coding Variants, JAMA Neurology, vol.76, issue.7, pp.809-826, 2019.

R. N. Pejic, D. T. Lee, and . Hypertriglyceridemia, J Am Board Fam Med, vol.19, issue.3, pp.310-316, 2006.

P. P. Toth, Triglyceride-rich lipoproteins as a causal factor for cardiovascular disease. Vasc Health Risk Manag, vol.12, pp.171-83, 2016.

J. Pang, D. C. Chan, and G. F. Watts, Origin and therapy for hypertriglyceridaemia in type 2 diabetes, World J Diabetes, vol.5, issue.2, pp.165-75, 2014.

A. Tenenbaum, R. Klempfner, and E. Z. Fisman, Hypertriglyceridemia: a too long unfairly neglected major cardiovascular risk factor, Cardiovasc Diabetol, vol.13, p.159, 2014.

S. Mora, N. Cook, J. E. Buring, P. M. Ridker, and I. M. Lee, Physical activity and reduced risk of cardiovascular events: potential mediating mechanisms. Circulation, vol.116, pp.2110-2118, 2007.

A. Mok, K. T. Khaw, R. Luben, N. Wareham, and S. Brage, Physical activity trajectories and mortality: population based cohort study, BMJ, vol.365, p.2323, 2019.

. Who and . Tobacco, Free Initiative, 2019.

D. G. Yanbaeva, M. A. Dentener, E. C. Creutzberg, G. Wesseling, and E. F. Wouters, Systemic effects of smoking, Chest, vol.131, issue.5, pp.1557-66, 2007.

S. Salahuddin, D. Prabhakaran, and A. Roy, Pathophysiological Mechanisms of Tobacco-Related CVD, Glob Heart, vol.7, issue.2, pp.113-133, 2012.

S. S. Hecht, Cigarette smoking: cancer risks, carcinogens, and mechanisms. Langenbeck's archives of surgery, vol.391, pp.603-616, 2006.

R. Dhingra and R. S. Vasan, Age as a risk factor, Med Clin North Am, vol.96, issue.1, pp.87-91, 2012.

L. Partridge, The new biology of ageing, Philos Trans R Soc Lond B Biol Sci, vol.365, pp.147-54, 1537.

, Global, regional, and national age-sex-specific mortality for 282 causes of death in 195 countries and territories, 1980-2017: a systematic analysis for the Global Burden of Disease Study, Lancet, vol.392, pp.1736-88, 2017.

V. Regitz-zagrosek, Sex and gender differences in health, Science & Society Series on Sex and Science. EMBO Rep, vol.13, issue.7, pp.596-603, 2012.

B. M. Altevogt, T. Wizemann, and D. E. Pankevich, Sex differences and implications for translational neuroscience research: workshop summary: National Academies Press, 2011.

L. C. Golden and R. Voskuhl, The importance of studying sex differences in disease: The example of multiple sclerosis, J Neurosci Res, vol.95, issue.1-2, pp.633-676, 2017.

C. Ober, D. A. Loisel, and Y. Gilad, Sex-specific genetic architecture of human disease, Nat Rev Genet, vol.9, issue.12, pp.911-933, 2008.

N. Genetic and N. S. Services, Understanding genetics: a New York, mid-Atlantic guide for patients and health professionals: Lulu. com, 2009.

. Center-gsl and . Learn, Genetics: Genetic Risk, 2019.

G. K. Alderton, Common disease pathogenesis pathways, Nature Reviews Genetics, vol.11, p.386, 2010.

B. A. Imhof and M. Aurrand-lions, Angiogenesis and inflammation face off, Nat Med, vol.12, issue.2, pp.171-173, 2006.

D. Ribatti and E. Crivellato, Immune cells and angiogenesis, J Cell Mol Med, vol.13, issue.9a, pp.2822-2855, 2009.

R. Medzhitov, Inflammation 2010: new adventures of an old flame, Cell, vol.140, issue.6, pp.771-777, 2010.

L. Chen, H. Deng, H. Cui, J. Fang, Z. Zuo et al., Inflammatory responses and inflammationassociated diseases in organs, Oncotarget, vol.9, issue.6, pp.7204-7222, 2018.

R. Medzhitov, Origin and physiological roles of inflammation, Nature, vol.454, issue.7203, pp.428-463, 2008.

R. Scrivo, M. Vasile, I. Bartosiewicz, and G. Valesini, Inflammation as "common soil" of the multifactorial diseases, Autoimmun Rev, vol.10, issue.7, pp.369-74, 2011.

S. Prasad, B. Sung, and B. B. Aggarwal, Age-associated chronic diseases require age-old medicine: role of chronic inflammation, Prev Med, vol.54, pp.29-37, 2012.

J. M. Zhang and J. An, Cytokines, inflammation, and pain, Int Anesthesiol Clin, vol.45, issue.2, pp.27-37, 2007.

C. A. Feghali and T. M. Wright, Cytokines in acute and chronic inflammation, Front Biosci, vol.2, issue.1, pp.12-26, 1997.

M. Genua, S. Rutella, C. Correale, and S. Danese, The triggering receptor expressed on myeloid cells (TREM) in inflammatory bowel disease pathogenesis, Journal of Translational Medicine, vol.12, issue.1, p.293, 2014.

J. W. Ford and D. W. Mcvicar, TREM and TREM-like receptors in inflammation and disease. Current opinion in immunology, vol.21, pp.38-46, 2009.

R. J. Arts, L. A. Joosten, J. W. Van-der-meer, and M. G. Netea, TREM-1: intracellular signaling pathways and interaction with pattern recognition receptors, Journal of leukocyte biology, vol.93, issue.2, pp.209-224, 2013.

C. Reitz and R. Mayeux, The New England journal of medicine, vol.369, pp.1564-1569, 2013.

D. L. Kober and T. J. Brett, TREM2-ligand interactions in health and disease, Journal of molecular biology, vol.429, issue.11, pp.1607-1636, 2017.

P. Carmeliet, Mechanisms of angiogenesis and arteriogenesis, Nat Med, vol.6, issue.4, pp.389-95, 2000.

W. Risau, Mechanisms of angiogenesis, Nature, vol.386, issue.6626, p.671, 1997.

M. Hofmann and J. Heineke, The Impact of Endothelial Transcription Factors in Sprouting Angiogenesis. Tumor Angiogenesis: A Key Target for Cancer Therapy, pp.1-18, 2018.

T. Adair, J. Montani, and . Angiogenesis, Morgan & Claypool Life Sciences, 2010.

F. Dehghanian, Z. Hojati, and M. Kay, New Insights into VEGF-A Alternative Splicing: Key Regulatory Switching in the Pathological Process, Avicenna J Med Biotechnol, vol.6, issue.4, pp.192-201, 2014.

K. H. Plate, G. Breier, and W. Risau, Molecular mechanisms of developmental and tumor angiogenesis, Brain Pathol, vol.4, issue.3, pp.207-225, 1994.

L. M. Ellis and D. J. Hicklin, VEGF-targeted therapy: mechanisms of anti-tumour activity, Nature reviews cancer, vol.8, issue.8, p.579, 2008.

N. Ferrara and R. S. Kerbel, Angiogenesis as a therapeutic target, Nature, vol.438, issue.7070, p.967, 2005.

T. Sakurai and M. Kudo, Signaling pathways governing tumor angiogenesis, Oncology, vol.81, issue.1, pp.24-33, 2011.

. Group and . Bdw, Biomarkers and surrogate endpoints: preferred definitions and conceptual framework, Clin Pharmacol Ther, vol.69, pp.89-95, 2001.

, Group F-NBW. BEST (Biomarkers, EndpointS, and other Tools) Resource. Silver Spring (MD): Food and Drug Administration

, Bethesda (MD): National Institutes of Health (US), 2016.

H. B. Burke, Predicting Clinical Outcomes Using Molecular Biomarkers, Biomark Cancer, vol.8, pp.89-99, 2016.

M. J. Selleck, M. Senthil, and N. R. Wall, Making Meaningful Clinical Use of Biomarkers, Biomark Insights, vol.12, p.1177271917715236, 2017.

G. Novelli, C. Ciccacci, P. Borgiani, P. Amati, M. Abadie et al., Genetic tests and genomic biomarkers: regulation, qualification and validation. Clin Cases Miner Bone Metab, vol.5, pp.149-54, 2008.

N. Ferrara, H. P. Gerber, and J. Lecouter, The biology of VEGF and its receptors, Nat Med, vol.9, issue.6, pp.669-76, 2003.

J. E. Nör, J. Christensen, D. J. Mooney, and P. J. Polverini, Vascular endothelial growth factor (VEGF)-mediated angiogenesis is associated with enhanced endothelial cell survival and induction of Bcl-2 expression, Am J Pathol, vol.154, issue.2, pp.375-84, 1999.

N. Ferrara, K. J. Hillan, W. Novotny, and . Bevacizumab, Avastin), a humanized anti-VEGF monoclonal antibody for cancer therapy, Biochem Biophys Res Commun, vol.333, issue.2, pp.328-363, 2005.

J. Woolard, H. S. Bevan, S. J. Harper, and D. O. Bates, Molecular diversity of VEGF-A as a regulator of its biological activity, Microcirculation, vol.16, issue.7, pp.572-92, 2009.

D. G. Nowak, E. M. Amin, E. S. Rennel, C. Hoareau-aveilla, M. Gammons et al., Regulation of vascular endothelial growth factor (VEGF) splicing from pro-angiogenic to antiangiogenic isoforms: a novel therapeutic strategy for angiogenesis, J Biol Chem, vol.285, issue.8, pp.5532-5572, 2010.

S. J. Harper and D. O. Bates, VEGF-A splicing: the key to anti-angiogenic therapeutics?, Nat Rev Cancer, vol.8, issue.11, pp.880-887, 2008.

K. L. Meadows and H. I. Hurwitz, Anti-VEGF therapies in the clinic, Cold Spring Harb Perspect Med, vol.2, issue.10, 2012.

W. Zhang, Z. Shen, H. Luo, X. Hu, L. Zheng et al., The Benefits and Side Effects of Bevacizumab for the Treatment of Recurrent Ovarian Cancer, Curr Drug Targets, vol.18, issue.10, pp.1125-1156, 2017.

S. Riondino, G. D. Monte, F. Fratangeli, F. Guadagni, M. Roselli et al., Anti-angiogenic drugs, vascular toxicity and thromboembolism in solid cancer, Cardiovasc Hematol Agents Med Chem, 2017.

M. R. Ladomery, S. J. Harper, and D. O. Bates, Alternative splicing in angiogenesis: the vascular endothelial growth factor paradigm, Cancer Lett, vol.249, issue.2, pp.133-175, 2007.

D. G. Nowak, J. Woolard, E. M. Amin, O. Konopatskaya, M. A. Saleem et al., Expression of pro-and anti-angiogenic isoforms of VEGF is differentially regulated by splicing and growth factors, J Cell Sci, vol.121, pp.3487-95, 2008.

R. K. Moyzis, J. M. Buckingham, L. S. Cram, M. Dani, L. L. Deaven et al., A highly conserved repetitive DNA sequence, (TTAGGG)n, present at the telomeres of human chromosomes, Proc Natl Acad Sci, vol.85, issue.18, pp.6622-6628, 1988.

R. J. O'sullivan and J. Karlseder, Telomeres: protecting chromosomes against genome instability, Nat Rev Mol Cell Biol, vol.11, issue.3, pp.171-81, 2010.

J. Lindsey, N. I. Mcgill, L. A. Lindsey, D. K. Green, and H. J. Cooke, In vivo loss of telomeric repeats with age in humans, Mutat Res, vol.256, issue.1, pp.45-53, 1991.

F. Fdad, P. M. Reaper, L. Clay-farrace, H. Fiegler, P. Carr et al., A DNA damage checkpoint response in telomere-initiated senescence, Nature, vol.426, p.194, 2003.

Y. Cong, W. E. Wright, and J. W. Shay, Human Telomerase and Its Regulation, Microbiol Mol Biol Rev, vol.66, pp.407-432, 2002.

N. W. Kim, M. A. Piatyszek, K. R. Prowse, C. B. Harley, M. D. West et al., Specific association of human telomerase activity with immortal cells and cancer, Science, vol.266, issue.5193, pp.2011-2016, 1994.

O. T. Njajou, R. M. Cawthon, C. M. Damcott, S. Wu, S. Ott et al., Telomere length is paternally inherited and is associated with parental lifespan, Proc Natl Acad Sci, vol.104, pp.12135-12144, 2007.

M. Vasa-nicotera, S. Brouilette, M. Mangino, J. R. Thompson, P. Braund et al., Mapping of a major locus that determines telomere length in humans, Am J Hum Genet, vol.76, pp.147-51, 2005.

T. Andrew, A. Aviv, M. Falchi, G. L. Surdulescu, J. P. Gardner et al., Mapping genetic loci that determine leukocyte telomere length in a large sample of unselected female sibling pairs, Am J Hum Genet, vol.78, pp.480-486, 2006.

G. Atzmon, M. Cho, R. M. Cawthon, T. Budagov, M. Katz et al., Evolution in health and medicine Sackler colloquium: Genetic variation in human telomerase is associated with telomere length in Ashkenazi centenarians, Proc Natl Acad Sci, vol.107, issue.1, pp.1710-1717, 2010.

M. Mangino, J. B. Richards, N. Soranzo, G. Zhai, A. Aviv et al., A genome-wide association study identifies a novel locus on chromosome 18q12.2 influencing white cell telomere length, J Med Genet, vol.46, pp.451-455, 2009.
URL : https://hal.archives-ouvertes.fr/hal-00552680

V. Codd, M. Mangino, P. Van-der-harst, P. S. Braund, M. Kaiser et al., Variants near TERC are associated with mean telomere length, Nat Genet, vol.42, pp.197-206, 2010.

D. Levy, S. L. Neuhausen, S. C. Hunt, M. Kimura, S. Hwang et al., Genome-wide association identifies OBFC1 as a locus involved in human leukocyte telomere biology, Proc Natl Acad Sci U S A, vol.107, pp.9293-9301, 2010.

J. Gu, M. Chen, S. Shete, C. I. Amos, A. Kamat et al., A genome-wide association study identifies a locus on chromosome 14q21 as a predictor of leukocyte telomere length and as a marker of susceptibility for bladder cancer, Cancer Prev Res (Phila), vol.4, pp.514-535, 2011.

J. Prescott, P. Kraft, D. I. Chasman, S. A. Savage, L. Mirabello et al., Genome-wide association study of relative telomere length, PLoS One, vol.6, p.19635, 2011.

M. Mangino, S. Hwang, T. D. Spector, S. C. Hunt, M. Kimura et al., Genome-wide meta-analysis points to CTC1 and ZNF676 as genes regulating telomere homeostasis in humans, Hum Mol Genet, vol.21, pp.5385-94, 2012.

V. Codd, C. P. Nelson, A. E. Mangino, M. Deelen, J. Buxton et al., Identification of seven loci affecting mean telomere length and their association with disease, Nat Genet, vol.45, pp.7-8, 2013.

K. A. Pooley, S. E. Bojesen, M. Weischer, S. F. Nielsen, D. Thompson et al., A genome-wide association scan (GWAS) for mean telomere length within the COGS project: identified loci show little association with hormone-related cancer risk, Hum Mol Genet, vol.22, pp.5056-64, 2013.

Y. Liu, L. Cao, Z. Li, D. Zhou, W. Liu et al., A genome-wide association study identifies a locus on TERT for mean telomere length in Han Chinese, PLoS One, vol.9, p.85043, 2014.

R. Saxena, A. Bjonnes, J. Prescott, P. Dib, P. Natt et al., Genome-wide association study identifies variants in casein kinase II (CSNK2A2) to be associated with leukocyte telomere length in a Punjabi Sikh diabetic cohort, Circ Cardiovasc Genet, vol.7, pp.287-95, 2014.

K. M. Walsh, V. Codd, I. V. Smirnov, T. Rice, P. A. Decker et al., Variants near TERT and TERC influencing telomere length are associated with high-grade glioma risk, Nat Genet, vol.46, pp.731-736, 2014.

E. R. Rietzschel, S. Bekaert, D. Meyer, and T. , Telomeres and Atherosclerosis: The Attrition of an Attractive Hypothesis, J Am Coll Cardiol, vol.67, issue.21, pp.2477-2486, 2016.

M. Armanios and E. H. Blackburn, The telomere syndromes, Nat Rev Genet, vol.13, pp.693-704, 2012.

M. A. Blasco, Telomeres and human disease: ageing, cancer and beyond, Nat Rev Genet, vol.6, pp.611-633, 2005.

R. T. Calado and N. S. Young, Telomere Diseases, The New England Journal of Medicine, vol.361, pp.2353-65, 2009.

M. L. Slattery, K. Curtin, R. Baumgartner, C. Sweeney, T. Byers et al., IL6, aspirin, nonsteroidal anti-inflammatory drugs, and breast cancer risk in women living in the southwestern United States. Cancer Epidemiology, Biomarkers & Prevention: A Publication of the, Cosponsored by the American Society of Preventive Oncology, vol.16, pp.747-55, 2007.

A. O'donovan, M. S. Pantell, E. Puterman, F. S. Dhabhar, E. H. Blackburn et al., Cumulative inflammatory load is associated with short leukocyte telomere length in the Health, Aging and Body Composition Study, PLoS One, vol.6, issue.5, p.19687, 2011.

D. A. Glei, N. Goldman, M. Weinstein, and R. A. Risques, Shorter Ends, Faster End? Leukocyte Telomere Length and Mortality Among Older Taiwanese, Biological Sciences and Medical Sciences, vol.70, pp.1490-1498, 2015.

Y. Maida, S. Kyo, T. Kanaya, Z. Wang, N. Yatabe et al., Direct activation of telomerase by EGF through Ets-mediated transactivation of TERT via MAP kinase signaling pathway, Oncogene, vol.21, pp.4071-4080, 2002.

E. Council, Council conclusions on personalised medicine for patients, Journal of the European Union, p.421, 2015.

W. K. Redekop and D. Mladsi, The faces of personalized medicine: a framework for understanding its meaning and scope. Value Health, vol.16, pp.4-9, 2013.

F. Goodsaid, F. Frueh, and M. E. Burczynski, Personalized Medicine. Drug Discovery and Evaluation: Methods in Clinical Pharmacology, pp.1-14, 2018.

D. Kim, Y. Kim, N. Son, C. Kang, and A. Kim, Recent omics technologies and their emerging applications for personalised medicine, IET Syst Biol, vol.11, issue.3, pp.87-98, 2017.

M. D. Ritchie, E. R. Holzinger, R. Li, S. A. Pendergrass, and D. Kim, Methods of integrating data to uncover genotype-phenotype interactions, Nature Reviews Genetics, vol.16, p.85, 2015.

F. Thibaut, From basic research to personalized medicine, Dialogues Clin Neurosci, vol.18, issue.3, pp.231-233, 2016.

R. L. Nussbaum, R. R. Mcinnes, and H. F. Willard, Thompson & Thompson genetics in medicine e-book, 2015.

F. Camastra, D. Taranto, M. D. Staiano, and A. , Statistical and computational methods for genetic diseases: An overview, Comput Math Methods Med, 2015.

J. S. Witte, Genome-wide association studies and beyond, Annu Rev Public Health, vol.31, p.9, 2010.

A. B. Zheutlin and D. A. Ross, Polygenic risk scores: What are they good for? Biological psychiatry, vol.83, pp.51-54, 2018.

A. V. Khera, M. Chaffin, K. G. Aragam, C. A. Emdin, D. Klarin et al., Genome-wide polygenic score to identify a monogenic risk-equivalent for coronary disease, p.218388

J. L. Jameson and D. L. Longo, Precision medicine-personalized, problematic, and promising, Obstet Gynecol Surv, vol.70, issue.10, pp.612-616, 2015.

L. R. Cardon and J. I. Bell, Association study designs for complex diseases, Nature Reviews Genetics, vol.2, issue.2, pp.91-100, 2001.

D. J. Balding, A tutorial on statistical methods for population association studies, Nature Reviews Genetics, vol.7, issue.10, pp.781-91, 2006.

C. M. Miles and M. Wayne, Quantitative Trait Locus (QTL) Analysis, Nature Education, vol.1, issue.1, 2008.

M. C. Mills and C. Rahal, A scientometric review of genome-wide association studies, Commun Biol, vol.2, p.9, 2019.

M. T. Dorak, Genetic association studies: background, conduct, analysis, interpretation: Garland Science, 2016.

K. Heggenhougen, International encyclopedia of public health, 2008.

J. I. Hoffman, Biostatistics for medical and biomedical practitioners, 2015.

P. K. Gupta, P. L. Kulwal, and J. V. Chapter, Association Mapping in Crop Plants: Opportunities and Challenges, vol.85, pp.109-156, 2014.

M. D. Gallagher and A. S. Chen-plotkin, The Post-GWAS Era: From Association to Function, Am J Hum Genet, vol.102, issue.5, pp.717-747, 2018.

M. V. Holmes, F. W. Asselbergs, T. M. Palmer, F. Drenos, M. B. Lanktree et al., Mendelian randomization of blood lipids for coronary heart disease, Eur Heart J, vol.36, issue.9, pp.539-50, 2014.

M. V. Holmes, M. Ala-korpela, and G. D. Smith, Mendelian randomization in cardiometabolic disease: challenges in evaluating causality, Nature Reviews Cardiology, vol.14, p.577, 2017.

N. M. Davies, M. V. Holmes, D. Smith, and G. , Reading Mendelian randomisation studies: a guide, glossary, and checklist for clinicians, BMJ, vol.362, p.601, 2018.

P. Sekula, F. D. Greco, M. Pattaro, C. Köttgen, and A. , Mendelian randomization as an approach to assess causality using observational data, J Am Soc Nephrol, vol.27, issue.11, pp.3253-65, 2016.

G. Thanassoulis, C. J. O'donnell, B. A. Raby, and H. N. Sokol, Mendelian randomization. Topic 13514 Version 1202018

A. D. Goldberg, C. D. Allis, and E. Bernstein, Epigenetics: a landscape takes shape, Cell, vol.128, issue.4, pp.635-643, 2007.

M. Bemer, Unraveling the Complex Epigenetic Mechanisms that Regulate Gene Activity, Methods in molecular biology, vol.1675, pp.205-236, 2018.

U. Weber-stadlbauer, Epigenetic and transgenerational mechanisms in infection-mediated neurodevelopmental disorders, Transl Psychiatry, vol.7, issue.5, p.1113, 2017.

G. Egger, G. Liang, A. Aparicio, and P. A. Jones, Epigenetics in human disease and prospects for epigenetic therapy, Nature, vol.429, issue.6990, pp.457-63, 2004.

A. Portela and M. Esteller, Epigenetic modifications and human disease, Nat Biotechnol, vol.28, issue.10, pp.1057-68, 2010.

X. S. Liu, H. Wu, J. X. Stelzer, Y. Wu, X. Czauderna et al., Editing DNA methylation in the mammalian genome, Cell, vol.167, issue.1, pp.233-280, 2016.

D. Schübeler, Function and information content of DNA methylation, Nature, vol.517, issue.7534, p.321, 2015.

K. D. Robertson, DNA methylation and human disease, Nature reviews Genetics, vol.6, issue.8, pp.597-610, 2005.

B. Jin, Y. Li, and K. D. Robertson, DNA methylation: superior or subordinate in the epigenetic hierarchy?, Genes & cancer, vol.2, issue.6, pp.607-624, 2011.

R. M. Kohli and Y. Zhang, TET enzymes, TDG and the dynamics of DNA demethylation, Nature, vol.502, issue.7472, pp.472-481, 2013.

T. F. Imperiale, D. F. Ransohoff, S. H. Itzkowitz, T. R. Levin, P. Lavin et al., Multitarget stool DNA testing for colorectal-cancer screening. The New England journal of medicine, vol.370, pp.1287-97, 2014.

C. C. Chang and H. C. Wang, The feasibility of detecting endometrial and ovarian cancer using DNA methylation biomarkers in cervical scrapings, vol.29, p.17, 2018.

W. Zhang, M. Song, J. Qu, and G. H. Liu, Epigenetic Modifications in Cardiovascular Aging and Diseases, Circulation research, vol.123, issue.7, pp.773-86, 2018.

V. K. Rakyan, T. A. Down, D. J. Balding, and S. Beck, Epigenome-wide association studies for common human diseases, Nat Rev Genet, vol.12, issue.8, pp.529-570, 2011.

S. Dedeurwaerder, M. Defrance, M. Bizet, E. Calonne, G. Bontempi et al., A comprehensive overview of Infinium HumanMethylation450 data processing, Brief Bioinform, vol.15, issue.6, pp.929-970, 2014.

B. Alberts, D. Bray, K. Hopkin, A. D. Johnson, J. Lewis et al., Essential cell biology: Garland Science, 2013.

L. Uo, Gene Expression and Regulation, 2019.

I. S. Segundo-val and C. S. Sanz-lozano, Introduction to the Gene Expression Analysis, Methods Mol Biol, vol.1434, pp.29-43, 2016.

Z. Wang, M. Gerstein, and M. Snyder, RNA-Seq: a revolutionary tool for transcriptomics, Nature Reviews Genetics, vol.10, p.57, 2009.

L. Tayebi and K. Moharamzadeh, Biomaterials for Oral and Dental Tissue Engineering, 2017.

P. Vidhyasekaran, Handbook of molecular technologies in crop disease management, 2013.

R. Lowe, N. Shirley, M. Bleackley, S. Dolan, and T. Shafee, Transcriptomics technologies, PLoS Comput Biol, vol.13, issue.5, p.1005457, 2017.

D. Huber, V. Voithenberg, L. V. Kaigala, and G. , Fluorescence in situ hybridization (FISH): history, limitations and what to expect from micro-scale FISH? Micro and Nano Engineering, 2018.

P. M. Loewenstein, C. Z. Song, and M. Green, The use of in vitro transcription to probe regulatory functions of viral protein domains, Methods Mol Med, vol.131, pp.15-31, 2007.

L. George, F. E. Indig, K. Abdelmohsen, and M. Gorospe, Intracellular RNA-tracking methods, Royal Society Open Biology, vol.8, issue.10, p.180104, 2018.

P. Deverka and R. J. Carlson, Personalized Medicine: Current and Future Perspectives, 2019.

M. Vivot, The 5 Challenges of Personalised Medicine: European Commission, 2016.

G. Siest, S. Visvikis, B. Herbeth, R. Gueguen, M. Vincent-viry 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, Clin Chem Lab Med, vol.36, issue.1, pp.35-42, 1998.

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.46, issue.6, pp.733-780, 2008.

A. C. Nica, L. Parts, D. Glass, J. Nisbet, A. Barrett et al., The architecture of gene regulatory variation across multiple human tissues: the MuTHER study, PLoS Genet, vol.7, issue.2, p.1002003, 2011.

S. A. Miller, D. D. Dykes, and H. F. Polesky, A simple salting out procedure for extracting DNA from human nucleated cells, Nucleic Acids Res, vol.16, issue.3, p.1215, 1988.

C. A. Glastonbury, A. Viñuela, A. Buil, G. H. Halldorsson, G. Thorleifsson et al., Adiposity-Dependent Regulatory Effects on Multi-tissue Transcriptomes, Am J Hum Genet, vol.99, issue.3, pp.567-79, 2016.

M. Bibikova, B. Barnes, C. Tsan, V. Ho, B. Klotzle et al., High density DNA methylation array with single CpG site resolution, Genomics, vol.98, issue.4, pp.288-95, 2011.

C. He, J. Holme, and J. Anthony, SNP genotyping: the KASP assay, Methods Mol Biol, vol.1145, pp.75-86, 2014.

A. Boyum, Isolation of mononuclear cells and granulocytes from human blood. Isolation of monuclear cells by one centrifugation, and of granulocytes by combining centrifugation and sedimentation at 1 g, Scand J Clin Lab Invest Suppl, vol.97, pp.77-89, 1968.

M. Azimi-nezhad, M. G. Stathopoulou, A. Bonnefond, M. Rancier, A. Saleh et al., Associations of vascular endothelial growth factor (VEGF) with adhesion and inflammation molecules in a healthy population, Cytokine, vol.61, issue.2, pp.602-609, 2013.
URL : https://hal.archives-ouvertes.fr/hal-01708258

J. B. Marteau, S. Mohr, M. Pfister, and S. Visvikis-siest, Collection and storage of human blood cells for mRNA expression profiling: a 15-month stability study, Clin Chem, vol.51, issue.7, pp.1250-1252, 2005.

W. T. Friedewald, R. I. Levy, and D. S. Fredrickson, Estimation of the concentration of low-density lipoprotein cholesterol in plasma, without use of the preparative ultracentrifuge, Clin Chem, vol.18, issue.6, pp.499-502, 1972.

M. B. Schulze, E. B. Rimm, I. Shai, N. Rifai, and F. B. Hu, Relationship between adiponectin and glycemic control, blood lipids, and inflammatory markers in men with type 2 diabetes, Diabetes Care, vol.27, issue.7, pp.1680-1687, 2004.

M. R. Irvin, D. Zhi, R. Joehanes, M. Mendelson, S. Aslibekyan et al., Epigenome-wide association study of fasting blood lipids in the genetics of lipid-lowering drugs and diet network study, Circulation, vol.130, issue.7, pp.565-72, 2014.

P. G. Kopelman, Obesity as a medical problem, Nature, vol.404, issue.6778, pp.635-678, 2000.

C. Lee, R. R. Huxley, R. P. Wildman, and M. Woodward, Indices of abdominal obesity are better discriminators of cardiovascular risk factors than BMI: a meta-analysis, J Clin Epidemiol, vol.61, issue.7, pp.646-53, 2008.

I. Janssen, P. T. Katzmarzyk, and R. Ross, Waist circumference and not body mass index explains obesity-related health risk, Am J Clin Nutr, vol.79, issue.3, pp.379-84, 2004.

J. Moustafa and P. Froguel, From obesity genetics to the future of personalized obesity therapy, Nature Reviews Endocrinology, vol.9, issue.7, p.402, 2013.

S. J. Van-dijk, P. L. Molloy, H. Varinli, J. L. Morrison, and B. S. Muhlhausler, Epigenetics and human obesity, Int J Obes (Lond), vol.39, issue.1, pp.85-97, 2015.

A. M. Byrne, D. J. Bouchier-hayes, and J. H. Harmey, Angiogenic and cell survival functions of vascular endothelial growth factor (VEGF), J Cell Mol Med, vol.9, issue.4, pp.777-94, 2005.

H. M. Berman, J. Westbrook, Z. Feng, G. Gilliland, T. N. Bhat et al., The Protein Data Bank, Nucleic Acids Res, vol.28, issue.1, pp.235-277, 2000.

Y. A. Muller, B. Li, H. W. Christinger, J. A. Wells, B. C. Cunningham et al., Vascular endothelial growth factor: crystal structure and functional mapping of the kinase domain receptor binding site, Proc Natl Acad Sci, vol.94, issue.14, pp.7192-7199, 1997.

F. Balkwill and A. Mantovani, Inflammation and cancer: back to Virchow? The lancet, vol.357, pp.539-584, 2001.

T. Muir, Peripheral Blood Mononuclear Cells: A Brief Review: stemexpress, 2019.

C. P. Corkum, D. P. Ings, C. Burgess, S. Karwowska, W. Kroll et al., Immune cell subsets and their gene expression profiles from human PBMC isolated by Vacutainer Cell Preparation Tube (CPT(?)) and standard density gradient, BMC Immunol, vol.16, 2015.

M. Colonna and Y. Wang, TREM2 variants: new keys to decipher Alzheimer disease pathogenesis, Nat Rev Neurosci, vol.17, issue.4, pp.201-208, 2016.

R. Guerreiro, A. Wojtas, J. Bras, M. Carrasquillo, E. Rogaeva et al., TREM2 Variants in Alzheimer's Disease, N Engl J Med, vol.368, issue.2, pp.117-144, 2013.

L. S. Boesten, A. S. Zadelaar, A. Van-nieuwkoop, M. J. Gijbels, M. P. De-winther et al., Tumor necrosis factor-alpha promotes atherosclerotic lesion progression in APOE*3-Leiden transgenic mice, Cardiovasc Res, vol.66, issue.1, pp.179-85, 2005.

J. Gariepy, A. Simon, M. Massonneau, A. Linhart, and J. Levenson, Wall thickening of carotid and femoral arteries in male subjects with isolated hypercholesterolemia. PCVMETRA Group. Prevention Cardio-Vasculaire en Medecine du Travail, Atherosclerosis, vol.113, issue.2, pp.141-51, 1995.

F. Zhao, M. Song, Y. Wang, and W. Wang, Genetic model, J Cell Mol Med, vol.20, issue.4, p.765, 2016.

S. H. Lee, N. Griffiths, and L. Shao, Can telomere length be used as a biomarker for cardiovascular diseases?: insights from a large clinical study, J Physiol, 2017.

M. Lee, C. E. Napier, S. F. Yang, J. W. Arthur, R. R. Reddel et al., Comparative analysis of whole genome sequencing-based telomere length measurement techniques, Methods, vol.114, pp.4-15, 2017.

P. C. Haycock, S. Burgess, A. Nounu, J. Zheng, G. N. Okoli et al., Association Between Telomere Length and Risk of Cancer and Non-Neoplastic Diseases: A Mendelian Randomization Study, JAMA Oncol, vol.3, issue.5, pp.636-51, 2017.

K. Mccloskey, A. L. Ponsonby, F. Collier, K. Allen, M. Tang et al., The association between higher maternal pre-pregnancy body mass index and increased birth weight, adiposity and inflammation in the newborn, Pediatr Obes, vol.13, issue.1, pp.46-53, 2018.

H. M. Salihu, A. Pradhan, L. King, A. Paothong, C. Nwoga et al., Impact of intrauterine tobacco exposure on fetal telomere length, Am J Obstet Gynecol, vol.212, issue.2, pp.205-206, 2015.

T. S. Send, M. Gilles, V. Codd, I. Wolf, S. Bardtke et al., Telomere Length in Newborns is Related to Maternal Stress During Pregnancy, Neuropsychopharmacology, vol.42, issue.12, pp.2407-2420, 2017.

I. Shalev, S. Entringer, P. D. Wadhwa, O. M. Wolkowitz, E. Puterman et al., Stress and telomere biology: a lifespan perspective, Psychoneuroendocrinology, vol.38, issue.9, pp.1835-1877, 2013.

D. S. Martens, M. Plusquin, W. Gyselaers, D. Vivo, I. Nawrot et al., Maternal pre-pregnancy body mass index and newborn telomere length, BMC Med, vol.14, issue.1, p.148, 2016.

U. Friedrich, M. Schwab, E. U. Griese, P. Fritz, and U. Klotz, Telomeres in neonates: new insights in fetal hematopoiesis, Pediatr Res, vol.49, issue.2, pp.252-258, 2001.

K. Okuda, A. Bardeguez, J. P. Gardner, P. Rodriguez, V. Ganesh et al., Telomere length in the newborn, Pediatr Res, vol.52, issue.3, pp.377-81, 2002.

M. G. Stathopoulou, A. M. Petrelis, J. L. Buxton, P. Froguel, and A. I. Blakemore, Visvikis-Siest S. Genetic determinants of leucocyte telomere length in children: a neglected and challenging field, Paediatr Perinat Epidemiol, vol.29, issue.2, pp.146-50, 2015.

A. M. Zeiger, M. J. White, C. Eng, S. S. Oh, J. Witonsky et al., Genetic Determinants of Telomere Length in African American Youth. Sci Rep, vol.8, issue.1, p.13265, 2018.

E. H. Blackburn, Structure and function of telomeres, Nature, vol.350, pp.569-73, 1991.

G. K. Hansson, Inflammation, atherosclerosis, and coronary artery disease, N Engl J Med, vol.352, issue.16, pp.1685-95, 2005.

P. Carmeliet, VEGF as a key mediator of angiogenesis in cancer, Oncology, vol.69, pp.4-10, 2005.

U. Testa, G. Pannitteri, and G. L. Condorelli, Vascular endothelial growth factors in cardiovascular medicine, J Cardiovasc Med (Hagerstown), vol.9, issue.12, pp.1190-221, 2008.

J. M. Houben, H. J. Moonen, F. J. Van-schooten, and G. J. Hageman, Telomere length assessment: biomarker of chronic oxidative stress?, Free Radic Biol Med, vol.44, issue.3, pp.235-281, 2008.

L. S. Honig, M. S. Kang, R. Cheng, J. H. Eckfeldt, B. Thyagarajan et al., Heritability of telomere length in a study of long-lived families, Neurobiol Aging, vol.36, issue.10, pp.2785-90, 2015.

M. L. Hetland, I. J. Christensen, T. Lottenburger, J. S. Johansen, M. N. Svendsen et al., Circulating VEGF as a biological marker in patients with rheumatoid arthritis? Preanalytical and biological variability in healthy persons and in patients, Dis Markers, vol.24, issue.1, pp.1-10, 2008.

S. Toupance, D. Villemonais, D. Germain, A. Gegout-petit, E. Albuisson et al., The individual's signature of telomere length distribution, Sci Rep, vol.9, issue.1, p.685, 2019.
URL : https://hal.archives-ouvertes.fr/hal-01925000

T. Poonpet, N. Saetan, A. Tanavalee, V. Wilairatana, P. Yuktanandana et al., Association between leukocyte telomere length and angiogenic cytokines in knee osteoarthritis, Int J Rheum Dis, vol.21, issue.1, pp.118-143, 2018.

M. N. Chahine, S. Toupance, S. El-hakim, C. Labat, S. Gautier et al., Telomere length and age-dependent telomere attrition: the blood-and-muscle model (1), Can J Physiol Pharmacol, vol.97, issue.4, pp.328-362, 2019.

M. Kimura, R. C. Stone, S. C. Hunt, J. Skurnick, X. Lu et al., Measurement of telomere length by the Southern blot analysis of terminal restriction fragment lengths, Nat Protoc, vol.5, issue.9, pp.1596-607, 2010.

I. Kocsis, B. Gyorffy, E. Nemeth, and B. Vasarhelyi, Examination of Hardy-Weinberg equilibrium in papers of Kidney International: an underused tool, Kidney Int, vol.65, issue.5, pp.1956-1964, 2004.

U. M. Schick, D. Jain, C. J. Hodonsky, J. V. Morrison, J. P. Davis et al., Genome-wide Association Study of Platelet Count Identifies Ancestry-Specific Loci in Hispanic/Latino Americans, Am J Hum Genet, vol.98, issue.2, pp.229-271, 2016.

J. Y. Wong, D. Vivo, I. Lin, X. Fang, S. C. Christiani et al., The relationship between inflammatory biomarkers and telomere length in an occupational prospective cohort study, PLoS One, vol.9, issue.1, p.87348, 2014.

A. D. Blann, F. M. Belgore, J. Constans, C. Conri, and G. Y. Lip, Plasma vascular endothelial growth factor and its receptor Flt-1 in patients with hyperlipidemia and atherosclerosis and the effects of fluvastatin or fenofibrate, The American journal of cardiology, vol.87, issue.10, pp.1160-1163, 2001.

. Blann-ad, . Belgore-fm, C. N. Mccollum, L. Silverman-s, . Pl et al., 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, vol.102, issue.2, pp.187-94, 2002.

H. F. Alber, M. Frick, J. Dulak, J. Dörler, R. H. Zwick et al., Vascular endothelial growth factor (VEGF) plasma concentrations in coronary artery disease, Heart, vol.91, issue.3, pp.365-371, 2005.

S. Visvikis-siest, V. Gorenjak, and M. G. Stathopoulou, Personalised Medicine: The Odyssey from Hope to Practice, J Pers Med, vol.8, issue.4, 2018.

J. H. Souverijn, Multitarget stool DNA testing for colorectal-cancer screening, N Engl J Med, vol.371, issue.2, p.187, 2014.

D. Carling, The AMP-activated protein kinase cascade--a unifying system for energy control, Trends Biochem Sci, vol.29, issue.1, pp.18-24, 2004.

P. Banankhah, G. A. Fishbein, A. Dota, and R. Ardehali, Cardiac manifestations of PRKAG2 mutation, BMC Med Genet, vol.19, issue.1, p.1, 2018.

A. C. Ha, J. M. Renaud, R. A. Dekemp, S. Thorn, J. Dasilva et al., In vivo assessment of myocardial glucose uptake by positron emission tomography in adults with the PRKAG2 cardiac syndrome, Circ Cardiovasc Imaging, vol.2, issue.6, pp.485-91, 2009.

M. Arad, D. W. Benson, A. R. Perez-atayde, W. J. Mckenna, E. A. Sparks et al., Constitutively active AMP kinase mutations cause glycogen storage disease mimicking hypertrophic cardiomyopathy, J Clin Invest, vol.109, issue.3, pp.357-62, 2002.

B. Mao and C. Niehrs, Kremen2 modulates Dickkopf2 activity during Wnt/LRP6 signaling, Gene, vol.302, issue.1-2, pp.179-83, 2003.

Y. Komiya and R. Habas, Wnt signal transduction pathways, Organogenesis, vol.4, issue.2, pp.68-75, 2008.

S. Foulquier, E. P. Daskalopoulos, G. Lluri, K. C. Hermans, A. Deb et al., WNT signaling in cardiac and vascular disease, Pharmacol Rev, vol.70, issue.1, pp.68-141, 2018.

D. Ferrari, G. V. Inestrosa, and N. C. , Wnt signaling function in Alzheimer's disease, Brain Res Rev, vol.33, issue.1, pp.1-12, 2000.

F. Tsai, G. Keller, F. C. Kuo, M. Weiss, J. Chen et al., An early haematopoietic defect in mice lacking the transcription factor GATA-2, Nature, vol.371, issue.6494, p.221, 1994.

N. Liu, D. Ding, W. Hao, F. Yang, X. Wu et al., hTERT promotes tumor angiogenesis by activating VEGF via interactions with the Sp1 transcription factor, Nucleic Acids Res, vol.44, issue.18, pp.8693-703, 2016.

D. Koboldt, Functional Validation of Genomic Discoveries, 2013.

, Epigenome-wide association study in healthy individuals identifies significant associations with DNA methylation and PBMCs extracts VEGF-A concentration

, Sophie Visvikis-Siest ¥, vol.1, p.3

. Université-de-lorraine, . Inserm, F. Ige-pcv, and . Nancy, United Kingdom 3 Department of Internal Medicine and Geriatrics

S. Dr and . Visvikis-siest,

. Inserm-umr-u1122;-ige-pcv,

, E-mail address: sophie.visvikis-siest@inserm.fr REFERENCES 1

K. A. Chester, L. Robson, R. H. Begent, I. C. Talbot, J. H. Pringle et al., Identification of a human ribosomal protein mRNA with increased expression in colorectal tumours, Biochim Biophys Acta, vol.1009, issue.3, pp.297-300, 1989.

Y. Maruyama, T. Miyazaki, K. Ikeda, T. Okumura, W. Sato et al., Short hairpin RNA library-based functional screening identified ribosomal protein L31 that modulates prostate cancer cell growth via p53 pathway, PLoS One, vol.9, issue.10, p.108743, 2014.

V. Senee, C. Chelala, S. Duchatelet, D. Feng, H. Blanc et al.,

D. R. Boileau, P. Cavener, D. Bougneres, C. Taha, and . Julier, Mutations in GLIS3 are responsible for a rare syndrome with neonatal diabetes mellitus and congenital hypothyroidism, vol.38, pp.682-689, 2006.

C. Tomii, M. Inokuchi, Y. Takagi, T. Ishikawa, S. Otsuki et al., TPX2 expression is associated with poor survival in gastric cancer, World J Surg Oncol, vol.15, 2017.

J. Jian, Y. Huang, L. Liu, S. Li, and F. Deng, TPX2 gene-silencing inhibits the proliferation and invasion of human colon cancer SW480 cells, TUMOR, vol.36, issue.6, pp.628-634, 2016.

K. Iwama, M. Sasaki, S. Hirabayashi, C. Ohba, E. Iwabuchi et al.,

S. Miyake, H. Ito, N. Saitsu, and . Matsumoto, Milder progressive cerebellar atrophy caused by biallelic SEPSECS mutations, J Hum Genet, issue.6, pp.527-558, 2016.

A. Sas-chen, M. R. Aure, L. Leibovich, S. Carvalho, Y. Enuka et al.,

S. Schwarz, N. Lavi, Y. Nevo, J. Kuznetsov, F. Yuan et al.,

H. G. Maelandsmo, E. Russnes, T. Schlichting, O. C. Sørlie, K. K. Lingjaerde et al., EMBO Mol Med, vol.8, issue.9, pp.1052-64, 2016.

Y. J. Choi, H. R. Oh, M. R. Choi, M. Gwak, C. H. An et al., Frameshift mutation of a histone methylation-related gene SETD1B and its regional heterogeneity in gastric and colorectal cancers with high microsatellite instability, Hum Pathol, vol.45, issue.8, pp.1674-81, 2014.

B. G. Jang, H. E. Lee, and W. H. Kim, ETV1 mRNA is specifically expressed in gastrointestinal stromal tumors, Virchows Arch, vol.467, issue.4, pp.393-403, 2015.

Y. Gokmen-polar, C. P. Goswami, R. A. Toroni, K. L. Sanders, R. Mehta et al., Gene Expression Analysis Reveals Distinct Pathways of Resistance to Bevacizumab in Xenograft Models of Human ER-Positive Breast Cancer, J Cancer, vol.5, issue.8, pp.633-678, 2014.

B. Fuchs, C. Y. Inwards, and R. Janknecht, Vascular endothelial growth factor expression is upregulated by EWS-ETS oncoproteins and Sp1 and may represent an independent predictor of survival in Ewing's sarcoma, Clin Cancer Res, vol.10, issue.4, pp.1344-53, 2004.

Y. Han, G. Q. Ru, X. Mou, H. J. Wang, Y. Ma et al., AUTS2 is a potential therapeutic target for pancreatic cancer patients with liver metastases, Med Hypotheses, vol.85, issue.2, pp.203-209, 2015.

M. G. Santos, A. Z. Machado, C. N. Martins, S. Domenice, E. M. Costa et al., Homozygous inactivating mutation in NANOS3 in two sisters with primary ovarian insufficiency, Biomed Res Int, vol.787465, 2014.

K. Kusz, L. Tomczyk, A. Spik, A. Latos-bielenska, P. Jedrzejczak et al., NANOS3 gene mutations in men with isolated sterility phenotype, Mol Reprod Dev, vol.76, issue.9, 2009.

S. Grelet, V. Andries, M. Polette, C. Gilles, K. Staes et al.,

C. W. Dalstein, C. Y. Cheng, P. Shen, F. Birembaut, B. Van-roy et al., The human NANOS3 gene contributes to lung tumour invasion by inducing epithelial-mesenchymal transition, J Pathol, vol.237, issue.1, pp.25-37, 2015.

Y. Murakami, N. Kanzawa, K. Saito, P. M. Krawitz, S. Mundlos et al., Mechanism for release of alkaline phosphatase caused by glycosylphosphatidylinositol deficiency in patients with hyperphosphatasia mental retardation syndrome, J Biol Chem, vol.287, issue.9, pp.959-965, 2012.

Z. Ertao, C. Jianhui, C. Chuangqi, Q. Changjiang, C. Sile et al., Low level of FOXL1 indicates a worse prognosis for gastric cancer patients, Tumour Biol, vol.37, issue.8, pp.11331-11338, 2016.

Y. Qin, W. Gong, M. Zhang, J. Wang, Z. Tang et al., Forkhead box L1 is frequently downregulated in gallbladder cancer and inhibits cell growth through apoptosis induction by mitochondrial dysfunction, Int J Clin Exp Pathol, vol.9, issue.7, pp.110-132, 2014.

M. G. Butler, S. L. Dagenais, J. L. Garcia-perez, P. Brouillard, M. Vikkula et al., Microcephaly, intellectual impairment, bilateral vesicoureteral reflux, distichiasis, and glomuvenous malformations associated with a 16q24.3 contiguous gene deletion and a Glomulin mutation, Am J Med Genet A, vol.158, issue.4, pp.3249-3258, 2012.

V. Easwaran, S. H. Lee, L. Inge, L. Guo, C. Goldbeck et al.,

H. Fuller, V. Chan, F. Randazzo, R. Gundel, R. S. Warren et al., Fantl: beta-Catenin regulates vascular endothelial growth factor expression in colon cancer, Cancer Res, vol.63, issue.12, pp.3145-53, 2003.

M. G. Lampugnani, A. Zanetti, M. Corada, T. Takahashi, G. Balconi et al.,

A. Orsenigo, R. Cattelino, T. O. Kemler, E. Daniel, and . Dejana, Contact inhibition of VEGF-induced proliferation requires vascular endothelial cadherin, beta-catenin, and the phosphatase DEP

/. Cd148, J Cell Biol, vol.161, issue.4, pp.793-804, 2003.

M. Orlandini, S. Semboloni, and S. Oliviero, Beta-catenin inversely regulates vascular endothelial growth factor-D mRNA stability, J Biol Chem, vol.278, issue.45, pp.44650-44656, 2003.

M. Ma and N. Yu, Over-Expression of TBL1XR1 Indicates Poor Prognosis of Serous Epithelial Ovarian Cancer, Tohoku J Exp Med, issue.3, pp.239-247, 2017.

N. Kwok, M. Risch, and . Sabbadini, Pierpont syndrome associated with the p.Tyr446Cys missense mutation in TBL1XR1, Eur J Med Genet, vol.60, issue.10, pp.504-508, 2017.

H. Miraoui, A. A. Dwyer, G. P. Sykiotis, L. Plummer, W. Chung et al., Mutations in FGF17, IL17RD, DUSP6, SPRY4, and FLRT3 are identified in individuals with congenital hypogonadotropic hypogonadism, vol.92, pp.725-768, 2013.

L. Wang, E. R. Hauser, S. H. Shah, M. A. Pericak-vance, C. Haynes et al.,

A. B. Nelson, C. B. Hale, J. L. Granger, C. J. Haines, D. Jones et al., Peakwide mapping on chromosome 3q13 identifies the kalirin gene as a novel candidate gene for coronary artery disease, Am J Hum Genet, vol.80, issue.4, pp.650-63, 2007.

G. Wang, W. Zhao, X. Gao, D. Zhang, Y. Li et al., HNF1AAS1 promotes growth and metastasis of esophageal squamous cell carcinoma by sponging miR214 to upregulate the expression of SOX-4, Int J Oncol, vol.51, issue.2, pp.657-667, 2017.

I. Feenstra, L. E. Vissers, R. J. Pennings, W. Nillessen, R. Pfundt et al., Cremers: Disruption of teashirt zinc finger homeobox 1 is associated with congenital aural atresia in humans, Am J Hum Genet, vol.89, issue.6, pp.813-822, 2011.

S. H. Choi, D. Ruggiero, R. Sorice, C. Song, T. Nutile et al.,

E. Vasan, A. L. Ingelsson, A. D. Leutenegger, A. L. Johnson, S. Destefano et al., Six Novel Loci Associated with Circulating VEGF Levels Identified by a Meta-analysis of Genome-Wide Association Studies, PLoS Genet, vol.12, issue.2, pp.1042-1046, 1980.
URL : https://hal.archives-ouvertes.fr/hal-01708531

M. E. Askarian-amiri, V. Seyfoddin, C. E. Smart, J. Wang, J. E. Kim et al., Emerging role of long non-coding RNA SOX2OT in SOX2 regulation in breast cancer, PLoS One, vol.9, issue.7, p.102140, 2014.