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.

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.1628-1654, 2018.

J. Chatelin, M. Stathopoulou, A. Arguinano, A. Xie, and T. Visvikis-siest,

S. *. , Pharmacogenomic challenges in cardiovascular diseases: examples of drugs and considerations for future integration in clinical practice, Curr Pharm Biotechnol

M. Mazidi, P. Rezaie, P. Kengne, A. Stathopoulou, M. G. Azimi-nezhad et al.,

, VEGF, the underlying factor for metabolic syndrome; fact or fiction? Diabetes Metab Syndr, vol.11, pp.61-64, 2017.

S. Visvikis-siest and M. G. Stathopoulou, Beyond genome-wide association studies: identifying variants using -omics approaches, Pers Med, vol.12, issue.6, pp.529-531, 2015.

S. Visvikis-siest, M. G. Stathopoulou, and N. C. Ndiaye, Common mutations and polymorphisms predicting adverse cardiovascular events: current view, Pharmacogenomics, vol.13, issue.16, pp.1875-1878, 2012.

S. Choi, D. Ruggiero, R. Sorice, C. Song, T. Nutile et al., Six novel loci associated with circulating VEGF levels identified by a meta-analysis of genome-wide association studies, PLoS Genet, 2016.
URL : https://hal.archives-ouvertes.fr/hal-01708531

, ):e1005874. * equal first authors, ** equal last authors, 64th American Society of Human Genetics Annual Meeting in, vol.24, p.16, 2014.

E. Grigoriou, G. Trovas, I. Dontas, N. Papaioannou, M. Stathopoulou et al.,

, Serum 25-hydroxyvitamin D levels of healthy adult women in Greece, p.17, 2014.

E. Grigoriou, G. Trovas, I. Dontas, N. Papaioannou, M. Stathopoulou et al.,

, Serum 25-hydroxyvitamin D levels of healthy adult women in Greece

O. Osteoporosis and M. Diseases, , 2014.

M. G. Stathopoulou, P. Monteiro, P. Shahabi, E. Penas-lledo, E. Shamieh et al.,

L. Santos, N. Thilly, G. Siest, A. Llerena, and S. Visvikis-siest, Effects of synergy between clopidogrel and calcium-channel blockers in blood pressure levels, pp.2-19

, ESPT Conference Pharmacogenomics: From Cell to Clinic, 2013.

M. G. Stathopoulou, P. Monteiro, P. Shahabi, E. Penas-lledo, E. Shamieh et al.,

L. Santos, N. Thilly, G. Siest, A. Llerena, and S. Visvikis-siest, Effects of synergy between clopidogrel and calcium-channel blockers in blood pressure levels, pp.2-19

, polymorphisms. 1st public LURIC meeting, p.20, 2013.

M. G. Stathopoulou, P. Monteiro, P. Shahabi, E. Penas-lledo, E. Shamieh et al.,

L. Santos, N. Thilly, G. Siest, A. Llerena, and S. Visvikis-siest, Effects of synergy between clopidogrel and calcium-channel blockers in blood pressure levels, pp.2-19

P. Froguel, N. C. Ndiaye, A. Bonnefond, N. Bouatia-naji, and A. Dechaume,

B. , F. M. Bottolo, L. Guéant-rodriguez, R. M. Lecoeur, C. Langlois et al., Visvikis-Siest S. A genome-wide association study identifies rs2000999 as a strong genetic determinant of Je participe

, Consortium VEGF: Je suis le chef de projet du consortium et membre du comité de pilotage

C. Consortium, Je suis l'analyste principal et le responsable de contact pour l'IGE-PCV

, Groupe mondial collaborant sur les facteurs de risque liés aux maladies chroniques (NCD-RisC): Je suis l'analyste principal et le responsable de contact pour l'IGE-PCV

, U1122. J'ai participé à 6 publications originales. Depuis 2015, je participe aux contrats suivants de l'IGE-PCV, vol.1122

, Traitement avec mastic des obèses en bonne santé avec NAFLD -MAST4HEALTH», financé par la Communauté européenne dans le cadre du projet H2020-MSCA-RISE, 2015.

, Marie Sklodowska actions de recherche et échange de personnel

. «combattre-l'insuffisance-cardiaque, FIGHT-HF», financé par l'ANR dans le cadre des projets «Recherche hospitalo-universitaire en santé, 2015.

«. Projet and . Telarta,

, «Projet GEENAGE (génomique fonctionnelle, épigénomique et environnement pour la gestion du vieillissement saine et pathologique)», financé dans le cadre du projet LUE

, «Développement d'un système de gestion et d'analyse des échantillons biologiques et des données « -omiques » liées au VEGF», financé par le Pôle BMS «Biologie, Médecine, Santé» dans le cadre du projet AAP-BMS interne « Projets incitatifs -Crédits CS-UL, 2018.

, Depuis ma nomination en tant qu'ingénieur de recherche à l'Inserm en, 2015.

, doctorants en obtenant l'Autorisation à Codiriger une Thèse (ACT) de l'école doctorale BioSE

T. Xie, «Interactions épistatiques pour le développement des algorithmes de prédiction des maladies cardiovasculaires: De l'approche gène-candidat au séquençage de prochaine génération

, Jérôme Chatelin: «Interactions gène x gène, gène x environnement et Médecine Prédictive

. Préventive, Personnalisée et Participative (4P) dans l'inflammation

T. Xie, S. Akbar, M. Stathopoulou, T. Oster, C. Masson et al., Visvikis-Siest S. Epistatic interaction of apolipoprotein E and lipolysis-stimulated lipoprotein receptor genetic variants is associated with Alzheimer's disease, Neurobiol Aging, issue.18, pp.30147-30154, 2018.

T. Xie, M. Stathopoulou, S. Akbar, T. Oster, G. Siest et al., Visvikis-Siest S. Effect of LSR polymorphism on blood lipid levels and age-specific epistatic interaction with the APOE common polymorphism, Clin Genet, vol.93, issue.4, pp.846-852, 2018.

M. G. Stathopoulou, T. Xie, D. Ruggiero, J. Chatelin, M. Rancier et al., Visvikis-Siest S -The VEGF Consortium. A transnational collaborative network dedicated to the study and applications of the Vascular Endothelial Growth Factor-A in medical practice: The VEGF Consortium, Clin Chem Lab Med, vol.56, issue.4, pp.83-86, 2018.

T. Xie, M. Stathopoulou, F. De-andrés, G. Siest, H. Murray et al., Visvikis-Siest S. VEGF-related polymorphisms identified by GWAS and risk for major depression. Transl Psychiatry, vol.7, 2017.

J. Chatelin, M. Stathopoulou, A. Arguinano, A. Xie, T. Visvikis-siest et al., Pharmacogenomic challenges in cardiovascular diseases: examples of drugs and considerations for future integration in clinical practice, Curr Pharm Biotechnol, vol.18, issue.3, pp.231-241, 2017.

M. G. Stathopoulou, T. Xie, D. Ruggiero, J. Chatelin, M. Rancier et al., Visvikis-Siest S -The VEGF Consortium. A transnational collaborative network dedicated to the study and applications of the Vascular Endothelial Growth Factor-A in medical practice: The VEGF Consortium, Clin Chem Lab Med, vol.56, issue.4, pp.83-86, 2018.

J. Chatelin, M. Stathopoulou, A. Arguinano, A. Xie, T. Visvikis-siest et al., Pharmacogenomic challenges in cardiovascular diseases: examples of drugs and considerations for future integration in clinical practice, Curr Pharm Biotechnol, vol.18, issue.3, pp.231-241, 2017.

M. Rancier, I. Zaaber, M. G. Stathopoulou, J. Chatelin, A. Saleh et al., Visvikis-Siest S. Pro-and anti-angiogenic VEGF mRNAs in autoimmune thyroid diseases, Autoimmunity, vol.5, pp.1-7, 2016.

V. Gorenjak, Outils biostatistiques /bioinformatiques pour une recherche de médecine personnalisée : télomères, facteur de croissance de l'endothélium vasculaire (VEGF) et molécules associées ». The defense is planned for the end of 2019 I will participate in the evaluation committee

, This thesis aims to study the telomere length and the VEGF as biomarkers for personalised medicine. Modern approaches will be used like mendelian randomisation and the study of

A. C. Kaliora, M. G. Stathopoulou, J. K. Triantafillidis, G. V. Dedoussis, and N. K. Andrikopoulos,

, Alterations in the function of circulating mononuclear cells derived from patients with crohn's disease treated with mastic, World journal of gastroenterology, vol.13, pp.6031-6036, 2007.

A. C. Kaliora, M. G. Stathopoulou, J. K. Triantafillidis, G. V. Dedoussis, and N. K. Andrikopoulos,

, Chios mastic treatment of patients with active crohn's disease. World journal of gastroenterology, vol.13, pp.748-753, 2007.

M. Skouroliakou, I. Giannopoulou, C. Kostara, K. Koutri, M. G. Stathopoulou et al., Effects of a nutritional intervention in obese postmenopausal women on atypical antipsychotics, Maturitas, vol.67, pp.166-170, 2010.

M. Skouroliakou, C. Kakavelaki, K. Diamantopoulos, M. Stathopoulou, E. Vourvouhaki et al., The development and implementation of a software tool and its effect on the quality of provided clinical nutritional therapy in hospitalized patients, Journal of the American Medical Informatics Association : JAMIA, vol.16, pp.802-805, 2009.

M. Skouroliakou, O. Kastanidou, M. Stathopoulou, and G. Vourli, Evaluation of the antioxidant effect of a new functional food enriched with sideritis euboea in healthy subjects, Journal of medicinal food, vol.12, pp.1105-1110, 2009.

M. Skouroliakou, D. Konstantinou, K. Koutri, C. Kakavelaki, M. Stathopoulou et al., A double-blind, randomized clinical trial of the effect of omega-3 fatty acids on the oxidative stress of preterm neonates fed through parenteral nutrition, European journal of clinical nutrition, vol.64, pp.940-947, 2010.

M. Skouroliakou, K. Koutri, M. Stathopoulou, E. Vourvouhaki, I. Giannopoulou et al., Comparison of two types of tpn prescription methods in preterm neonates, Pharmacy world & science : PWS, vol.31, pp.202-208, 2009.

M. Skouroliakou, M. Stathopoulou, A. Koulouri, I. Giannopoulou, D. Stamatiades et al., Determinants of resting energy expenditure in hemodialysis patients, and comparison with healthy subjects, Journal of renal nutrition : the official journal of the Council on Renal Nutrition of the National Kidney Foundation, vol.19, pp.283-290, 2009.

, Osteoporosis prevention, diagnosis, and therapy, NIH Consens Statement, vol.17, pp.1-45, 2000.

. Iof, Osteoporosis facts and statistics, 2018.

S. Ferrari, Human genetics of osteoporosis, Best Pract Res Clin Endocrinol Metab, vol.22, pp.723-735, 2008.

W. F. Li, S. X. Hou, B. Yu, M. M. Li, C. Ferec et al., Genetics of osteoporosis: Accelerating pace in gene identification and validation, Hum Genet, vol.127, pp.249-285, 2010.

D. Karasik, F. Rivadeneira, and M. L. Johnson, The genetics of bone mass and susceptibility to bone diseases, Nature reviews. Rheumatology, vol.12, pp.323-334, 2016.

K. Zhu and R. L. Prince, Calcium and bone, Clin Biochem, vol.45, pp.936-942, 2012.

I. Charopoulos, G. Trovas, M. Stathopoulou, P. Kyriazopoulos, A. Galanos et al., Lack of association between vitamin d and calcitonin receptor gene polymorphisms and forearm bone values of young greek males, Journal of musculoskeletal & neuronal interactions, vol.8, pp.196-203, 2008.

M. G. Stathopoulou, G. V. Dedoussis, G. Trovas, A. Katsalira, N. Hammond et al., Low-density lipoprotein receptor-related protein 5 polymorphisms are associated with bone mineral density in greek postmenopausal women: An interaction with calcium intake, Journal of the American Dietetic Association, vol.110, pp.1078-1083, 2010.

M. G. Stathopoulou, G. V. Dedoussis, G. Trovas, E. V. Theodoraki, A. Katsalira et al., The role of vitamin d receptor gene polymorphisms in the bone mineral density of greek postmenopausal women with low calcium intake, The Journal of nutritional biochemistry, vol.22, pp.752-757, 2011.

L. Stolk, J. R. Perry, D. I. Chasman, C. He, M. Mangino et al., Nature genetics, vol.44, pp.260-268, 2012.

N. Ferrara, Vegf-a: A critical regulator of blood vessel growth, Eur Cytokine Netw, vol.20, pp.158-163, 2009.

J. A. Nagy, A. M. Dvorak, and H. F. Dvorak, Vegf-a and the induction of pathological angiogenesis, Annu Rev Pathol, vol.2, pp.251-275, 2007.

A. Amini, S. M. Moghaddam, D. L. Morris, and M. H. Pourgholami, The critical role of vascular endothelial growth factor in tumor angiogenesis, Curr Cancer Drug Targets, vol.12, pp.23-43, 2012.

P. W. Holm, R. H. Slart, C. J. Zeebregts, J. L. Hillebrands, and R. A. Tio, Atherosclerotic plaque development and instability: A dual role for vegf, Ann Med, vol.41, pp.257-264, 2009.

P. R. Moreno, M. Purushothaman, and K. R. Purushothaman, Plaque neovascularization: Defense mechanisms, betrayal, or a war in progress, Ann N Y Acad Sci, vol.1254, pp.7-17, 2012.

P. Carmeliet and R. K. Jain, Molecular mechanisms and clinical applications of angiogenesis, Nature, vol.473, pp.298-307, 2011.

M. R. Ladomery, S. J. Harper, and D. O. Bates, Alternative splicing in angiogenesis: The vascular endothelial growth factor paradigm, Cancer Lett, vol.249, pp.133-142, 2007.

M. Medinger, N. Fischer, and A. Tzankov, Vascular endothelial growth factor-related pathways in hemato-lymphoid malignancies, J Oncol, vol.2010, p.729725, 2010.

F. L. Celletti, J. M. Waugh, P. G. Amabile, A. Brendolan, P. R. Hilfiker et al., Vascular endothelial growth factor enhances atherosclerotic plaque progression, Nat Med, vol.7, pp.425-429, 2001.

M. Inoue, H. Itoh, M. Ueda, T. Naruko, A. Kojima et al., Vascular endothelial growth factor (vegf) expression in human coronary atherosclerotic lesions: Possible pathophysiological significance of vegf in progression of atherosclerosis, Circulation, vol.98, pp.2108-2116, 1998.

R. Khurana, Z. Zhuang, S. Bhardwaj, M. Murakami, D. Muinck et al., Angiogenesis-dependent and independent phases of intimal hyperplasia, Circulation, vol.110, pp.2436-2443, 2004.

K. B. Lemstrom, R. Krebs, A. I. Nykanen, J. M. Tikkanen, R. K. Sihvola et al., Vascular endothelial growth factor enhances cardiac allograft arteriosclerosis, Circulation, vol.105, pp.2524-2530, 2002.

K. S. Moulton, K. Vakili, D. Zurakowski, M. Soliman, C. Butterfield et al., Inhibition of plaque neovascularization reduces macrophage accumulation and progression of advanced atherosclerosis, Proc Natl Acad Sci U S A, vol.100, pp.4736-4741, 2003.

J. Rutanen, P. Leppanen, T. T. Tuomisto, T. T. Rissanen, M. O. Hiltunen et al.,

, Vascular endothelial growth factor-d expression in human atherosclerotic lesions, Cardiovasc Res, vol.59, pp.971-979, 2003.

M. Laitinen, Z. I. Breier, G. Pakkanen, T. Hakkinen, T. Luoma et al., Vegf gene transfer reduces intimal thickening via increased production of nitric oxide in carotid arteries, Hum Gene Ther, vol.8, pp.1737-1744, 1997.

P. Leppanen, S. Koota, I. Kholova, J. Koponen, C. Fieber et al., Gene transfers of vascular endothelial growth factor-a, vascular endothelial growth factor-b, vascular endothelial growth factor-c, and vascular endothelial growth factor-d have no effects on atherosclerosis in hypercholesterolemic low-density lipoprotein-receptor/apolipoprotein b48-deficient mice, Circulation, vol.112, pp.1347-1352, 2005.

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 anti-angiogenic isoforms: A novel therapeutic strategy for angiogenesis, J Biol Chem, vol.285, pp.5532-5540, 2010.

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, pp.572-592, 2009.

S. J. Harper and D. O. Bates, Vegf-a splicing: The key to anti-angiogenic therapeutics?, Nat Rev Cancer, vol.8, pp.880-887, 2008.

Y. S. Ng, D. Krilleke, and D. T. Shima, Vegf function in vascular pathogenesis, Exp Cell Res, vol.312, pp.527-537, 2006.

H. Berrahmoune, B. Herbeth, J. V. Lamont, C. Masson, and P. S. Fitzgerald, Visvikis-Siest S. Heritability for plasma vegf concentration in the stanislas family study, Ann Hum Genet, vol.71, pp.54-63, 2007.

H. Berrahmoune, J. V. Lamont, B. Herbeth, and P. S. Fitzgerald, Visvikis-Siest S. Biological determinants of and reference values for plasma interleukin-8, monocyte chemoattractant protein-1, epidermal growth factor, and vascular endothelial growth factor: Results from the stanislas cohort, Clin Chem, vol.52, pp.504-510, 2006.

M. Azimi-nezhad, . Ld, C. Ottone, C. Perrin, C. Chapel et al., Influence of pre-analytical variables on vefga gene expression and circulating protein concentrations, Biopreservation and Biobanking, vol.10, pp.454-461, 2012.

S. Debette, S. Visvikis-siest, M. H. Chen, N. C. Ndiaye, C. Song et al.,

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

M. G. Stathopoulou, A. Bonnefond, N. C. Ndiaye, M. Azimi-nezhad, E. Shamieh et al., Visvikis-Siest S. A common variant highly associated with plasma vegfa levels also contributes to the variation of both ldlc and hdl-c, J Lipid Res, vol.54, pp.535-541, 2013.

M. Azimi-nezhad, M. G. Stathopoulou, A. Bonnefond, M. Rancier, A. Saleh et al., Visvikis-Siest S. Associations of vascular endothelial growth factor (vegf) with adhesion and inflammation molecules in a healthy population, Cytokine, vol.61, pp.602-607, 2013.

A. Bonnefond, P. J. Saulnier, M. G. Stathopoulou, N. Grarup, N. C. Ndiaye et al., What is the contribution of two genetic variants regulating vegf levels to type 2 diabetes risk and to microvascular complications?, PLoS One, vol.8, p.55921, 2013.
URL : https://hal.archives-ouvertes.fr/hal-01708333

C. M. Lawes, S. Vander-hoorn, and A. Rodgers, Global burden of blood-pressure-related disease, Lancet, vol.371, pp.1513-1518, 2001.

G. B. Ehret, P. B. Munroe, K. M. Rice, M. Bochud, A. D. Johnson et al., Nature, vol.478, pp.103-109, 2011.

S. El-shamieh, N. C. Ndiaye, M. G. Stathopoulou, H. A. Murray, C. Masson et al.,

P. Fitzgerald and A. Benetos, Visvikis-Siest S. Functional epistatic interaction between rs6046g>a in f7 and rs5355c>t in sele modifies systolic blood pressure levels, PLoS One, vol.7, p.40777, 2012.

N. C. Ndiaye, S. Said-el, M. G. Stathopoulou, G. Siest, M. Y. Tsai et al., Epistatic study reveals two genetic interactions in blood pressure regulation, BMC medical genetics, vol.14, issue.2, 2013.
URL : https://hal.archives-ouvertes.fr/hal-01708271

P. Froguel, N. C. Ndiaye, A. Bonnefond, N. Bouatia-naji, A. Dechaume et al., Visvikis-Siest S. A genome-wide association study identifies rs2000999 as a strong genetic determinant of circulating haptoglobin levels, PLoS One, vol.7, p.32327, 2012.

M. Armanios, Telomeres and age-related disease: How telomere biology informs clinical paradigms, J Clin Invest, vol.123, pp.996-1002, 2013.

B. B. De-jesus and M. A. Blasco, Assessing cell and organ senescence biomarkers, Circ Res, vol.111, pp.97-109, 2012.

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

D. Meyer, T. Rietzschel, E. R. , D. Buyzere, M. L. Van-criekinge et al., Telomere length and cardiovascular aging: The means to the ends?, Ageing Res Rev, vol.10, pp.297-303, 2011.

V. Codd, M. Mangino, P. Van-der-harst, P. S. Braund, M. Kaiser et al.,

J. Moore, C. Nelson, N. Soranzo, G. Zhai, and A. M. Valdes,

R. A. Boer, M. Kimura, A. Aviv, A. H. Goodall, W. Ouwehand et al.,

W. H. Gilst, G. Navis, P. R. Burton, M. D. Tobin, A. S. Hall et al., Common variants near terc are associated with mean telomere length, Nature genetics, vol.42, pp.197-199, 2010.

V. Codd, C. P. Nelson, A. E. Mangino, M. Deelen, J. Buxton et al., Nature genetics, vol.45, pp.427-421, 2013.

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.514-521, 2011.

D. Levy, S. L. Neuhausen, S. C. Hunt, M. Kimura, S. J. Hwang et al., Genomewide association identifies obfc1 as a locus involved in human leukocyte telomere biology, Proc Natl Acad Sci U S A, vol.107, pp.9293-9298, 2010.

M. Mangino, S. J. 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.5385-5394, 2012.

M. Mangino, J. B. Richards, N. Soranzo, G. Zhai, A. Aviv et al.,

P. Deloukas and T. D. Spector, A genome-wide association study identifies a novel locus on chromosome 18q12.2 influencing white cell telomere length, J Med Genet, vol.46, pp.451-454, 2009.
URL : https://hal.archives-ouvertes.fr/hal-00552680

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. 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. Paediatric and perinatal epidemiology, vol.29, pp.146-150, 2015.

S. W. Brouilette, J. S. Moore, A. D. Mcmahon, J. R. Thompson, I. Ford et al., Telomere length, risk of coronary heart disease, and statin treatment in the west of scotland primary prevention study: A nested case-control study, Lancet, vol.369, pp.107-114, 2007.

A. Aviv, A. M. Valdes, and T. D. Spector, Human telomere biology: Pitfalls of moving from the laboratory to epidemiology, International journal of epidemiology, vol.35, pp.1424-1429, 2006.

S. Bekaert, D. Meyer, T. Rietzschel, E. R. , D. Buyzere et al., Van Oostveldt P. Telomere length and cardiovascular risk factors in a middle-aged population free of overt cardiovascular disease, Aging Cell, vol.6, pp.639-647, 2007.

L. Wang, H. L. Mcleod, and R. M. Weinshilboum, Genomics and drug response, N Engl J Med, vol.364, pp.1144-1153, 2011.

E. C. Hayden, Cardiovascular disease gets personal, Nature, vol.460, pp.940-941, 2009.

I. Postmus, J. J. Verschuren, A. J. De-craen, P. E. Slagboom, R. G. Westendorp et al., Pharmacogenetics of statins: Achievements, whole-genome analyses and future perspectives, Pharmacogenomics, vol.13, pp.831-840, 2012.

D. Voora and G. S. Ginsburg, Clinical application of cardiovascular pharmacogenetics, J Am Coll Cardiol, vol.60, pp.9-20, 2012.

G. W. Dorn, Decoding the cardiac message: The 2011 thomas w. Smith memorial lecture, Circ Res, vol.110, pp.755-763, 2012.

M. G. Stathopoulou, P. Monteiro, P. Shahabi, E. Penas-lledo, E. Shamieh et al., Visvikis-Siest S. Newly identified synergy between clopidogrel and calcium-channel blockers for blood pressure regulation possibly involves cyp2c19 rs4244285, Int J Cardiol, vol.168, pp.3057-3058, 2013.

P. Shahabi, T. Cuisset, M. G. Stathopoulou, P. E. Morange, C. Grosdidier et al.,

G. , A. Mc, and S. Visvikis-siest, Genetic determined low response to thienopyridines is associated with higher systemic inflammation in smokers, Pharmacogenomics, vol.16, pp.459-469, 2015.

B. Herbeth, A. Samara, M. Stathopoulou, and G. Siest, Visvikis-Siest S. Alcohol consumption, beverage preference, and diet in middle-aged men from the stanislas study, Journal of nutrition and metabolism, vol.2012, p.987243, 2012.

N. C. Ndiaye, A. Nehzad, M. , E. Shamieh, S. Stathopoulou et al., Visvikis-Siest S. Cardiovascular diseases and genome-wide association studies, Clin Chim Acta, vol.412, pp.1697-1701, 2011.

S. Visvikis-siest, M. G. Stathopoulou, and N. C. Ndiaye, Common mutations and polymorphisms predicting adverse cardiovascular events: Current view, Pharmacogenomics, vol.13, pp.1875-1878, 2012.

A. Saleh, M. G. Stathopoulou, S. Dade, N. C. Ndiaye, M. Azimi-nezhad et al.,

C. Masson, J. Lamont, P. Fitzgerald, and S. Visvikis-siest, Angiogenesis related genes nos3, cd14, mmp3 and il4r are associated to vegf gene expression and circulating levels in healthy adults, BMC medical genetics, vol.16, p.90, 2015.
URL : https://hal.archives-ouvertes.fr/inserm-01264504

M. Rancier, I. Zaaber, M. G. Stathopoulou, J. Chatelin, A. Saleh et al., Visvikis-Siest S. Pro-and anti-angiogenic vegf mrnas in autoimmune thyroid diseases, Autoimmunity, vol.49, pp.366-372, 2016.

I. Zaaber, M. Rancier, M. G. Stathopoulou, A. Saleh, H. Marmouch et al., Visvikis-Siest S. Plasma vegf-related polymorphisms are implied in autoimmune thyroid diseases, vol.49, pp.229-235, 2016.

S. H. Choi, D. Ruggiero, R. Sorice, C. Song, T. Nutile et al., Six novel loci associated with circulating vegf levels identified by a meta-analysis of genome-wide association studies, PLoS genetics, vol.12, p.1005874, 2016.
URL : https://hal.archives-ouvertes.fr/hal-01708531

T. Xie, M. G. Stathopoulou, F. De-andres, G. Siest, H. Murray et al., Visvikis-Siest S. Vegf-related polymorphisms identified by gwas and risk for major depression, Translational psychiatry, vol.7, p.1055, 2017.

M. Azimi-nezhad, S. R. Mirhafez, M. G. Stathopoulou, H. Murray, N. C. Ndiaye et al., The relationship between vascular endothelial growth factor cis-and trans-acting genetic variants and metabolic syndrome. The American journal of the medical sciences, vol.355, pp.559-565, 2018.

M. Mazidi, P. Rezaie, A. P. Kengne, M. G. Stathopoulou, M. Azimi-nezhad et al., Vegf, the underlying factor for metabolic syndrome; fact or fiction? Diabetes & metabolic syndrome, vol.11, pp.61-64, 2017.

M. G. Stathopoulou, T. Xie, D. Ruggiero, J. Chatelin, M. Rancier et al., Visvikis-Siest S. A transnational collaborative network dedicated to the study and applications of the vascular endothelial growth factor-a in medical practice: The vegf consortium, Clinical genetics, vol.56, pp.83-86, 2018.

L. Kopin, C. Lowenstein, and . Dyslipidemia, Annals of internal medicine, vol.167, pp.81-96, 2017.

R. K. Upadhyay, Emerging risk biomarkers in cardiovascular diseases and disorders, Institute. NHGR. Gwas catalog, pp.971453-96, 2015.

R. W. Mahley and S. C. Rall, Type iii hyperlipoproteinemia (dysbetalipoproteinemia): The role of apolipoprotein e in normal and abnormal lipoprotein metabolism, The metabolic and molecular bases of inherited disease, pp.2835-2562, 2001.

D. Salah, K. Bohnet, R. Gueguen, G. Siest, and S. Visvikis, Combined effects of lipoprotein lipase and apolipoprotein e polymorphisms on lipid and lipoprotein levels in the stanislas cohort, Journal of lipid research, vol.38, pp.904-912, 1997.

C. Pallaud, R. Gueguen, C. Sass, M. Grow, S. Cheng et al., Genetic influences on lipid metabolism trait variability within the stanislas cohort, Journal of lipid research, vol.42, pp.1879-1890, 2001.

E. Boerwinkle, S. Visvikis, D. Welsh, J. Steinmetz, S. M. Hanash et al., The use of measured genotype information in the analysis of quantitative phenotypes in man. Ii. The role of the apolipoprotein e polymorphism in determining levels, variability, and covariability of cholesterol, betalipoprotein, and triglycerides in a sample of unrelated individuals, American journal of medical genetics, vol.27, pp.567-582, 1987.

N. Haddy, D. Bacquer, D. Chemaly, M. M. Maurice, M. Ehnholm et al., The importance of plasma apolipoprotein e concentration in addition to its common polymorphism on interindividual variation in lipid levels: Results from apo europe, European journal of human genetics : EJHG, vol.10, pp.841-850, 2002.
URL : https://hal.archives-ouvertes.fr/hal-00176672

D. Evans, F. U. Beil, and J. Aberle, Resequencing the apoe gene reveals that rare mutations are not significant contributory factors in the development of type iii hyperlipidemia, Journal of clinical lipidology, vol.7, pp.671-674, 2013.

P. N. Hopkins, E. A. Brinton, and M. N. Nanjee, Hyperlipoproteinemia type 3: The forgotten phenotype, Current atherosclerosis reports, vol.16, p.440, 2014.

C. Reitz, Dyslipidemia and the risk of alzheimer's disease, Current atherosclerosis reports, vol.15, p.307, 2013.

N. E. Shepardson, G. M. Shankar, and D. J. Selkoe, Cholesterol level and statin use in alzheimer disease: Ii. Review of human trials and recommendations, Archives of neurology, vol.68, pp.1385-1392, 2011.

L. Mahoney-sanchez, A. A. Belaidi, A. I. Bush, and S. Ayton, The complex role of apolipoprotein e in alzheimer's disease: An overview and update, vol.60, pp.325-335, 2016.

C. Lane-donovan, W. M. Wong, and M. S. Durakoglugil, Genetic restoration of plasma apoe improves cognition and partially restores synaptic defects in apoe-deficient mice, Journal of molecular neuroscience : MN, vol.36, pp.10141-10150, 2016.

F. T. Yen, O. Roitel, L. Bonnard, V. Notet, D. Pratte et al., Lipolysis stimulated lipoprotein receptor: A novel molecular link between hyperlipidemia, weight gain, and atherosclerosis in mice, The Journal of biological chemistry, vol.283, pp.25650-25659, 2008.

C. Stenger, C. Corbier, and F. T. Yen, Structure and function of the lipolysis stimulated lipoprotein receptor in chemical biology, pp.267-292, 2012.

P. Narvekar, B. Diaz, M. Krones-herzig, A. Hardeland, U. Strzoda et al., Liver-specific loss of lipolysis-stimulated lipoprotein receptor triggers systemic hyperlipidemia in mice, Diabetes, vol.58, pp.1040-1049, 2009.

B. E. Bihain and F. T. Yen, Free fatty acids activate a high-affinity saturable pathway for degradation of low-density lipoproteins in fibroblasts from a subject homozygous for familial hypercholesterolemia, Biochemistry, vol.31, pp.4628-4636, 1992.

F. T. Yen, C. J. Mann, L. M. Guermani, N. F. Hannouche, N. Hubert et al., Identification of a lipolysis-stimulated receptor that is distinct from the ldl receptor and the ldl receptor-related protein, Biochemistry, vol.33, pp.1172-1180, 1994.

C. Stenger, A. Pincon, M. Hanse, L. Royer, A. Comte et al., Brain region-specific immunolocalization of the lipolysis-stimulated lipoprotein receptor (lsr) and altered cholesterol distribution in aged lsr+/-mice, Journal of neurochemistry, vol.123, pp.467-476, 2012.

A. Pincon, M. H. Thomas, M. Huguet, A. Allouche, J. C. Colin et al., Increased susceptibility of dyslipidemic lsr+/-mice to amyloid stress is associated with changes in cortical cholesterol levels, Journal of Alzheimer's disease : JAD, vol.45, pp.195-204, 2015.
URL : https://hal.archives-ouvertes.fr/hal-01575780

R. Daneman, L. Zhou, D. Agalliu, J. D. Cahoy, A. Kaushal et al., The mouse bloodbrain barrier transcriptome: A new resource for understanding the development and function of brain endothelial cells, PLoS One, vol.5, p.13741, 2010.

F. Sohet, C. Lin, R. N. Munji, S. Y. Lee, N. Ruderisch et al., Lsr/angulin-1 is a tricellular tight junction protein involved in blood-brain barrier formation, The Journal of cell biology, vol.208, pp.703-711, 2015.
URL : https://hal.archives-ouvertes.fr/hal-01575779

G. L. Bowman, J. A. Kaye, and J. F. Quinn, Dyslipidemia and blood-brain barrier integrity in alzheimer's disease. Current gerontology and geriatrics research, vol.2012, p.184042, 2012.

T. S. Salameh, E. M. Rhea, W. A. Banks, and A. J. Hanson, Insulin resistance, dyslipidemia, and apolipoprotein e interactions as mechanisms in cognitive impairment and alzheimer's disease. Experimental biology and medicine (Maywood, N.J.), vol.241, pp.1676-1683, 2016.

M. Brulliard, V. Ogier, L. Mejean, L. Bonnard, B. Bihain et al., A polymorphism of the lipolysis-stimulated lipoprotein receptor gene is associated with elevated plasma triglycerides and ldl cholesterol in obese subjects, ATVB, vol.28, issue.6, p.136, 2008.

T. Xie, M. G. Stathopoulou, S. Akbar, T. Oster, G. Siest et al., Effect of lsr polymorphism on blood lipid levels and age-specific epistatic interaction with the apoe common polymorphism, vol.93, pp.846-852, 2018.
URL : https://hal.archives-ouvertes.fr/hal-01908961

T. Xie, S. Akbar, M. G. Stathopoulou, T. Oster, C. Masson et al., Epistatic interaction of apolipoprotein e and lipolysis-stimulated lipoprotein receptor genetic variants is associated with alzheimer's disease, Neurobiology of aging, vol.69, pp.292-291, 2018.

A. Bouchon, J. Dietrich, and M. Colonna, Cutting edge: Inflammatory responses can be triggered by trem-1, a novel receptor expressed on neutrophils and monocytes, J Immunol, vol.164, pp.4991-4995, 2000.

A. Bouchon, F. Facchetti, M. A. Weigand, and M. Colonna, Trem-1 amplifies inflammation and is a crucial mediator of septic shock, Nature, vol.410, pp.1103-1107, 2001.

M. Colonna and F. Facchetti, Trem-1 (triggering receptor expressed on myeloid cells): A new player in acute inflammatory responses, J Infect Dis, vol.187, issue.2, pp.397-401, 2003.

J. W. Ford and D. W. Mcvicar, Trem and trem-like receptors in inflammation and disease, Curr Opin Immunol, vol.21, pp.38-46, 2009.

J. Kuai, B. Gregory, A. Hill, D. D. Pittman, J. L. Feldman et al., Trem-1 expression is increased in the synovium of rheumatoid arthritis patients and induces the expression of pro-inflammatory cytokines, Rheumatology (Oxford), vol.48, pp.1352-1358, 2009.

J. Joffre, S. Potteaux, L. Zeboudj, X. Loyer, A. Boufenzer et al., Ait-Oufella H. Genetic and pharmacological inhibition of trem-1 limits the development of experimental atherosclerosis, J Am Coll Cardiol, vol.68, pp.2776-2793, 2016.

L. Jeremie, B. Amir, D. Marc, and G. Sebastien, The triggering receptor expressed on myeloid cells-1: A new player during acute myocardial infarction, Pharmacol Res, vol.100, pp.261-265, 2015.

J. F. Dopheide, C. Doppler, M. Scheer, V. Obst, M. C. Radmacher et al., Critical limb ischaemia is characterised by an increased production of whole blood reactive oxygen species and expression of trem-1 on neutrophils, Atherosclerosis, vol.229, pp.396-403, 2013.

R. P. Mcever, Selectins: Initiators of leucocyte adhesion and signalling at the vascular wall, Cardiovasc Res, vol.107, pp.331-339, 2015.

R. L. Zemans, S. P. Colgan, and G. P. Downey, Transepithelial migration of neutrophils: Mechanisms and implications for acute lung injury, Am J Respir Cell Mol Biol, vol.40, pp.519-535, 2009.

W. A. Muller, The regulation of transendothelial migration: New knowledge and new questions, Cardiovasc Res, vol.107, pp.310-320, 2015.

A. Arguinano, A. A. Dade, S. Stathopoulou, M. Derive, M. et al., Trem-1 snp rs2234246 regulates trem-1 protein and mrna levels and is associated with plasma levels of l-selectin, vol.12, p.182226, 2017.

L. L. Lanier, Dap10-and dap12-associated receptors in innate immunity, Immunol Rev, vol.227, pp.150-160, 2009.

H. Ito and J. A. Hamerman, Trem-2, triggering receptor expressed on myeloid cell-2, negatively regulates tlr responses in dendritic cells, Eur J Immunol, vol.42, pp.176-185, 2012.

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

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, Mol Med Rep, vol.7, pp.921-926, 2013.

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, Mol Cell Biochem, vol.394, pp.155-161, 2014.

V. Gorenjak, A. Arguinano, A. A. Dade, S. Stathopoulou, M. G. Vance et al., The polymorphism rs6918289 located in the downstream region of the trem2 gene is associated with tnf-alpha levels and imt-f, Scientific reports, vol.8, p.7160, 2018.

V. Gorenjak, S. Akbar, and M. G. Stathopoulou, Visvikis-Siest S. The future of telomere length in personalized medicine, Frontiers in bioscience, vol.23, pp.1628-1654, 2018.

P. M. Ridker, J. E. Buring, N. Rifai, and N. R. Cook, Development and validation of improved algorithms for the assessment of global cardiovascular risk in women: The reynolds risk score, JAMA, vol.297, pp.611-619, 2007.

A. A. Arguinano, E. Naderi, N. C. Ndiaye, M. Stathopoulou, S. Dade et al., Visvikis-Siest S. Il6r haplotype rs4845625*t/rs4537545*c is a risk factor for simultaneously high crp, ldl and apob levels, Genes and immunity, vol.18, pp.163-169, 2017.

S. Ligthart, A. Vaez, U. Vosa, M. G. Stathopoulou, P. S. De-vries et al.,

D. Hernandez, C. Tiesler, V. Giedraitis, D. Liewald, K. Fischer et al.,

, Am J Hum Genet, vol.103, pp.691-706, 2018.

P. B. Balagopal, S. D. De-ferranti, S. Cook, S. R. Daniels, S. S. Gidding et al., Nontraditional risk factors and biomarkers for cardiovascular disease: Mechanistic, research, and clinical considerations for youth: A scientific statement from the american heart association, Circulation, vol.123, pp.2749-2769, 2011.

D. G. Cook, M. A. Mendall, P. H. Whincup, I. M. Carey, L. Ballam et al., C-reactive protein concentration in children: Relationship to adiposity and other cardiovascular risk factors, Atherosclerosis, vol.149, pp.139-150, 2000.

E. S. Ford, C-reactive protein concentration and cardiovascular disease risk factors in children: Findings from the national health and nutrition examination survey, Circulation, vol.108, pp.1053-1058, 1999.

A. W. Zieske, R. P. Tracy, C. A. Mcmahan, E. E. Herderick, S. Homma et al., Elevated serum c-reactive protein levels and advanced atherosclerosis in youth, Arterioscler Thromb Vasc Biol, vol.25, pp.1237-1243, 2005.

M. Juonala, J. S. Viikari, T. Ronnemaa, L. Taittonen, J. Marniemi et al., Childhood c-reactive protein in predicting crp and carotid intima-media thickness in adulthood: The cardiovascular risk in young finns study, Arterioscler Thromb Vasc Biol, vol.26, pp.1883-1888, 2006.

L. K. Heilbronn and L. V. Campbell, Adipose tissue macrophages, low grade inflammation and insulin resistance in human obesity, Curr Pharm Des, vol.14, pp.1225-1230, 2008.

M. Kong and C. Lee, Genetic associations with c-reactive protein level and white blood cell count in the kare study, Int J Immunogenet, vol.40, pp.120-125, 2013.

Y. Wu, T. W. Mcdade, C. W. Kuzawa, J. Borja, Y. Li et al.,

, Genome-wide association with c-reactive protein levels in clhns: Evidence for the crp and hnf1a loci and their interaction with exposure to a pathogenic environment, Inflammation, vol.35, pp.574-583, 2012.

F. Tekola-ayele, A. Doumatey, H. Huang, J. Zhou, B. Charles et al., Genome-wide associated loci influencing interleukin (il)-10, il-1ra, and il-6 levels in african americans, Immunogenetics, vol.64, pp.351-359, 2012.

A. P. Reiner, S. Beleza, N. Franceschini, P. L. Auer, J. G. Robinson et al., Genome-wide association and population genetic analysis of c-reactive protein in african american and hispanic american women, Am J Hum Genet, vol.91, pp.502-512, 2012.

S. Naitza, E. Porcu, M. Steri, D. D. Taub, A. Mulas et al., A genome-wide association scan on the levels of markers of inflammation in sardinians reveals associations that underpin its complex regulation, PLoS genetics, vol.8, p.1002480, 2012.

A. P. Doumatey, G. Chen, T. Ayele, F. Zhou, J. Erdos et al., C-reactive protein (crp) promoter polymorphisms influence circulating crp levels in a genome-wide association study of african americans, Hum Mol Genet, vol.21, pp.3063-3072, 2012.

S. Aslibekyan, E. K. Kabagambe, M. R. Irvin, R. J. Straka, I. B. Borecki et al., A genome-wide association study of inflammatory biomarker changes in response to fenofibrate treatment in the genetics of lipid lowering drug and diet network, Pharmacogenet Genomics, vol.22, pp.191-197, 2012.

Y. Okada, A. Takahashi, H. Ohmiya, N. Kumasaka, Y. Kamatani et al., Genomewide association study for c-reactive protein levels identified pleiotropic associations in the il6 locus, Hum Mol Genet, vol.20, pp.1224-1231, 2011.

R. P. Middelberg, M. A. Ferreira, A. K. Henders, A. C. Heath, P. A. Madden et al., Genetic variants in lpl, oasl and tomm40/apoe-c1-c2-c4 genes are associated with multiple cardiovascular-related traits, BMC medical genetics, vol.12, p.123, 2011.

A. Dehghan, J. Dupuis, M. Barbalic, J. C. Bis, G. Eiriksdottir et al., Circulation, vol.123, pp.731-738, 2011.

P. Elliott, J. C. Chambers, W. Zhang, R. Clarke, J. C. Hopewell et al., Genetic loci associated with c-reactive protein levels and risk of coronary heart disease, JAMA, vol.302, pp.37-48, 2009.

M. S. Cunnington, B. M. Mayosi, D. H. Hall, P. J. Avery, M. Farrall et al., Novel genetic variants linked to coronary artery disease by genome-wide association are not associated with carotid artery intima-media thickness or intermediate risk phenotypes, Atherosclerosis, vol.203, pp.41-44, 2009.

P. M. Ridker, G. Pare, A. Parker, R. Y. Zee, J. S. Danik et al., Loci related to metabolic-syndrome pathways including lepr,hnf1a, il6r, and gckr associate with plasma c-reactive protein: The women's genome health study, Am J Hum Genet, vol.82, pp.1185-1192, 2008.

A. P. Reiner, M. J. Barber, Y. Guan, P. M. Ridker, L. A. Lange et al., Polymorphisms of the hnf1a gene encoding hepatocyte nuclear factor-1 alpha are associated with c-reactive protein, Am J Hum Genet, vol.82, pp.1193-1201, 2008.

D. Melzer, J. R. Perry, D. Hernandez, A. M. Corsi, K. Stevens et al., PLoS genetics, vol.4, p.1000072, 2008.

E. J. Benjamin, J. Dupuis, M. G. Larson, K. L. Lunetta, S. L. Booth et al., Genome-wide association with select biomarker traits in the framingham heart study, BMC medical genetics, vol.8, issue.1, p.11, 2007.

A. E. Locke, B. Kahali, S. I. Berndt, A. E. Justice, T. H. Pers et al.,

J. Shi, V. Smith, A. Smolonska, J. Stanton, A. V. Steinthorsdottir et al.,

Q. Zhang, E. P. Brennan, M. Choi, Z. Dastani, A. W. Drong et al., Genetic studies of body mass index yield new insights for obesity biology, vol.518, pp.197-206, 2015.

D. Shungin, T. W. Winkler, D. C. Croteau-chonka, T. Ferreira, A. E. Locke et al.,

P. Lichtner, L. Lind, J. Lindstrom, S. Lobbens, M. Lorentzon et al.,

M. , E. R. Eriksson, J. G. Farrall, M. Ferrannini, E. Ferrieres et al.,

C. M. Duijn, C. J. Willer, P. M. Visscher, J. Yang, J. N. Hirschhorn et al., New genetic loci link adipose and insulin biology to body fat distribution, Nature, vol.518, pp.187-196, 2015.
URL : https://hal.archives-ouvertes.fr/hal-01132692

P. K. Joshi, T. Esko, H. Mattsson, N. Eklund, I. Gandin et al.,

P. Froguel, G. Pasterkamp, S. F. Grant, H. Hakonarson, L. Ferrucci et al., Nature, vol.523, pp.459-462, 2015.

, A century of trends in adult human height. eLife, vol.5, 2016.

, Worldwide trends in diabetes since 1980: A pooled analysis of 751 population-based studies with 4.4 million participants, Lancet, vol.387, pp.1513-1530, 2016.

, Worldwide trends in body-mass index, underweight, overweight, and obesity from 1975 to 2016: A pooled analysis of 2416 population-based measurement studies in 128.9 million children, adolescents, and adults, Lancet, vol.390, pp.2627-2642, 2017.

, Worldwide trends in blood pressure from 1975 to 2015: A pooled analysis of 1479 population-based measurement studies with 19.1 million participants, Lancet, vol.389, pp.37-55, 2017.

, Trends in adult body-mass index in 200 countries from 1975 to 2014: A pooled analysis of 1698 population-based measurement studies with 19.2 million participants, Lancet, vol.387, pp.1377-1396, 2016.

, Contributions of mean and shape of blood pressure distribution to worldwide trends and variations in raised blood pressure: A pooled analysis of 1018 population-based measurement studies with 88.6 million participants, International journal of epidemiology, 2018.

J. Chatelin, M. G. Stathopoulou, A. A. Arguinano, T. Xie, and S. Visvikis-siest, Pharmacogenomic challenges in cardiovascular diseases: Examples of drugs and considerations for future integration in clinical practice, Curr Pharm Biotechnol, vol.18, pp.231-241, 2017.

S. Visvikis-siest, V. Gorenjak, and M. G. Stathopoulou, Personalised medicine: The odyssey from hope to practice, Journal of personalized medicine, vol.8, 2018.

S. Visvikis-siest and M. G. Stathopoulou, Beyond genome-wide association studies: Identifying variants using -omics approaches, Pers Med, vol.12, pp.529-531, 2015.

, European Heart Network. European cardiovascular disease statistics, 2017.

M. Presta, G. Andres, D. Leali, P. Dell'era, and R. Ronca, Inflammatory cells and chemokines sustain fgf2-induced angiogenesis, Eur Cytokine Netw, vol.20, pp.39-50, 2009.

D. Ribatti, F. Levi-schaffer, and P. T. Kovanen, Inflammatory angiogenesis in atherogenesis--a double-edged sword, Ann Med, vol.40, pp.606-621, 2008.

I. Zachary and R. D. Morgan, Therapeutic angiogenesis for cardiovascular disease: Biological context, challenges, prospects, Heart, vol.97, pp.181-189, 2011.

J. Luo, Y. Xiong, X. Han, and Y. Lu, Vegf non-angiogenic functions in adult organ homeostasis: Therapeutic implications, J Mol Med (Berl), vol.89, pp.635-645, 2011.

, Neurodegenerative disease research in europe gets it together, 2014.

, Internationa Osteoporosis Foundation. Osteoporosis: Facts and statistics, 2018.

S. Koch and L. Claesson-welsh, Signal transduction by vascular endothelial growth factor receptors. Cold Spring Harbor perspectives in medicine, vol.2, p.6502, 2012.

Z. G. Zhang, L. Zhang, Q. Jiang, R. Zhang, K. Davies et al., Vegf enhances angiogenesis and promotes blood-brain barrier leakage in the ischemic brain, J Clin Invest, vol.106, pp.829-838, 2000.

T. Inai, M. Mancuso, H. Hashizume, F. Baffert, A. Haskell et al., Inhibition of vascular endothelial growth factor (vegf) signaling in cancer causes loss of endothelial fenestrations, regression of tumor vessels, and appearance of basement membrane ghosts, The American journal of pathology, vol.165, pp.35-52, 2004.

J. Provias and B. Jeynes, Reduction in vascular endothelial growth factor expression in the superior temporal, hippocampal, and brainstem regions in alzheimer's disease, Current neurovascular research, vol.11, pp.202-209, 2014.

J. B. Zhang, M. F. Li, H. X. Zhang, Z. G. Li, H. R. Sun et al., Association of serum vascular endothelial growth factor levels and cerebral microbleeds in patients with alzheimer's disease, European journal of neurology, vol.23, pp.1337-1342, 2016.

I. Mateo, J. Llorca, J. Infante, E. Rodriguez-rodriguez, C. Fernandez-viadero et al., Low serum vegf levels are associated with alzheimer's disease, Acta neurologica Scandinavica, vol.116, pp.56-58, 2007.

A. Chakraborty, M. Chatterjee, H. Twaalfhoven, D. C. Milan, M. Teunissen et al., Vascular endothelial growth factor remains unchanged in cerebrospinal fluid of patients with alzheimer's disease and vascular dementia, vol.10, p.58, 2018.

E. Tarkowski, R. Issa, M. Sjogren, A. Wallin, K. Blennow et al., Increased intrathecal levels of the angiogenic factors vegf and tgf-beta in alzheimer's disease and vascular dementia, Neurobiology of aging, vol.23, pp.237-243, 2002.

L. Thirumangalakudi, P. G. Samany, A. Owoso, B. Wiskar, and P. Grammas, Angiogenic proteins are expressed by brain blood vessels in alzheimer's disease, Journal of Alzheimer's disease : JAD, vol.10, pp.111-118, 2006.

S. Y. Liu, F. F. Zeng, Z. W. Chen, C. Y. Wang, B. Zhao et al., Vascular endothelial growth factor gene promoter polymorphisms and alzheimer's disease risk: A meta-analysis, CNS neuroscience & therapeutics, vol.19, pp.469-476, 2013.

D. Bo, R. Ghezzi, S. Scarpini, E. Bresolin, N. Comi et al., Vegf genetic variability is associated with increased risk of developing alzheimer's disease, Journal of the neurological sciences, vol.283, pp.66-68, 2009.

D. He, W. Lu, K. Chang, Y. Liu, J. Zhang et al., Vascular endothelial growth factor polymorphisms and risk of alzheimer's disease: A meta-analysis, Gene, vol.518, pp.296-302, 2013.

M. Grellier, P. Granja, J. Fricain, S. Bidarra, R. M. Bareille et al., The effect of the co-immobilization of human osteoprogenitors and endothelial cells within alginate microspheres on mineralization in a bone defect, Biomaterials, vol.30, pp.3271-3278, 2009.

J. Trueta and A. Buhr, The vascular contribution to osteogenesis. V. The vasculature supplying the epiphysial cartilage in rachitic rats, Journal of Bone and Joint Surgery British, vol.45, pp.572-581, 1963.

M. Brandi, C. , and P. , Vascular biology of the skeleton, Journal of Bone and Mineral Research, vol.21, pp.183-192, 2006.

I. Kim, S. Moon, S. Kim, H. Kim, Y. Koy et al., Vascular endothelial growth factor expression of intercellular adhesion molecule 1 (icam-1), vascular cell adhesion molecule 1 (vcam-1), and e-selectin through nuclear factor-kappa b activation in endothelial cells, J Biol Chem, vol.276, pp.7614-7620, 2001.

V. Midy and J. Plouët, Vasculotropin/vascular endothelial growth factor induces differentiation in cultured osteoblasts, Biochem Biophys Res Commun, vol.199, pp.380-386, 1994.

U. Mayr-wohlfart, J. Waltenberger, H. Hausser, S. Kessler, K. Günther et al., Vascular endothelial growth factor stimulates chemotactic migration of primary human osteoblasts, Bone, vol.30, pp.472-477, 2002.

Y. Tan, Y. Yang, L. Chai, R. Wong, and A. Rabie, Effects of vascular endothelial growth factor (vegf] on mc3t3-e1, Orthod Craniofac Res, vol.13, pp.223-228, 2010.

W. Lieb, R. Safa, E. J. Benjamin, V. Xanthakis, X. Yin et al., Vascular endothelial growth factor, its soluble receptor, and hepatocyte growth factor: Clinical and genetic correlates and association with vascular function, Eur Heart J, vol.30, pp.1121-1127, 2009.

D. Ruggiero, C. Dalmasso, T. Nutile, R. Sorice, L. Dionisi et al., Genetics of vegf serum variation in human isolated populations of cilento: Importance of vegf polymorphisms, PLoS One, vol.6, p.16982, 2011.

R. M. Conroy, K. Pyorala, A. P. Fitzgerald, S. Sans, A. Menotti et al., Estimation of ten-year risk of fatal cardiovascular disease in europe: The score project, Eur Heart J, vol.24, pp.987-1003, 2003.

R. B. D'agostino, S. Vasan, R. S. Pencina, M. J. Wolf, P. A. Cobain et al., General cardiovascular risk profile for use in primary care: The framingham heart study, Circulation, vol.117, pp.743-753, 2008.

J. A. Kanis, E. V. Mccloskey, H. Johansson, A. Oden, O. Strom et al., Osteoporosis international : a journal established as result of cooperation between the European Foundation for Osteoporosis and the National Osteoporosis Foundation of the USA, vol.21, pp.407-413, 2010.

M. T. Cooney, A. Dudina, D. 'agostino, R. Graham, and I. M. , Cardiovascular risk-estimation systems in primary prevention: Do they differ? Do they make a difference? Can we see the future?, Circulation, vol.122, pp.300-310, 2010.

M. J. Pencina, D. 'agostino, R. B. Vasan, and R. S. , Statistical methods for assessment of added usefulness of new biomarkers. Clinical chemistry and laboratory medicine, vol.48, pp.1703-1711, 2010.

E. W. Steyerberg and Y. Vergouwe, Towards better clinical prediction models: Seven steps for development and an abcd for validation, Eur Heart J, 2014.