Y. Hojo, U. Ikeda, Y. Zhu, M. Okada, and S. Ueno, Expression of vascular endothelial growth factor in patients with acute myocardial infarction, Journal of the American College of Cardiology, vol.35, issue.4, pp.968-973, 2000.
DOI : 10.1016/S0735-1097(99)00632-4

R. Matsuo, T. Ago, M. Kamouchi, J. Kuroda, and T. Kuwashiro, Clinical significance of plasma VEGF value in ischemic stroke - research for biomarkers in ischemic stroke (REBIOS) study, BMC Neurology, vol.12, issue.2, pp.32-32, 2013.
DOI : 10.1016/S1050-1738(01)00149-9

M. Slevin, J. Krupinski, A. Slowik, P. Kumar, and A. Szczudlik, Serial Measurement of Vascular Endothelial Growth Factor and Transforming Growth Factor-??1 in Serum of Patients With Acute Ischemic Stroke, Stroke, vol.31, issue.8, pp.1863-1870, 2000.
DOI : 10.1161/01.STR.31.8.1863

A. Karayiannakis, K. Syrigos, A. Polychronidis, A. Zbar, and G. Kouraklis, Circulating VEGF Levels in the Serum of Gastric Cancer Patients, Annals of Surgery, vol.236, issue.1, pp.37-42, 2002.
DOI : 10.1097/00000658-200207000-00007

I. Hyodo, T. Doi, H. Endo, Y. Hosokawa, and Y. Nishikawa, Clinical significance of plasma vascular endothelial growth factor in gastrointestinal cancer, European Journal of Cancer, vol.34, issue.13, pp.2041-2045, 1998.
DOI : 10.1016/S0959-8049(98)00282-2

I. Kishiro, S. Kato, D. Fuse, T. Yoshida, and S. Machida, Clinical significance of vascular endothelial growth factor in patients with primary lung cancer, Respirology, vol.74, issue.2, pp.93-98, 2002.
DOI : 10.1002/1097-0142(19941015)74:8<2245::AID-CNCR2820740807>3.0.CO;2-X

H. Li, P. Kantoff, J. Ma, M. Stampfer, and D. George, Prediagnostic Plasma Vascular Endothelial Growth Factor Levels and Risk of Prostate Cancer, Cancer Epidemiology Biomarkers & Prevention, vol.14, issue.6, pp.1557-1561, 2005.
DOI : 10.1158/1055-9965.EPI-04-0456

B. Wirostko, T. Wong, and R. Simo, Vascular endothelial growth factor and diabetic complications, Progress in Retinal and Eye Research, vol.27, issue.6, pp.608-621, 2008.
DOI : 10.1016/j.preteyeres.2008.09.002

N. Ferrara, G. Frantz, J. Lecouter, L. Dillard-telm, and T. Pham, Differential Expression of the Angiogenic Factor Genes Vascular Endothelial Growth Factor (VEGF) and Endocrine Gland-Derived VEGF in Normal and Polycystic Human Ovaries, The American Journal of Pathology, vol.162, issue.6, pp.1881-1893, 2003.
DOI : 10.1016/S0002-9440(10)64322-2

P. Peitsidis and R. Agrawal, Role of vascular endothelial growth factor in women with PCO and PCOS: a systematic review, Reproductive BioMedicine Online, vol.20, issue.4, pp.444-452, 2010.
DOI : 10.1016/j.rbmo.2010.01.007

G. Smith and H. Wear, The perinatal implications of angiogenic factors, Current Opinion in Obstetrics and Gynecology, vol.21, issue.2, pp.111-116, 2009.
DOI : 10.1097/GCO.0b013e328328cf7d

B. Oosthuyse, L. Moons, E. Storkebaum, H. Beck, and D. Nuyens, Deletion of the hypoxia-response element in the vascular endothelial growth factor promoter causes motor neuron degeneration, Nature Genetics, vol.807, issue.2, pp.131-138, 2001.
DOI : 10.1016/S0006-8993(98)00569-1

B. Sopher, P. Thomas, M. Lafevre-bernt, I. Holm, and S. Wilke, Androgen Receptor YAC Transgenic Mice Recapitulate SBMA Motor Neuronopathy and Implicate VEGF164 in the Motor Neuron Degeneration, Neuron, vol.41, issue.5, pp.687-699, 2004.
DOI : 10.1016/S0896-6273(04)00082-0

W. Lieb, R. Safa, E. Benjamin, V. Xanthakis, and X. Yin, Vascular endothelial growth factor, its soluble receptor, and hepatocyte growth factor: clinical and genetic correlates and association with vascular function, European Heart Journal, vol.109, issue.9, pp.1121-1127, 2009.
DOI : 10.1042/CS20040307

URL : https://academic.oup.com/eurheartj/article-pdf/30/9/1121/17353285/ehp007.pdf

I. Pantsulaia, S. Trofimov, E. Kobyliansky, and G. Livshits, Heritability of circulating growth factors involved in the angiogenesis in healthy human population, Cytokine, vol.27, issue.6, pp.152-158, 2004.
DOI : 10.1016/j.cyto.2004.04.005

H. Berrahmoune, B. Herbeth, J. Lamont, C. Masson, and P. Fitzgerald, Heritability for Plasma VEGF Concentration in the Stanislas Family Study, Annals of Human Genetics, vol.266, issue.1, pp.54-63, 2007.
DOI : 10.1161/hc0902.104718

D. Ruggiero, C. Dalmasso, T. Nutile, R. Sorice, and L. Dionisi, Genetics of VEGF Serum Variation in Human Isolated Populations of Cilento: Importance of VEGF Polymorphisms, PLoS ONE, vol.42, issue.2, p.21347390, 2011.
DOI : 10.1371/journal.pone.0016982.s004

J. Rosenstein, N. Mani, A. Khaibullina, and J. Krum, Neurotrophic effects of vascular endothelial growth factor on organotypic cortical explants and primary cortical neurons, J Neurosci, vol.23, pp.11036-11044, 2003.

J. Rosenstein and J. Krum, New roles for VEGF in nervous tissue?beyond blood vessels, Experimental Neurology, vol.187, issue.2, pp.246-253, 2004.
DOI : 10.1016/j.expneurol.2004.01.022

N. Costa, S. Paramanathan, M. Donald, D. Wierzbicki, A. Hampson et al., Factors regulating circulating vascular endothelial growth factor (VEGF): Association with bone mineral density (BMD) in post-menopausal osteoporosis, Cytokine, vol.46, issue.3, pp.376-381, 2009.
DOI : 10.1016/j.cyto.2009.03.012

A. Medford, S. Godinho, L. Keen, J. Bidwell, and A. Millar, Relationship Between Vascular Endothelial Growth Factor + 936 Genotype and Plasma/Epithelial Lining Fluid Vascular Endothelial Growth Factor Protein Levels in Patients With and at Risk for ARDS, Chest, vol.136, issue.2, pp.457-464, 2009.
DOI : 10.1378/chest.09-0383

U. Langsenlehner, G. Wolf, T. Langsenlehner, A. Gerger, and G. Hofmann, Genetic polymorphisms in the vascular endothelial growth factor gene and breast cancer risk. The Austrian ???tumor of breast tissue: incidence, genetics, and environmental risk factors??? study, Breast Cancer Research and Treatment, vol.103, issue.2, pp.297-304, 2008.
DOI : 10.1007/s10549-007-9655-z

I. Mateo, J. Llorca, J. Infante, E. Rodriguez-rodriguez, and C. Fernandez-viadero, Low serum VEGF levels are associated with Alzheimer's disease, Acta Neurologica Scandinavica, vol.31, issue.1, pp.56-58, 2007.
DOI : 10.1016/S0197-4580(03)00111-8

S. Balasubramanian, A. Cox, S. Cross, S. Higham, and N. Brown, Influence of VEGF-A gene variation and protein levels in breast cancer susceptibility and severity, International Journal of Cancer, vol.92, issue.5, pp.1009-1016, 2007.
DOI : 10.1177/002215540405200408

K. Steffensen, M. Waldstrom, I. Brandslund, and A. Jakobsen, The relationship of VEGF polymorphisms with serum VEGF levels and progression-free survival in patients with epithelial ovarian cancer, Gynecologic Oncology, vol.117, issue.1, 2010.
DOI : 10.1016/j.ygyno.2009.11.011

R. Zhai, M. Gong, W. Zhou, T. Thompson, and P. Kraft, Genotypes and haplotypes of the VEGF gene are associated with higher mortality and lower VEGF plasma levels in patients with ARDS, Thorax, vol.62, issue.8, pp.718-722, 2007.
DOI : 10.1136/thx.2006.069393

M. Ferrante, M. Pierik, L. Henckaerts, M. Joossens, and K. Claes, The role of vascular endothelial growth factor (VEGF) in inflammatory bowel disease, Inflammatory Bowel Diseases, vol.46, issue.9, pp.870-878, 2006.
DOI : 10.1093/rheumatology/kei013

P. Krippl, U. Langsenlehner, W. Renner, B. Yazdani-biuki, and G. Wolf, A common 936 C/T gene polymorphism of vascular endothelial growth factor is associated with decreased breast cancer risk, International Journal of Cancer, vol.22, issue.4, pp.468-471, 2003.
DOI : 10.1002/ijc.11238

T. Awata, K. Inoue, S. Kurihara, T. Ohkubo, and M. Watanabe, A Common Polymorphism in the 5'-Untranslated Region of the VEGF Gene Is Associated With Diabetic Retinopathy in Type 2 Diabetes, Diabetes, vol.51, issue.5, pp.1635-1639, 2002.
DOI : 10.2337/diabetes.51.5.1635

M. Petrovic, P. Korosec, M. Kosnik, J. Osredkar, and M. Hawlina, Local and genetic determinants of vascular endothelial growth factor expression in advanced proliferative diabetic retinopathy, Mol Vis, vol.14, pp.1382-1387, 2008.

W. Renner, S. Kotschan, C. Hoffmann, B. Obermayer-pietsch, and E. Pilger, A Common 936 C/T Mutation in the Gene for Vascular Endothelial Growth Factor Is Associated with Vascular Endothelial Growth Factor Plasma Levels, Journal of Vascular Research, vol.37, issue.6, pp.443-448, 2000.
DOI : 10.1159/000054076

S. Debette, S. Visvikis-siest, M. Chen, N. Ndiaye, and C. Song, Identification of cis- and trans-Acting Genetic Variants Explaining Up to Half the Variation in Circulating Vascular Endothelial Growth Factor Levels, Circulation Research, vol.109, issue.5, pp.554-563, 2011.
DOI : 10.1161/CIRCRESAHA.111.243790

H. Verheul, K. Hoekman, S. Luykx-de-bakker, C. Eekman, and C. Folman, Platelet: transporter of vascular endothelial growth factor, Clin Cancer Res, vol.3, pp.2187-2190, 1997.

N. Webb, M. Bottomley, C. Watson, and P. Brenchley, Vascular Endothelial Growth Factor (VEGF) is Released from Platelets during Blood Clotting: Implications for Measurement of Circulating VEGF Levels in Clinical Disease, Clinical Science, vol.94, issue.4, pp.395-404, 1998.
DOI : 10.1042/cs0940395

L. Ward and M. Kellis, HaploReg: a resource for exploring chromatin states, conservation, and regulatory motif alterations within sets of genetically linked variants, Nucleic Acids Research, vol.41, issue.11, pp.930-934, 2012.
DOI : 10.1038/ng.472

A. Segre, L. Groop, V. Mootha, M. Daly, and D. Altshuler, Common Inherited Variation in Mitochondrial Genes Is Not Enriched for Associations with Type 2 Diabetes or Related Glycemic Traits, PLoS Genetics, vol.102, issue.8, 2010.
DOI : 10.1371/journal.pgen.1001058.s024

E. Speliotes, C. Willer, S. Berndt, K. Monda, and G. Thorleifsson, Association analyses of 249,796 individuals reveal 18 new loci associated with body mass index, Nature Genetics, vol.74, issue.11, pp.937-948, 2010.
DOI : 10.1016/j.ajhg.2010.02.020

D. Shungin, T. Winkler, D. Croteau-chonka, T. Ferreira, and A. Locke, New genetic loci link adipose and insulin biology to body fat distribution, Nature, vol.9, issue.7538, pp.187-196, 2015.
DOI : 10.1038/nmeth.2212

URL : https://hal.archives-ouvertes.fr/hal-01132692

C. Gieger, A. Radhakrishnan, A. Cvejic, W. Tang, and E. Porcu, New gene functions in megakaryopoiesis and platelet formation, Nature, vol.2, issue.7376, pp.201-208, 2011.
DOI : 10.1038/nprot.2007.324

P. Van-der-harst, W. Zhang, M. Leach, I. Rendon, A. Verweij et al., Seventy-five genetic loci influencing the human red blood cell, Nature, vol.141, issue.7429, pp.369-375, 2012.
DOI : 10.1042/0264-6021:3590099

H. Engels, E. Wohlleber, A. Zink, J. Hoyer, and K. Ludwig, A novel microdeletion syndrome involving 5q14.3-q15: clinical and molecular cytogenetic characterization of three patients, European Journal of Human Genetics, vol.77, issue.12, pp.1592-1599, 2009.
DOI : 10.1086/379793

C. Cardoso, A. Boys, E. Parrini, C. Mignon-ravix, and J. Mcmahon, Periventricular heterotopia, mental retardation, and epilepsy associated with 5q14.3-q15 deletion, Neurology, vol.72, issue.9, pp.784-792, 2009.
DOI : 10.1212/01.wnl.0000336339.08878.2d

URL : https://hal.archives-ouvertes.fr/inserm-00483473

M. Ikram, X. Sim, S. Xueling, R. Jensen, and M. Cotch, Four Novel Loci (19q13, 6q24, 12q24, and 5q14) Influence the Microcirculation In Vivo, PLoS Genetics, vol.32, issue.10, 2010.
DOI : 10.1371/journal.pgen.1001184.s003

D. Maiti, Z. Xu, and E. Duh, Vascular Endothelial Growth Factor Induces MEF2C and MEF2-Dependent Activity in Endothelial Cells, Investigative Opthalmology & Visual Science, vol.49, issue.8, pp.3640-3648, 2008.
DOI : 10.1167/iovs.08-1760

URL : http://iovs.arvojournals.org/data/journals/iovs/932954/z7g00808003640.pdf

S. Watanabe, K. Watanabe, V. Akimov, J. Bartkova, and B. Blagoev, JMJD1C demethylates MDC1 to regulate the RNF8 and BRCA1???mediated chromatin response to DNA breaks, Nature Structural & Molecular Biology, vol.20, issue.12, pp.1425-1433, 2013.
DOI : 10.1111/j.1474-9726.2006.00231.x

J. Lee, H. Choi, J. Gyuris, R. Brent, and D. Moore, Two classes of proteins dependent on either the presence or absence of thyroid hormone for interaction with the thyroid hormone receptor, Mol Endocrinol, vol.9, pp.243-254, 1995.

E. Speliotes, J. Butler, C. Palmer, B. Voight, and J. Hirschhorn, variants specifically confer increased risk for histologic nonalcoholic fatty liver disease but not metabolic disease, Hepatology, vol.28, issue.3, pp.904-912, 2010.
DOI : 10.1055/s-0028-1091981

URL : http://onlinelibrary.wiley.com/doi/10.1002/hep.23768/pdf

J. Chambers, W. Zhang, J. Sehmi, X. Li, and M. Wass, Genome-wide association study identifies loci influencing concentrations of liver enzymes in plasma, Nature Genetics, vol.87, issue.11, pp.1131-1138, 2011.
DOI : 10.1039/c0mb00066c

E. Taniguchi, S. Sakisaka, K. Matsuo, K. Tanikawa, and M. Sata, Expression and Role of Vascular Endothelial Growth Factor in Liver Regeneration After Partial Hepatectomy in Rats, Journal of Histochemistry & Cytochemistry, vol.9, issue.1, pp.121-130, 2001.
DOI : 10.1002/hep.1840180622

R. Qayyum, B. Snively, E. Ziv, M. Nalls, and Y. Liu, A Meta-Analysis and Genome-Wide Association Study of Platelet Count and Mean Platelet Volume in African Americans, PLoS Genetics, vol.34, issue.3, 2012.
DOI : 10.1371/journal.pgen.1002491.s014

A. Johnson, L. Yanek, M. Chen, N. Faraday, and M. Larson, Genome-wide meta-analyses identifies seven loci associated with platelet aggregation in response to agonists, Nature Genetics, vol.26, issue.7, pp.608-613, 2010.
DOI : 10.1172/JCI22973

G. Jin, J. Sun, S. Kim, J. Feng, and Z. Wang, Genome-wide association study identifies a new locus JMJD1C at 10q21 that may influence serum androgen levels in men, Human Molecular Genetics, vol.8, issue.23, pp.5222-5228, 2012.
DOI : 10.1371/journal.pgen.1002805

A. Coviello, R. Haring, M. Wellons, D. Vaidya, and T. Lehtimaki, A Genome-Wide Association Meta-Analysis of Circulating Sex Hormone???Binding Globulin Reveals Multiple Loci Implicated in Sex Steroid Hormone Regulation, PLoS Genetics, vol.24, issue.7, 2012.
DOI : 10.1371/journal.pgen.1002805.s004

R. Stewart, D. Panigrahy, E. Flynn, and J. Folkman, VASCULAR ENDOTHELIAL GROWTH FACTOR EXPRESSION AND TUMOR ANGIOGENESIS ARE REGULATED BY ANDROGENS IN HORMONE RESPONSIVE HUMAN PROSTATE CARCINOMA: : EVIDENCE FOR ANDROGEN DEPENDENT DESTABILIZATION OF VASCULAR ENDOTHELIAL GROWTH FACTOR TRANSCRIPTS, The Journal of Urology, vol.165, issue.2, pp.688-693, 2001.
DOI : 10.1097/00005392-200102000-00095

P. Mark, B. Radlinski, N. Core, A. Fryer, and E. Kirk, Narrowing the Critical Region for Congenital Vertical Talus in Patients With Interstitial 18q Deletions, American Journal of Medical Genetics Part A, vol.3, issue.5, pp.1117-1121, 2013.
DOI : 10.1002/ajmg.1320030207

B. Schick, S. Wemmert, V. Willnecker, J. Dlugaiczyk, and P. Nicolai, Genome-wide copy number profiling using a 100K SNP array reveals novel disease-related genes BORIS and TSHZ1 in juvenile angiofibroma, International Journal of Oncology, vol.39, pp.1143-1151, 2011.
DOI : 10.3892/ijo.2011.1166

J. Brieger, M. Wierzbicka, M. Sokolov, Y. Roth, and W. Szyfter, Vessel Density, Proliferation, and Immunolocalization of Vascular Endothelial Growth Factor in Juvenile Nasopharyngeal Angiofibromas, Archives of Otolaryngology???Head & Neck Surgery, vol.130, issue.6, pp.727-731, 2004.
DOI : 10.1001/archotol.130.6.727

K. Freson, C. Thys, C. Wittewrongel, J. Vermylen, and M. Hoylaerts, Molecular cloning and characterization of the GATA1 cofactor human FOG1 and assessment of its binding to GATA1 proteins carrying D218 substitutions, Human Genetics, vol.112, issue.1, pp.42-49, 2003.
DOI : 10.1007/s00439-002-0832-1

K. Nichols, J. Crispino, M. Poncz, J. White, and S. Orkin, Familial dyserythropoietic anaemia and thrombocytopenia due to an inherited mutation in GATA1, Nature Genetics, vol.17, issue.3, pp.266-270, 2000.
DOI : 10.1128/MCB.17.3.1642

R. Baker and P. Board, Unequal crossover generates variation in ubiquitin coding unit number at the human UbC polyubiquitin locus, Am J Hum Genet, vol.44, pp.534-542, 1989.

J. Satoh and Y. Kuroda, Ubiquitin C-terminal hydrolase-L1 (PGP9.5) expression in human neural cell lines following induction of neuronal differentiation and exposure to cytokines, neurotrophic factors or heat stress, Neuropathology and Applied Neurobiology, vol.69, issue.2, pp.95-9104, 2001.
DOI : 10.1146/annurev.nutr.15.1.161

S. Debette, I. Verbaas, C. Bressler, J. Schuur, M. Smith et al., Genome-wide Studies of Verbal Declarative Memory in Nondemented Older People: The Cohorts for Heart and Aging Research in Genomic Epidemiology Consortium, Biological Psychiatry, vol.77, issue.8, pp.749-763, 2015.
DOI : 10.1016/j.biopsych.2014.08.027

M. Hayashi, C. Fearns, B. Eliceiri, Y. Yang, and J. Lee, Big Mitogen-Activated Protein Kinase 1/Extracellular Signal-Regulated Kinase 5 Signaling Pathway Is Essential for Tumor-Associated Angiogenesis, Cancer Research, vol.65, issue.17, pp.7699-7706, 2005.
DOI : 10.1158/0008-5472.CAN-04-4540

URL : http://cancerres.aacrjournals.org/content/canres/65/17/7699.full.pdf

X. Wang and C. Tournier, Regulation of cellular functions by the ERK5 signalling pathway, Cellular Signalling, vol.18, issue.6, pp.753-760, 2006.
DOI : 10.1016/j.cellsig.2005.11.003

S. Sohn, B. Sarvis, D. Cado, and A. Winoto, ERK5 MAPK Regulates Embryonic Angiogenesis and Acts as a Hypoxia-sensitive Repressor of Vascular Endothelial Growth Factor Expression, Journal of Biological Chemistry, vol.280, issue.45, pp.43344-43351, 2002.
DOI : 10.1073/pnas.142293999

X. Pi, G. Garin, L. Xie, Q. Zheng, and H. Wei, BMK1/ERK5 Is a Novel Regulator of Angiogenesis by Destabilizing Hypoxia Inducible Factor 1??, Circulation Research, vol.96, issue.11, pp.1145-1151, 2005.
DOI : 10.1161/01.RES.0000168802.43528.e1

S. Cameron, S. Ture, D. Mickelsen, E. Chakrabarti, and K. Modjeski, Platelet Extracellular Regulated Protein Kinase 5 Is a Redox Switch and Triggers Maladaptive Platelet Responses and Myocardial Infarct ExpansionCLINICAL PERSPECTIVES, Circulation, vol.132, issue.1, pp.47-58, 2015.
DOI : 10.1161/CIRCULATIONAHA.115.015656

A. Walz, R. Burgener, B. Car, M. Baggiolini, and S. Kunkel, Structure and neutrophil-activating properties of a novel inflammatory peptide (ENA-78) with homology to interleukin 8, Journal of Experimental Medicine, vol.174, issue.6, pp.1355-1362, 1991.
DOI : 10.1084/jem.174.6.1355

A. Koch, S. Kunkel, L. Harlow, D. Mazarakis, and G. Haines, Epithelial neutrophil activating peptide-78: a novel chemotactic cytokine for neutrophils in arthritis., Journal of Clinical Investigation, vol.94, issue.3, pp.1012-1018, 1994.
DOI : 10.1172/JCI117414

D. Arenberg, M. Keane, B. Digiovine, S. Kunkel, and S. Morris, Epithelial-neutrophil activating peptide (ENA-78) is an important angiogenic factor in non-small cell lung cancer., Journal of Clinical Investigation, vol.102, issue.3, pp.465-472, 1998.
DOI : 10.1172/JCI3145

P. Kuo, Y. Chen, T. Chen, K. Shen, and Y. Hsu, CXCL5/ENA78 increased cell migration and epithelial-to-mesenchymal transition of hormone-independent prostate cancer by early growth response-1/snail signaling pathway, Journal of Cellular Physiology, vol.66, issue.5, pp.1224-1231, 2011.
DOI : 10.1002/pros.20488

M. Kawamura, Y. Toiyama, K. Tanaka, S. Saigusa, and Y. Okugawa, CXCL5, a promoter of cell proliferation, migration and invasion, is a novel serum prognostic marker in patients with colorectal cancer, European Journal of Cancer, vol.48, issue.14, pp.2244-2251, 2012.
DOI : 10.1016/j.ejca.2011.11.032

L. Begley, S. Kasina, R. Mehra, S. Adsule, and A. Admon, CXCL5 Promotes Prostate Cancer Progression, Neoplasia, vol.10, issue.3, pp.244-254, 2008.
DOI : 10.1593/neo.07976

URL : https://doi.org/10.1593/neo.07976

J. Park, K. Park, S. Bang, M. Kim, and J. Lee, CXCL5 overexpression is associated with late stage gastric cancer, Journal of Cancer Research and Clinical Oncology, vol.12, issue.11, pp.835-840, 2007.
DOI : 10.1007/s00432-007-0225-x

A. Li, J. King, A. Moro, M. Sugi, and D. Dawson, Overexpression of CXCL5 Is Associated With Poor Survival in Patients With Pancreatic Cancer, The American Journal of Pathology, vol.178, issue.3, pp.1340-1349, 2011.
DOI : 10.1016/j.ajpath.2010.11.058

Y. Yahata, Y. Shirakata, S. Tokumaru, K. Yamasaki, and K. Sayama, Nuclear Translocation of Phosphorylated STAT3 Is Essential for Vascular Endothelial Growth Factor-induced Human Dermal Microvascular Endothelial Cell Migration and Tube Formation, Journal of Biological Chemistry, vol.13, issue.41, pp.40026-40031, 2003.
DOI : 10.1006/bbrc.1998.8254

B. Eliceiri, R. Paul, P. Schwartzberg, J. Hood, and J. Leng, Selective Requirement for Src Kinases during VEGF-Induced Angiogenesis and Vascular Permeability, Molecular Cell, vol.4, issue.6, pp.915-924, 1999.
DOI : 10.1016/S1097-2765(00)80221-X

M. Saino, T. Maruyama, T. Sekiya, T. Kayama, and Y. Murakami, Inhibition of angiogenesis in human glioma cell lines by antisense RNA from the soluble guanylate cyclase genes, GUCY1A3 and GUCY1B3, Oncology Reports, vol.12, pp.47-52, 2004.
DOI : 10.3892/or.12.1.47

L. Morbidelli, A. Pyriochou, S. Filippi, I. Vasileiadis, and C. Roussos, The soluble guanylyl cyclase inhibitor NS-2028 reduces vascular endothelial growth factor-induced angiogenesis and permeability, American Journal of Physiology-Regulatory, Integrative and Comparative Physiology, vol.269, issue.3, pp.824-832, 2009.
DOI : 10.1172/JCI117557

M. Moro, R. Russel, S. Cellek, I. Lizasoain, and Y. Su, cGMP mediates the vascular and platelet actions of nitric oxide: confirmation using an inhibitor of the soluble guanylyl cyclase., Proceedings of the National Academy of Sciences, vol.93, issue.4, pp.1480-1485, 1996.
DOI : 10.1073/pnas.93.4.1480

C. Balduini, A. Iolascon, and A. Savoia, Inherited thrombocytopenias: from genes to therapy, Haematologica, vol.87, pp.860-880, 2002.

K. Heath, A. Campos-barros, A. Toren, G. Rozenfeld-granot, and L. Carlsson, Nonmuscle Myosin Heavy Chain IIA Mutations Define a Spectrum of Autosomal Dominant Macrothrombocytopenias: May-Hegglin Anomaly and Fechtner, Sebastian, Epstein, and Alport-Like Syndromes, The American Journal of Human Genetics, vol.69, issue.5, pp.1033-1045, 2001.
DOI : 10.1086/324267

L. Gilles, D. Bluteau, S. Boukour, Y. Chang, and Y. Zhang, MAL/SRF complex is involved in platelet formation and megakaryocyte migration by regulating MYL9 (MLC2) and MMP9, Blood, vol.114, issue.19, pp.4221-4232, 2009.
DOI : 10.1182/blood-2009-03-209932

G. Jalagadugula, G. Mao, G. Kaur, L. Goldfinger, and D. Dhanasekaran, Regulation of platelet myosin light chain (MYL9) by RUNX1: implications for thrombocytopenia and platelet dysfunction in RUNX1 haplodeficiency, Blood, vol.116, issue.26, pp.6037-6045, 2010.
DOI : 10.1182/blood-2010-06-289850

P. Bray and M. Shuman, Identification of an abnormal gene for the GPIIIa subunit of the platelet fibrinogen receptor resulting in Glanzmann's thrombasthenia, Blood, vol.75, pp.881-888, 1990.

A. Leutenegger, M. Sahbatou, S. Gazal, H. Cann, and E. Genin, Consanguinity around the world: what do the genomic data of the HGDP-CEPH diversity panel tell us?, European Journal of Human Genetics, vol.26, issue.5, pp.583-587, 2011.
DOI : 10.1159/000067666

C. Fuchsberger, D. Taliun, P. Pramstaller, and C. Pattaro, GWAtoolbox: an R package for fast quality control and handling of genome-wide association studies meta-analysis data, Bioinformatics, vol.81, issue.3, pp.444-445, 2012.
DOI : 10.1086/519795

C. Willer, Y. Li, and G. Abecasis, METAL: fast and efficient meta-analysis of genomewide association scans, Bioinformatics, vol.40, issue.5, pp.2190-2191, 2010.
DOI : 10.1038/ng.120

C. Newton-cheh, M. Eijgelsheim, K. Rice, P. De-bakker, and X. Yin, Common variants at ten loci influence QT interval duration in the QTGEN Study, Nature Genetics, vol.79, issue.4, pp.399-406, 2009.
DOI : 10.1002/gepi.20303

S. Purcell, N. B. Todd-brown, K. Thomas, L. Ferreira, and M. , PLINK: A Tool Set for Whole-Genome Association and Population-Based Linkage Analyses, The American Journal of Human Genetics, vol.81, issue.3, pp.559-575, 2007.
DOI : 10.1086/519795

URL : https://doi.org/10.1086/519795