L. Bordenave, P. Menu, C. Baquey, G. W. Bos, A. A. Poot et al., Developments towards tissue-engineered, small-diameter arterial substitutes, Arch Physiol Biochem, vol.5, issue.3, pp.100-115, 1998.

D. E. Cohen and D. Melton, Turning straw into gold: directing cell fate for regenerative medicine, Nat Rev Genet, vol.12, issue.4, pp.243-52, 2011.

K. Takahashi and S. Yamanaka, Induction of Pluripotent Stem Cells from Mouse Embryonic and Adult Fibroblast Cultures by Defined Factors, Cell, vol.126, issue.4, pp.663-76, 2006.

J. Yu, M. A. Vodyanik, K. Smuga-otto, A. , J. Frane et al.,

S. , Induced pluripotent stem cell lines derived from human somatic cells. science, vol.318, pp.1917-1920, 2007.

R. Langer and J. P. Vacanti, Tissue Engineering. Science, vol.260, issue.5110, pp.920-926, 1993.

J. Mansbridge, Skin substitutes to enhance wound healing, Expert Opin Investig Drugs, vol.7, issue.5, p.803, 1998.

D. G. Seifu, A. Purnama, K. Mequanint, and D. Mantovani, Small-diameter vascular tissue engineering, Nat Rev Cardiol, vol.10, issue.7, pp.410-431, 2013.

, The European Society for Biomaterials 9th European Conference on

B. Chester, and IUPAC working party on blood compatibility, UK 9-11th September, 1991 in conjunction with 2nd Consensus Conference on Definitions in Biomaterials 7-8th September, vol.2, pp.62-62, 1991.

D. F. Williams, On the nature of biomaterials, Biomaterials, 2009.

N. R. Patel, P. Gohil, A. Teo, A. Mishra, I. Park et al., Biomaterials and Modifications in the Development of Small-Diameter Vascular Grafts, A review on biomaterials: scope, applications & human anatomy significance, vol.2, pp.227-51, 2012.

W. Suchanek and M. Yoshimura, Processing and properties of hydroxyapatitebased biomaterials for use as hard tissue replacement implants, J Mater Res, vol.13, issue.01, pp.94-117, 1998.

S. M. Zakaria, S. Zein, S. H. Othman, M. R. Yang, F. Jansen et al., Nanophase Hydroxyapatite as a Biomaterial in Advanced Hard Tissue Engineering: A Review, Tissue Eng Part B Rev, vol.19, issue.5, pp.431-472, 2013.

, Biodegradable polymers and composites in biomedical applications: from catgut to tissue engineering. Part 2 Systems for temporary replacement and advanced tissue regeneration, An Introduction to Materials in Medicine. Biomater Sci, vol.3, issue.5, pp.274-85, 2004.

C. Erisken, D. M. Kalyon, and H. Wang, Functionally graded electrospun polycaprolactone and ?-tricalcium phosphate nanocomposites for tissue engineering applications, Schmidt CE, Baier JM. Acellular vascular tissues: natural biomaterials for tissue repair and tissue engineering, vol.29, pp.2215-2231, 2000.

K. C. Dee, D. A. Puleo, R. Bizios, R. Sridharan, A. R. Cameron et al., Biomaterial based modulation of macrophage polarization: a review and suggested design principles, Mater Today, vol.18, issue.6, pp.313-325, 2002.

J. M. Morais, F. Papadimitrakopoulos, and D. J. Burgess, Biomaterials/Tissue Interactions: Possible Solutions to Overcome Foreign Body Response, AAPS J, vol.12, issue.2, pp.188-96, 2010.

J. M. Anderson, Agence de la santé publique du Canada. Le fardeau économique de la maladie au Canada, Cardiovascular diseases (CVDs) Global status report on noncommunicable diseases, vol.31, 2001.

D. Srivasta, C. Olson-en, L. Kr, C. P. Aj, and J. Rk, Vascular biology, Nature, issue.6801, p.407, 2000.

P. Libby, Inflammation in atherosclerosis, vol.420, p.7, 2002.

R. S. Reneman, T. Arts, and A. Hoeks, Wall Shear Stress-an Important Determinant of Endothelial Cell Function and Structure-in the Arterial System in vivo, J Vasc Res, vol.43, issue.3, pp.251-69, 2006.

C. Hahn and M. A. Schwartz, Mechanotransduction in vascular physiology and atherogenesis, Nat Rev Mol Cell Biol, vol.10, issue.1, pp.53-62, 2009.
DOI : 10.1038/nrm2596

URL : http://europepmc.org/articles/pmc2719300?pdf=render

K. S. Cunningham and A. I. Gotlieb, The role of shear stress in the pathogenesis of atherosclerosis, Lab Invest, vol.85, issue.1, pp.9-23, 2005.

S. Jono, M. D. Mckee, C. E. Murry, A. Shioi, Y. Nishizawa et al., Phosphate regulation of vascular smooth muscle cell calcification, Circ Res, vol.87, issue.7, pp.10-17, 2000.
DOI : 10.1161/01.res.87.7.e10

URL : http://circres.ahajournals.org/content/87/7/e10.full.pdf

E. Kasper-hoebe, B. Janssen, and . Beutler, The interface between innate and adaptive immunity, Nat Immunol, vol.5, issue.10, p.971, 2004.

G. Kikuchi, T. Yoshida, and M. Noguchi, Heme oxygenase and heme degradation

, Biochem Biophys Res Commun, vol.338, issue.1, pp.558-67, 2005.

L. Lanotte, J. Mauer, S. Mendez, D. A. Fedosov, J. Fromental et al., Red cells' dynamic morphologies govern blood shear thinning under microcirculatory flow conditions, Mulligan-Kehoe MJ, Simons M. Vasa Vasorum in Normal and Diseased Arteries, vol.113, pp.2557-66, 2014.
DOI : 10.1073/pnas.1608074113

URL : https://hal.archives-ouvertes.fr/hal-01435953

S. Moncada and A. Higgs, The vascular endothelium II

, =%22and+production:+LETEX+Jelonek,+Schmidt+%26+V%C3%B6ckler+GbR,+Leipzig,%22+%22been+very+pr oductive+and+its+results+have+contributed,+arguably+more%22+%22result,+we+have+ produced+two+volumes+which+is+a+good+representation+of%22+&ots=pWvPS484sk, pp.55-56, 2006.

S. Chien, Molecular basis of rheological modulation of endothelial functions: importance of stress direction, 40. Fox SI. Human Physiology, vol.43, issue.2, pp.33-43, 1983.

L. J. Ignarro, J. M. Fukuto, J. M. Griscavage, N. E. Rogers, R. E. Byrns et al., Multiple Mesoderm Subsets Give Rise to Endothelial Cells, Whereas Hematopoietic Cells Are Differentiated Only from a Restricted Subset in Embryonic Stem Cell Differentiation Culture, J Hematother Stem Cell Res, vol.90, issue.17, pp.401-412, 1993.

P. Carmeliet, Angiogenesis in life, disease and medicine, Nature, vol.438, issue.7070, pp.932-938, 2005.
DOI : 10.1038/nature04478

P. Carmeliet, Semenza GL. Vasculogenesis, angiogenesis, and arteriogenesis: Mechanisms of blood vessel formation and remodeling, J Cell Biochem, vol.6, issue.4, pp.840-847, 2000.

S. Azzi and J. Gavard, Vaisseaux sanguins et tumeurs ou l'art du dialogue. médecine/sciences, vol.30, pp.408-422, 2014.
DOI : 10.1051/medsci/20143004015

URL : https://www.medecinesciences.org/articles/medsci/pdf/2014/05/medsci20143004p408.pdf

T. Asahara, T. Murohara, A. Sullivan, M. Silver, R. Van-der-zee et al., Isolation of putative progenitor endothelial cells for angiogenesis, Science, vol.275, issue.5302, pp.964-966, 1997.
DOI : 10.1126/science.275.5302.964

D. Ribatti, A. Vacca, B. Nico, L. Roncali, and F. Dammacco, Postnatal vasculogenesis, Mech Dev, vol.100, issue.2, pp.157-163, 2001.
DOI : 10.1016/s0925-4773(00)00522-0

URL : https://doi.org/10.1016/s0925-4773(00)00522-0

S. Balaji, A. King, T. M. Crombleholme, S. Keswani, M. S. Goligorsky et al., The Role of Endothelial Progenitor Cells in Postnatal Vasculogenesis: Implications for Therapeutic Neovascularization and Wound Healing, Nat Rev Nephrol, vol.2, issue.6, pp.423-429, 2007.

A. M. Malek and S. Izumo, Mechanism of endothelial cell shape change and cytoskeletal remodeling in response to fluid shear stress, J Am Coll Cardiol, vol.109, issue.4, pp.1538-1585, 1996.

L. E. Mead, D. Prater, M. C. Yoder, D. Ingram, J. M. Martin et al., In vivo vasculogenic potential of human blood-derived endothelial progenitor cells, Curr Protoc Stem Cell Biol, vol.109, issue.11, pp.4761-4768, 2007.

T. Murohara, H. Ikeda, I. Onitsuka, T. Ueno, and K. Matsui, Endothelial progenitor cells in the human umbilical cord blood, CIRCULATION, vol.98, issue.17, pp.729-729, 1998.

M. Peichev, A. J. Naiyer, D. Pereira, Z. Zhu, W. J. Lane et al., Ingram DA. Identification of a novel hierarchy of endothelial progenitor cells using human peripheral and umbilical cord blood, J Société Biol, vol.95, issue.3, pp.2752-60, 2000.

M. Hristov, C. Weber, F. Timmermans, J. Plum, M. C. Yöder et al., Endothelial progenitor cells: characterization, pathophysiology, and possible clinical relevance, J Cell Mol Med, vol.8, issue.4, pp.498-508, 2004.
DOI : 10.1111/j.1582-4934.2004.tb00474.x

J. , Endothelial progenitor cells: identity defined?, J Cell Mol Med, vol.13, issue.1, pp.87-102, 2008.

J. Eggermann, S. Kliche, G. Jarmy, K. Hoffmann, U. Mayr-beyrle et al.,

M. , Endothelial progenitor cell culture and differentiation in vitro: a methodological comparison using human umbilical cord blood, Cardiovasc Res, vol.58, issue.2, pp.478-486, 2003.

X. Sun, L. Cheng, H. Duan, G. Lin, G. Lu et al., Nitric oxide production contributes to the angiogenic properties of vascular endothelial growth factor in human endothelial cells, J Tissue Eng Regen Med, vol.105, issue.7, pp.530-574, 1997.

C. J. Drake, L. Tilling, P. Chowienczyk, and B. Clapp, 72. Petit I, Jin D, Rafii S. The SDF-1-CXCR4 signaling pathway: a molecular hub modulating neo-angiogenesis, Br J Clin Pharmacol, vol.69, issue.1, pp.299-307, 2003.

M. Cheng and G. Qin, Progenitor Cell Mobilization and Recruitment, p.1

C. , Progress in Molecular Biology and Translational Science, pp.243-64, 2012.

M. Eguchi, H. Masuda, and T. Asahara, Endothelial progenitor cells for postnatal vasculogenesis, Clin Exp Nephrol, vol.11, issue.1, pp.18-25, 2007.
DOI : 10.1007/s10157-006-0448-1

T. Asahara and A. Kawamoto, Smadja DM, Gaussem P. Caractérisation des progéniteurs endothéliaux et stratégies d'expansion in vitro, AJP Cell Physiol, vol.287, issue.3, pp.197-207, 2004.

H. Masuda, C. Alev, H. Akimaru, R. Ito, T. Shizuno et al.,

, Methodological Development of a Clonogenic Assay to Determine Endothelial Progenitor Cell Potential, Circ Res, vol.109, issue.1, pp.20-37, 2011.

S. Ferratge, G. Ha, G. Carpentier, N. Arouche, R. Bascetin et al.,

, Initial clonogenic potential of human endothelial progenitor cells is predictive of their further properties and establishes a functional hierarchy related to immaturity, Stem Cell Res

. Apr, , 2017.

I. Ahrens, H. Domeij, D. Topcic, I. Haviv, R. Merivirta et al.,

, Successful In Vitro Expansion and Differentiation of Cord Blood Derived CD34+ Cells into Early Endothelial Progenitor Cells Reveals Highly Differential Gene Expression, PLoS ONE, vol.6, issue.8, p.23210, 2011.

K. K. Hirschi, D. A. Ingram, and M. C. Yoder, Assessing Identity, Phenotype, and Fate of Endothelial Progenitor Cells, Arterioscler Thromb Vasc Biol, vol.28, issue.9, pp.1584-95, 2008.

H. A. Hadi, C. S. Carr, J. Suwaidi, D. G. Seifu, A. Purnama et al., Endothelial dysfunction: cardiovascular risk factors, therapy, and outcome. Vasc Health Risk Manag, Nat Rev Cardiol, vol.1, issue.3, pp.410-431, 2005.

P. Graft-past and F. , Tissue Eng Part B Rev, p.2017, 2015.

J. G. Meinhart, M. Deutsch, T. Fischlein, N. Howanietz, A. Fröschl et al.,

M. Deutsch, J. Meinhart, P. Zilla, N. Howanietz, M. Gorlitzer et al., Long-term experience in autologous in vitro endothelialization of infrainguinal ePTFE grafts, Ann Thorac Surg, vol.71, issue.5, pp.352-62, 2001.

J. Kunlin, Long vein transplantation in treatment of ischemia caused by

R. A. Tineli, F. Viaro, M. B. Dalio, G. S. Reis, S. Basseto et al., Rev Chir, vol.70, issue.7-8, p.206, 1951.

N. Chakfé, F. Dieval, F. Thaveau, S. Rinckenbach, O. Hassani et al., Rev Bras Cir Cardiovasc, Forças mecânicas e veias safenas humanas: implicação na revascularização do miocárdio, vol.22, pp.301-310, 2004.

R. M. Langer and B. D. Kahan, Blakemore AH, Voorhees AB. The Use of Tubes Constructed from Vinyon "N" Cloth in Bridging Arterial Defects-Experimental and Clinical, Transplantation proceedings, vol.90, pp.324-357, 1947.

O. Creech, M. E. Debakey, M. Self, B. Halpert, R. Y. Kannan et al., The fate of heterologous arterial grafts: An experimental study, J Biomed Mater Res B Appl Biomater, vol.36, issue.3, pp.570-81, 1954.

H. He, T. Shirota, H. Yasui, and T. Matsuda, Canine endothelial progenitor celllined hybrid vascular graft with nonthrombogenic potential, J Thorac Cardiovasc Surg, vol.126, issue.2, pp.455-64, 2003.

A. H. Huang and L. E. Niklason, Engineering of arteries in vitro, Cell Mol Life Sci

C. M. Vaz, S. Van-tuijl, C. Bouten, and F. Baaijens, Design of scaffolds for blood vessel tissue engineering using a multi-layering electrospinning technique, Acta Biomater, vol.1, issue.5, pp.575-82, 2005.

C. Huang, R. Chen, Q. Ke, Y. Morsi, K. Zhang et al., Electrospun collagenchitosan-TPU nanofibrous scaffolds for tissue engineered tubular grafts, Colloids Surf B Biointerfaces, vol.82, issue.2, pp.307-322, 2011.

M. Poh, M. Boyer, A. Solan, S. Dahl, D. Pedrotty et al., Stem Cell Sources for Vascular Tissue Engineering and Regeneration, Tissue Eng Part B Rev, vol.365, issue.5, pp.405-430, 2005.

, Biomaterials, vol.35, issue.38, pp.10009-10023, 2014.

B. D. Ratner, The atherosusceptible endothelium: endothelial phenotypes in complex haemodynamic shear stress regions in vivo, The Biomaterials: Silver Jubilee Compendium, vol.99, pp.315-342, 2004.

M. S. Lemson, J. Tordoir, M. Daemen, P. Kitslaar, W. M. Abbott et al., Effects of an Artery/Vascular Graft Compliance Mismatch on Protein Transport: A Numerical Study, Eur J Vasc Endovasc Surg, vol.19, issue.4, pp.991-1006, 1987.

F. Khoffi, Contribution à l'étude de la compliance et du vieillissement des prothèses artérielles, p.189

J. M. Anderson, A. Rodriguez, and D. T. Chang, Foreign body reaction to biomaterials, Semin Immunol, vol.20, issue.2, pp.86-100, 2008.

R. A. Jonas, G. Ziemer, F. J. Schoen, L. Britton, and A. R. Castaneda, A new sealant for knitted Dacron prostheses: minimally cross-linked gelatin, J Vasc Surg, vol.7, issue.3, pp.414-419, 1988.

U. Hersel, C. Dahmen, H. Kessler, H. Shin, J. S. Mikos et al., RGD modified polymers: biomaterials for stimulated cell adhesion and beyond, Biomaterials, vol.24, issue.24, pp.4385-415, 2003.

, Biomaterials, vol.24, issue.24, pp.4353-64, 2003.

J. H. Collier and T. Segura, Evolving the use of peptides as components of biomaterials, Biomaterials, vol.32, issue.18, pp.4198-204, 2011.

M. Sgarioto, P. Vigneron, J. Patterson, F. Malherbe, M. Nagel et al.,

, Collagen type I together with fibronectin provide a better support for endothelialization, C R Biol, vol.335, issue.8, pp.520-528, 2012.

K. Lee, E. A. Silva, and D. J. Mooney, Growth factor delivery-based tissue engineering: general approaches and a review of recent developments, J R Soc Interface, vol.8, issue.55, pp.153-70, 2011.

E. S. Wijelath, S. Rahman, J. Murray, Y. Patel, G. Savidge et al., Fibronectin promotes VEGF-induced CD34+ cell differentiation into endothelial cells, J Vasc Surg, vol.39, issue.3, pp.655-60, 2004.

U. Edlund, T. Sauter, and A. Albertsson, Covalent VEGF protein immobilization on resorbable polymeric surfaces, Polym Adv Technol, 2011.

L. Saux, G. Plawinski, L. Parrot, C. Nlate, S. Servant et al., Surface bound VEGF mimicking peptide maintains endothelial cell proliferation in the absence of soluble VEGF in vitro: SVVYGLR TO MAINTAIN HUVEC PROLIFERATION, J Biomed Mater Res A, vol.104, issue.6, pp.1425-1461, 2016.

C. K. Poh, Z. Shi, T. Y. Lim, K. G. Neoh, and W. Wang, The effect of VEGF functionalization of titanium on endothelial cells in vitro, Biomaterials, vol.31, issue.7, pp.1578-85, 2010.

B. Randone, G. Cavallaro, A. Polistena, A. Cucina, P. Coluccia et al., Dual Role of VEGF in Pretreated Experimental ePTFE Arterial Grafts, J Surg Res, vol.127, issue.2, pp.70-79, 2005.

Y. Suzuki, K. Yamamoto, J. Ando, K. Matsumoto, and T. Matsuda, Arterial shear stress augments the differentiation of endothelial progenitor cells adhered to VEGF-bound surfaces, Biochem Biophys Res Commun, vol.423, issue.1, pp.91-98, 2012.

D. Visscher, G. Mesure, L. Meuris, B. Ivanova, A. Flameng et al., Improved endothelialization and reduced thrombosis by coating a synthetic vascular graft with fibronectin and stem cell homing factor SDF-1?, J Controlled Release, vol.8, issue.3, pp.68-75, 2012.

M. Zhou, Z. Liu, K. Li, W. Qiao, X. Jiang et al., Beneficial effects of granulocyte-colony stimulating factor on small-diameter heparin immobilized decellularized vascular graft, J Biomed Mater Res A, vol.95, issue.2, pp.1636-1681, 2010.

O. D. Krishna and K. L. Kiick, Protein-and peptide-modified synthetic polymeric biomaterials, Biopolymers, vol.94, issue.1, pp.32-48, 2010.

F. Caiado, S. Dias, V. Kähäri, J. Heino, and K. Elenius, Endothelial progenitor cells and integrins: adhesive needs, Fibrogenesis Tissue Repair, vol.5, issue.1, pp.13-22, 2002.

M. D. Pierschbacher and E. Ruoslahti, Variants of the cell recognition site of fibronectin that retain attachment-promoting activity, Proc Natl Acad Sci, vol.81, pp.5985-5993, 1984.

E. Ruoslahti and M. D. Pierschbacher, New Perspectives in Cell Adhesion: RGD and Integrins, Science, vol.238, issue.4826, pp.491-498, 1987.

Y. Lei, M. Rémy, C. Labrugère, and M. Durrieu, Peptide immobilization on polyethylene terephthalate surfaces to study specific endothelial cell adhesion, spreading and migration, J Mater Sci Mater Med, vol.23, issue.11, pp.2761-72, 2012.
URL : https://hal.archives-ouvertes.fr/inserm-00724079

S. L. Bellis, Advantages of RGD peptides for directing cell association with biomaterials, Biomaterials, vol.32, issue.18, pp.4205-4215, 2011.

X. Ren, Y. Feng, J. Guo, H. Wang, Q. Li et al., Surface modification and endothelialization of biomaterials as potential scaffolds for vascular tissue engineering applications, Chem Soc Rev, vol.44, issue.15, pp.5680-742, 2015.

J. Ruiz-rodríguez, M. Miguel, S. Preciado, G. A. Acosta, A. J. Bidon-chanal et al., Polythiazole linkers as functional rigid connectors: a new RGD cyclopeptide with enhanced integrin selectivity, Chem Sci, vol.5, issue.10, p.3929, 2014.

X. Ren, Y. Feng, J. Guo, H. Wang, Q. Li et al., Surface modification and endothelialization of biomaterials as potential scaffolds for vascular tissue engineering applications, Chem Soc Rev, vol.44, issue.15, pp.5680-742, 2015.

K. Kanie, Y. Narita, Y. Zhao, F. Kuwabara, M. Satake et al., Collagen type IV-specific tripeptides for selective adhesion of endothelial and smooth muscle cells, Biotechnol Bioeng, vol.109, issue.7, pp.1808-1824, 2012.

L. Saux, G. Magenau, A. Gunaratnam, K. Kilian, K. A. Böcking et al., Spacing of Integrin Ligands Influences Signal Transduction in Endothelial Cells, Biophys J, vol.101, issue.4, pp.764-73, 2011.

C. Chollet, C. Chanseau, M. Remy, A. Guignandon, R. Bareille et al., The effect of RGD density on osteoblast and endothelial cell behavior on RGD-grafted polyethylene terephthalate surfaces, Biomaterials, vol.30, issue.5, pp.711-731, 2009.
URL : https://hal.archives-ouvertes.fr/inserm-00395608

M. Yin, Y. Yuan, C. Liu, and J. Wang, Combinatorial coating of adhesive polypeptide and anti-CD34 antibody for improved endothelial cell adhesion and proliferation, J Mater Sci Mater Med, vol.20, issue.7, pp.1513-1536, 2009.

J. I. Rotmans, In Vivo Cell Seeding With Anti-CD34 Antibodies Successfully Accelerates Endothelialization but Stimulates Intimal Hyperplasia in Porcine Arteriovenous Expanded Polytetrafluoroethylene Grafts. Circulation, vol.112, pp.12-20, 2005.

J. Li, D. Li, F. Gong, S. Jiang, H. Yu et al., Capture of Flowing Endothelial Cells Using Surface-Immobilized Anti-Kinase Insert Domain Receptor Antibody, Tissue Eng Part C Methods, vol.2014, p.110306233138079, 2011.

M. Zhang, Z. Wang, Z. Wang, S. Feng, H. Xu et al., Immobilization of anti-CD31 antibody on electrospun poly(?-caprolactone) scaffolds through hydrophobins for specific adhesion of endothelial cells, Colloids Surf B Biointerfaces, vol.85, issue.1, pp.32-41, 2011.

, Surface Functionalization of Poly(ethylene terephthalate) Film and Membrane by Controlled Wet Chemistry: Chemical Characterization of Carboxylated Surfaces, J Colloid Interface Sci, vol.173, issue.1, pp.236-280, 1995.

P. Mougenot, M. Koch, I. Dupont, Y. Schneider, and J. Marchand-brynaert,

, Surface Functionalization of Polyethylene Terephthalate Film and Membranes by Controlled Wet Chemistry: II. Reactivity Assays of Hydroxyl Chain Ends, J Colloid Interface Sci, vol.177, issue.1, pp.162-170, 1996.

T. Boxus, M. Deldimerubbens, P. Mougenot, Y. Schneider, and M. ,

J. Fischer-colbrie, G. Heumann, S. Liebminger, S. Almansa, E. Cavaco-paulo et al., Chemical assays of end-groups displayed on the surface of poly(ethylene terephthalate) (PET) films and membranes by radiolabeling, Biocatal Biotransformation, vol.7, issue.7, pp.341-347, 1996.

F. R. Pu, R. L. Williams, T. K. Markkula, and J. A. Hunt, Effects of plasma treated PET and PTFE on expression of adhesion molecules by human endothelial cells in vitro, Biomaterials, vol.23, issue.11, pp.2411-2428, 2002.

K. S. Siow, L. Britcher, S. Kumar, H. Griesser, R. Jingrun et al., Plasma Methods for the Generation of Chemically Reactive Surfaces for Biomolecule Immobilization and Cell Colonization-A Review. Plasma Process Polym, Appl Surf Sci, vol.3, issue.6-7, pp.263-269, 2006.

P. K. Thalla, A. Contreras-garcía, H. Fadlallah, J. Barrette, D. Crescenzo et al.,

Y. Merhi, A Versatile Star PEG Grafting Method for the Generation of Nonfouling and Nonthrombogenic Surfaces, BioMed Res Int, vol.2013, pp.1-12, 2013.

C. Chollet, C. Chanseau, B. Brouillaud, and M. C. Durrieu, RGD peptides grafting onto poly(ethylene terephthalate) with well controlled densities, Biomol Eng, vol.24, issue.5, pp.477-82, 2007.

R. G. Lebaron, K. Athanasiou, T. T. Lee, J. R. García, J. I. Paez et al., Lighttriggered in vivo activation of adhesive peptides regulates cell adhesion, inflammation and vascularization of biomaterials, Smadja DM. PAR-1 Activation on Human Late Endothelial Progenitor Cells Enhances Angiogenesis In Vitro, vol.6, pp.85-103, 2000.

, Arterioscler Thromb Vasc Biol, vol.25, issue.11, pp.2321-2328, 2005.

P. Zilla, D. Bezuidenhout, and P. Human, Prosthetic vascular grafts: Wrong models, wrong questions and no healing, Biomaterials, vol.28, issue.34, pp.5009-5036, 2007.

M. Zhou, Z. Liu, C. Liu, X. Jiang, Z. Wei et al., Tissue engineering of small-diameter vascular grafts by endothelial progenitor cells seeding heparin-coated decellularized scaffolds, J Biomed Mater Res B Appl Biomater, vol.100, issue.1, pp.111-131, 2012.

J. Aoki, P. W. Serruys, H. Van-beusekom, A. Ong, E. P. Mcfadden et al., Endothelial Progenitor Cell Capture by Stents Coated With Antibody Against CD34, J Am Coll Cardiol, vol.45, issue.10, pp.1574-1583, 2005.

X. Wang, Peptide linked polymers for cadiovascular applications

A. N. Veleva, D. E. Heath, S. L. Cooper, and C. Patterson, Selection and initial characterization of novel peptide ligands that bind specifically to human blood outgrowth endothelial cells, Veleva AN, vol.29, pp.306-318, 2007.

T. Takahashi, C. Kalka, H. Masuda, D. Chen, M. Silver et al.,

-. Ischemia, D. M. Smadja, I. Bieche, J. Emmerich, M. Aiach et al., PAR-1 activation has different effects on the angiogenic activity of endothelial progenitor cells derived from human adult and cord blood, J Thromb Haemost, vol.5, issue.4, pp.2729-2731, 1999.

C. Brenner, N. Kränkel, S. Kühlenthal, L. Israel, F. Remm et al.,

, Short-term inhibition of DPP-4 enhances endothelial regeneration after acute arterial injury via enhanced recruitment of circulating progenitor cells, Int J Cardiol, vol.177, issue.1, pp.266-75, 2014.

G. Chang, P. Zhang, L. Ye, K. Lu, Y. Wang et al., Protective effects of sitagliptin on myocardial injury and cardiac function in an ischemia/reperfusion rat model, Eur J Pharmacol, vol.718, issue.1-3, pp.105-118, 2013.

Z. Liu, V. Stanojevic, S. Avadhani, T. Yano, and J. F. Habener, Stromal cell-derived factor-1 (SDF-1)/chemokine (C-X-C motif) receptor 4 (CXCR4) axis activation induces intra-islet glucagon-like peptide-1 (GLP-1) production and enhances beta cell survival, Diabetologia, vol.54, issue.8, pp.2067-76, 2011.

C. Urbich, A. Aicher, C. Heeschen, E. Dernbach, W. Hofmann et al., Soluble factors released by endothelial progenitor cells promote migration of endothelial cells and cardiac resident progenitor cells, J Mol Cell Cardiol, vol.39, issue.5, pp.733-775, 2005.

C. Zhang, Y. Srinivasan, D. H. Arlow, J. J. Fung, D. Palmer et al., Highresolution crystal structure of human protease-activated receptor 1, Nature, vol.492, issue.7429, pp.387-92, 2009.

M. A. Arnaout, B. Mahalingam, J. Xiong, M. Barczyk, S. Carracedo et al., Integrin structure, allostery, and bidirectional signaling, Annu Rev Cell Dev Biol, vol.21, 2005.

E. F. Plow, T. A. Haas, L. Zhang, J. Loftus, and J. W. Smith, Ligand Binding to Integrins, J Biol Chem, vol.339, issue.1, pp.21785-21793, 2000.

U. Wudebwe, A. Bannerman, P. Goldberg-oppenheimer, J. Z. Paxton, R. L. Williams et al., Exploiting cell-mediated contraction and adhesion to structure tissues in vitro, Philos Trans R Soc B Biol Sci, vol.370, pp.20140200-20140200, 1661.

C. S. Chen, M. Mrksich, S. Huang, G. M. Whitesides, D. Ingber et al., Cell Spreading and Focal Adhesion Dynamics Are Regulated by Spacing of Integrin Ligands, Biophys J, vol.276, issue.5317, pp.2964-74, 1997.

T. Pompe, C. Mitdank, and C. Werner, Quantitative analysis of fibronectin fibrillogenesis by endothelial cells on biomaterials, J Phys Condens Matter, vol.16, issue.26, pp.2421-2427, 2004.

T. Pompe, L. Renner, and C. Werner, Nanoscale Features of Fibronectin Fibrillogenesis Depend on Protein-Substrate Interaction and Cytoskeleton Structure, Biophys J, vol.88, issue.1, pp.527-561, 2005.

R. Peng, X. Yao, and J. Ding, Effect of cell anisotropy on differentiation of stem cells on micropatterned surfaces through the controlled single cell adhesion, Biomaterials, vol.32, issue.32, pp.8048-57, 2011.

K. A. Kilian, B. Bugarija, B. T. Lahn, and M. Mrksich, Geometric cues for directing the differentiation of mesenchymal stem cells, Proc Natl Acad Sci, vol.107, issue.11, pp.4872-4879, 2010.

J. P. Cooke and N. O. Flow, Proc Natl Acad Sci, vol.100, issue.3, pp.768-770, 2003.

D. E. Conway, M. R. Williams, S. G. Eskin, L. Mcintire, C. A. Hoesli et al., Endothelial cell responses to atheroprone flow are driven by two separate flow components: low timeaverage shear stress and fluid flow reversal, AJP Heart Circ Physiol, vol.298, issue.2, 2010.

, A fluorophore-tagged RGD peptide to control endothelial cell adhesion to micropatterned surfaces, Biomaterials, vol.35, issue.3, pp.879-90, 2014.

D. Canio, C. Lamponi, S. Barbucci, and R. , Spiral and square microstructured surfaces: The effect of the decreasing size of photo-immobilized hyaluronan domains on cell growth, J Biomed Mater Res A, vol.92, issue.1, pp.276-84, 2010.

S. Li, S. Bhatia, Y. L. Hu, Y. T. Shiu, Y. S. Li et al., Effects of morphological patterning on endothelial cell migration, Biorheology, vol.38, issue.2-3, pp.491-502, 2001.

A. Cerf, C. Vieu, ;. Kim, . Zhao, P. Rogers et al., Circumferential alignment of vascular smooth muscle cells in a circular microfluidic channel, Soft Lithography, a Tool to Address SingleObjects Investigations. INTECH Open Access Publisher; 2010. 186. Xia, vol.273, pp.63-70, 1996.

F. Ye, J. Jiang, H. Chang, L. Xie, J. Deng et al., Improved single-cell culture achieved using micromolding in capillaries technology coupled with poly (HEMA)

, Biomicrofluidics, vol.9, issue.4, p.44106, 2015.

J. Norman and T. Desai, Methods for Fabrication of Nanoscale Topography for Tissue Engineering Scaffolds, Ann Biomed Eng, vol.34, issue.1, pp.89-101, 2006.

L. E. Murr, K. N. Amato, S. J. Li, Y. X. Tian, X. Y. Cheng et al.,

, Microstructure and mechanical properties of open-cellular biomaterials prototypes for total knee replacement implants fabricated by electron beam melting, J Mech Behav Biomed Mater, vol.4, issue.7, pp.1396-411, 2011.

S. Barcelo and Z. Li, Nanoimprint lithography for nanodevice fabrication, Imprint Materials for Nanoscale Devices. MRS Bull, vol.3, issue.1, pp.947-51, 2005.

E. Engel, E. Martínez, C. A. Mills, M. Funes, J. A. Planell et al.,

, Mesenchymal stem cell differentiation on microstructured poly (methyl methacrylate) substrates, Ann Anat, vol.191, issue.1, pp.136-180, 2009.

D. Franco, M. Klingauf, M. Bednarzik, M. Cecchini, V. Kurtcuoglu et al.,

J. , Control of initial endothelial spreading by topographic activation of focal adhesion kinase, Soft Matter, vol.7, issue.16, pp.7313-7337, 2011.

S. A. Bustin, V. Benes, J. A. Garson, J. Hellemans, J. Huggett et al.,

M. The and . Guidelines, Minimum Information for Publication of Quantitative Real-Time PCR Experiments, Clin Chem, vol.55, issue.4, pp.611-633, 2009.

S. Taylor, M. Wakem, G. Dijkman, M. Alsarraj, and M. Nguyen, A practical approach to RT-qPCR-Publishing data that conform to the MIQE guidelines, Methods, vol.50, issue.4, pp.1-5, 2010.

S. C. Taylor and E. M. Mrkusich, The State of RT-Quantitative PCR: Firsthand Observations of Implementation of Minimum Information for the Publication of Quantitative Real-Time PCR Experiments (MIQE), J Mol Microbiol Biotechnol, vol.24, issue.1, pp.46-52, 2014.

H. Hung, W. Shyu, C. Tsai, S. Hsu, and S. Lin, Transplantation of

, Urbich C. Endothelial Progenitor Cells: Characterization and Role in Vascular Biology, Cell Transplant, vol.18, issue.9, pp.343-53, 2004.

W. Risau and F. I. Vasculogenesis, Annu Rev Cell Dev Biol, vol.11, issue.1, pp.73-91, 1995.

T. Asahara, T. Murohara, A. Sullivan, M. Silver, R. Van-der-zee et al., Isolation of putative progenitor endothelial cells for angiogenesis, Science, vol.275, issue.5302, pp.964-966, 1997.

S. Patan, Vasculogenesis and Angiogenesis as Mechanisms of Vascular Network Formation, Growth and Remodeling, J Neurooncol, vol.50, issue.1, pp.1-15, 2000.

J. A. Nagy, L. Benjamin, H. Zeng, A. M. Dvorak, and H. F. Dvorak, Vascular permeability, vascular hyperpermeability and angiogenesis, Angiogenesis, vol.11, issue.2, pp.109-128, 2008.

Q. Shi, M. Wu, N. Hayashida, A. R. Wechezak, A. W. Clowes et al., Proof of fallout endothelialization of impervious dacron grafts in the aorta and inferior vena cava of the dog, J Vasc Surg, vol.20, issue.4, pp.546-57, 1994.

T. Liu, S. Liu, K. Zhang, J. Chen, and N. Huang, Endothelialization of implanted cardiovascular biomaterial surfaces: The development from in vitro to in vivo: Endothelialization of Implanted Cardiovascular Biomaterial Surfaces, J Biomed Mater Res A, vol.102, issue.10, pp.3754-72, 2014.

A. Kawamoto and D. W. Losordo, Endothelial Progenitor Cells for Cardiovascular Regeneration, Trends Cardiovasc Med, vol.18, issue.1, pp.33-40, 2008.

M. C. Yoder, Human Endothelial Progenitor Cells. Cold Spring Harb Perspect Med, vol.2, pp.6692-006692, 2012.

C. Cheng, S. Chang, Y. Chueh, T. Huang, P. Huang et al., Distinct angiogenesis roles and surface markers of early and late endothelial progenitor cells revealed by functional group analyses, Arterioscler Thromb Vasc Biol, vol.14, issue.1, p.1527, 2000.

M. C. Yoder, L. E. Mead, D. Prater, T. R. Krier, K. N. Mroueh et al., Redefining endothelial progenitor cells via clonal analysis and hematopoietic stem/progenitor cell principals, Blood, vol.109, issue.5, pp.1801-1809, 2007.

S. A. Bustin, Quantification of mRNA using real-time reverse transcription PCR (RT-PCR): trends and problems, J Mol Endocrinol, vol.29, issue.1, pp.23-39, 2002.

E. Deindl, K. Boengler, N. Van-royen, and W. Schaper, Differential expression of GAPDH and ?-actin in growing collateral arteries, Mol Cell Biochem, vol.236, issue.1, pp.139-146, 2002.

A. Radoni?, S. Thulke, I. M. Mackay, O. Landt, W. Siegert et al., Guideline to reference gene selection for quantitative real-time PCR, Biochem Biophys Res Commun, vol.313, issue.4, pp.856-62, 2004.

J. Synnergren, T. L. Giesler, S. Adak, R. Tandon, K. Noaksson et al., , p.161

N. B. Thébaud, R. Bareille, R. Daculsi, C. Bourget, M. Rémy et al., Polyelectrolyte multilayer films allow seeded human progenitor-derived endothelial cells to remain functional under shear stress in vitro, Acta Biomater, vol.25, issue.2, pp.1437-1482, 2007.

J. Kim, Y. Jeon, H. E. Kim, J. M. Shin, H. M. Chung et al., Comparative proteomic analysis of endothelial cells progenitor cells derived from cord blood-and peripheral blood for cell therapy, Biomaterials, vol.34, issue.6, pp.1669-85, 2013.

M. Boivin, P. Chevallier, C. A. Hoesli, J. Lagueux, R. Bareille et al., Human saphenous vein endothelial cell adhesion and expansion on micropatterned polytetrafluoroethylene, J Biomed Mater Res A, vol.101, issue.3, pp.694-703, 2013.

A. Tichopad, Pfaffl MW, Tichopad A, Prgomet C, Neuvians TP. Determination of stable housekeeping genes, differentially regulated target genes and sample integrity: BestKeeper-Excel-based tool using pair-wise correlations, Nucleic Acids Res, vol.31, issue.6, pp.509-515, 2003.

C. L. Andersen, J. L. Jensen, and T. F. Ørntoft, Normalization of real-time quantitative reverse transcription-PCR data: a model-based variance estimation approach to identify genes suited for normalization, applied to bladder and colon cancer data sets, Cancer Res, vol.64, issue.15, pp.5245-5250, 2004.

J. Vandesompele, D. Preter, K. Pattyn, F. Poppe, B. Van-roy et al., Accurate normalization of real-time quantitative RT-PCR data by geometric averaging of multiple internal control genes, Genome Biol, vol.3, issue.7, pp.34-35, 2002.

N. Silver, S. Best, J. Jiang, S. Thein, A. Forootan et al., Methods to determine limit of detection and limit of quantification in quantitative real-time PCR (qPCR), BMC Mol Biol, vol.7, issue.1, pp.1-6, 2006.

E. Ragni, M. Viganò, P. Rebulla, R. Giordano, and L. Lazzari, What is beyond a qRT-PCR study on mesenchymal stem cell differentiation properties: how to choose the most reliable housekeeping genes, J Cell Mol Med, vol.17, issue.1, pp.168-80, 2013.

P. Rajendran, T. Rengarajan, J. Thangavel, Y. Nishigaki, and D. Sakthisekaran,

G. Sethi, The Vascular Endothelium and Human Diseases, Int J Biol Sci, vol.9, issue.10, pp.1057-69, 2013.

K. Stellos, H. Langer, K. Daub, T. Schoenberger, A. Gauss et al.,

, Platelet-Derived Stromal Cell-Derived Factor-1 Regulates Adhesion and Promotes Differentiation of Human CD34+ Cells to Endothelial Progenitor Cells. Circulation, vol.117, pp.206-221, 2008.

G. Chen, L. Zhao, J. Feng, G. You, Q. Sun et al., Validation of Reliable Reference Genes for Real-Time PCR in Human Umbilical Vein Endothelial Cells on Substrates with Different Stiffness, PLoS ONE, vol.8, issue.6, p.67360, 2013.

P. Brett, J. Harle, V. Salih, R. Mihoc, I. Olsen et al., Roughness response genes in osteoblasts, Bone, vol.35, issue.1, pp.124-157, 2004.

W. Chen, Y. Chen, C. Ko, Y. Lin, T. Kuo et al., Interaction of progenitor bone cells with different surface modifications of titanium implant, Mater Sci Eng C, vol.37, pp.305-318, 2014.

D. B. Oturai, H. B. Søndergaard, L. Börnsen, F. Sellebjerg, R. Christensen et al., Identification of Suitable Reference Genes for Peripheral Blood Mononuclear Cell Subset Studies in Multiple Sclerosis, Scand J Immunol, vol.83, issue.1, pp.72-80, 2016.

B. ?y?y?ska-granica and K. Koziak, Identification of Suitable Reference Genes for Real-Time PCR Analysis of Statin-Treated Human Umbilical Vein Endothelial Cells, PLoS ONE, vol.7, issue.12, p.51547, 2012.

T. Li, H. Diao, L. Zhao, Y. Xing, J. Zhang et al., Identification of suitable reference genes for real-time quantitative PCR analysis of hydrogen peroxide-treated human umbilical vein endothelial cells, J Am Heart Assoc, vol.18, issue.1, pp.504-000504, 2014.

J. Chlupac, E. Filova, and L. Bacakova, Blood vessel replacement: 50 years of development and tissue engineering paradigms in vascular surgery, Physiol Res, vol.58, p.119, 2009.

R. A. Jonas, G. Ziemer, F. J. Schoen, L. Britton, and A. R. Castaneda, A new sealant for knitted Dacron prostheses: minimally cross-linked gelatin, J Vasc Surg, vol.7, issue.3, pp.414-419, 1988.

Y. Takami, K. Tajima, W. Kato, K. Fujii, M. Hibino et al., Comparison of different vascular prostheses and matrices in relation to endothelial seeding, Br J Surg, vol.14, issue.5, pp.417-420, 1991.

P. C. Begovac, R. C. Thomson, J. L. Fisher, A. Hughson, and A. Gällhagen, Improvements in GORE-TEX® vascular graft performance by Carmeda® bioactive surface heparin immobilization, Eur J Vasc Endovasc Surg, vol.25, issue.5, pp.432-439, 2003.
DOI : 10.1053/ejvs.2002.1909

URL : https://doi.org/10.1053/ejvs.2002.1909

P. Feugier, R. A. Black, J. A. Hunt, and T. V. How, Attachment, morphology and adherence of human endothelial cells to vascular prosthesis materials under the action of shear stress, Biomaterials, vol.26, issue.13, pp.1457-66, 2005.

T. Kumar and L. K. Krishnan, Fibrin-mediated endothelial cell adhesion to vascular biomaterials resists shear stress due to flow, J Mater Sci Mater Med, vol.13, issue.8, pp.93-106, 2002.

D. Schmidt, C. Breymann, A. Weber, C. I. Guenter, S. Neuenschwander et al.,

G. , Fabrication of endothelial progenitor cell (EPC)-seeded intravascular stent devices and in vitro endothelialization on hybrid vascular tissue, Ann Thorac Surg, vol.78, issue.6, pp.2295-302, 2003.

R. Blindt, F. Vogt, I. Astafieva, C. Fach, M. Hristov et al., A Novel Drug-Eluting Stent Coated With an Integrin-Binding Cyclic Arg-Gly-Asp Peptide Inhibits Neointimal Hyperplasia by Recruiting Endothelial Progenitor Cells, J Am Coll Cardiol, vol.47, issue.9, pp.1786-95, 2006.
DOI : 10.1016/j.jacc.2005.11.081

URL : https://doi.org/10.1016/j.jacc.2005.11.081

C. Kang, W. Lim, S. Kyeong, W. Choe, H. Kim et al., Fabrication of biofunctional stents with endothelial progenitor cell specificity for vascular re-endothelialization, Colloids Surf B Biointerfaces, vol.102, pp.744-51, 2013.

J. M. Lee, W. Choe, B. Kim, W. Seo, W. Lim et al., Comparison of endothelialization and neointimal formation with stents coated with antibodies against CD34 and vascular endothelial-cadherin, Biomaterials, vol.33, issue.35, pp.8917-8944, 2012.

Q. Lin, X. Ding, F. Qiu, X. Song, G. Fu et al., In situ endothelialization of intravascular stents coated with an anti-CD34 antibody functionalized heparin-collagen multilayer, Biomaterials, vol.31, issue.14, pp.4017-4042, 2010.

X. Ye, X. Hu, H. Wang, J. Liu, and Q. Zhao, Polyelectrolyte multilayer film on decellularized porcine aortic valve can reduce the adhesion of blood cells without affecting the growth of human circulating progenitor cells, Acta Biomater, vol.8, issue.3, pp.1057-67, 2012.

Q. Li, Z. Wang, S. Zhang, W. Zheng, Q. Zhao et al., Functionalization of the surface of electrospun poly(epsilon-caprolactone) mats using zwitterionic poly(carboxybetaine methacrylate) and cell-specific peptide for endothelial progenitor cells capture, Mater Sci Eng C, vol.33, issue.3, pp.1646-53, 2013.

X. Chen, J. Wang, A. Q. Li, D. Liu, P. Zhu et al., Electrospun poly(l-lactic acid-co-?-caprolactone) fibers loaded with heparin and vascular endothelial growth factor to improve blood compatibility and endothelial progenitor cell proliferation, Colloids Surf B Biointerfaces, vol.128, pp.106-120, 2015.
DOI : 10.1016/j.colsurfb.2015.02.023

X. Bérard, M. Rémy-zolghadri, C. Bourget, N. Turner, R. Bareille et al., Capability of human umbilical cord blood progenitor-derived endothelial cells to form an efficient lining on a polyester vascular graft in vitro, Acta Biomater, vol.5, issue.4, pp.1147-57, 2009.

N. Chang, C. Lam, C. Lin, W. Chen, C. Li et al.,

, Transplantation of autologous endothelial progenitor cells in porous PLGA scaffolds create 164

, a microenvironment for the regeneration of hyaline cartilage in rabbits. Osteoarthritis Cartilage, vol.21, pp.1613-1635, 2013.

J. I. Rotmans, J. M. Heyligers, E. S. Stroes, and G. Pasterkamp, Schultz GS, Wysocki A. Interactions between extracellular matrix and growth factors in wound healing, Wound Repair Regen, vol.22, issue.11, pp.153-62, 2006.

U. Hersel, C. Dahmen, H. Kessler, T. Colin, M. Durrieu et al., RGD modified polymers: biomaterials for stimulated cell adhesion and beyond, Biomaterials, vol.24, issue.24, pp.4385-415, 2003.
DOI : 10.1016/s0142-9612(03)00343-0

, Modeling of the migration of endothelial cells on bioactive micropatterned polymers, Math Biosci Eng, vol.10, issue.4, pp.997-1015, 2013.

T. G. Kapp, F. Rechenmacher, S. Neubauer, O. V. Maltsev, E. A. Cavalcanti-adam et al., Efficacy and Safety of the Dipeptidyl Peptidase-4 Inhibitor Sitagliptin Added to Ongoing Metformin Therapy in Patients With Type 2 Diabetes Inadequately Controlled With Metformin Alone. Diabetes Care, A Comprehensive Evaluation of the Activity and Selectivity Profile of Ligands for RGD-binding Integrins. Sci Rep, vol.7, pp.522-556, 2001.

S. Kidoaki and T. Matsuda, Shape-engineered vascular endothelial cells: Nitric oxide production, cell elasticity, and actin cytoskeletal features, J Biomed Mater Res A, vol.81, issue.3, pp.728-763, 2007.

S. Lamponi, M. Forbicioni, and R. Barbucci, The role of fibronectin in cell adhesion to spiral patterned TiO2 nanoparticles, J Appl Biomater Biomech JABB, vol.7, issue.2, p.104, 2009.

E. Uchida, Y. Uyama, Y. Ikada, O. Sorption, and . Film, LANGMUIR, vol.9, issue.4, pp.1121-1125, 1993.

L. Pichavant, G. Amador, J. C. Brouillaud, B. Héroguez, V. Durrieu et al., pH-controlled delivery of gentamicin sulfate from orthopedic devices preventing nosocomial infections, J Controlled Release, vol.162, issue.2, pp.373-81, 2012.
URL : https://hal.archives-ouvertes.fr/hal-00757362

I. Bilem, P. Chevallier, L. Plawinski, E. D. Sone, M. C. Durrieu et al., RGD 165 and BMP-2 mimetic peptide crosstalk enhances osteogenic commitment of human bone marrow stem cells, Acta Biomater, vol.36, pp.132-174, 2016.

C. Royer, A. Bégin, L. Plawinski, L. Lévesque, M. Durrieu et al., Validation of reference genes for real-time PCR of cord blood mononuclear cells, differentiating endothelial progenitor cells, and mature endothelial cells, Exp Cell Res, 2018.

K. J. Livak and T. D. Schmittgen, Analysis of Relative Gene Expression Data Using Real-Time Quantitative PCR and the 2???CT Method, Methods, vol.25, issue.4, pp.402-410, 2001.

D. E. Discher, P. Janmey, Y. Wang, M. M. Stevens, and J. H. George, Tissue cells feel and respond to the stiffness of their substrate, Science, vol.310, issue.5751, pp.1135-1138, 2005.

M. P. Lutolf and J. A. Hubbell, Synthetic biomaterials as instructive extracellular microenvironments for morphogenesis in tissue engineering, Nat Biotechnol, vol.23, issue.1, pp.47-55, 2005.

S. P. Massia and J. A. Hubbell, An RGD spacing of 440 nm is sufficient for integrin alpha V beta 3-mediated fibroblast spreading and 140 nm for focal contact and stress fiber formation, J Cell Biol, vol.114, issue.5, pp.1089-1100, 1991.

L. Saux, G. Magenau, A. Böcking, T. Gaus, K. Gooding et al., The Relative Importance of Topography and RGD Ligand Density for Endothelial Cell Adhesion, PLoS ONE, vol.6, issue.7, p.21869, 2011.

C. Chollet, C. Labrugère, and M. Durrieu, Influence de la densité de peptides RGD greffés en surface de polyéthylène téréphtalate sur l'attachement des MC3T3, IRBM, vol.29, issue.1, pp.7-12, 2008.

H. Jin, A homing mechanism for bone marrow-derived progenitor cell recruitment to the neovasculature, J Clin Invest, vol.116, issue.3, pp.652-62, 2006.

C. Igreja, R. Fragoso, F. Caiado, N. Clode, A. Henriques et al.,

, Detailed molecular characterization of cord blood-derived endothelial progenitors, Exp Hematol, vol.36, issue.2, pp.193-194, 2008.

H. Bompais, J. Chagraoui, X. Canron, M. Crisan, X. H. Liu et al., Human endothelial cells derived from circulating progenitors display specific functional properties compared with mature vessel wall endothelial cells, Blood, vol.103, issue.7, pp.2577-2584, 2004.

W. Qiao, L. Niu, Z. Liu, T. Qiao, and C. Liu, Endothelial nitric oxide synthase as a marker for human endothelial progenitor cells, Tohoku J Exp Med, vol.221, issue.1, p.19, 2010.

N. F. Huang, B. Patlolla, O. Abilez, H. Sharma, J. Rajadas et al., A matrix micropatterning platform for cell localization and stem cell fate determination, Acta Biomater, vol.6, issue.12, pp.4614-4635, 2010.

N. F. Huang, E. S. Lai, A. Ribeiro, S. Pan, B. L. Pruitt et al., Spatial patterning of endothelium modulates cell morphology, adhesiveness and transcriptional signature, Biomaterials, vol.34, issue.12, pp.2928-2965, 2013.

S. Kusuma, Q. Smith, A. Facklam, and S. Gerecht, Micropattern size-dependent endothelial differentiation from a human induced pluripotent stem cell line: Micropattern size-dependent endothelial differentiation from a human induced pluripotent stem cell line, J Tissue Eng Regen Med, vol.11, issue.3, pp.855-61, 2017.

Y. C. Wang and C. Ho, Micropatterning of proteins and mammalian cells on biomaterials, FASEB J, vol.18, issue.3, pp.525-532, 2004.

R. F. Ankeny, C. J. Ankeny, R. M. Nerem, and H. Jo, Fluid shear stress induces differentiation of circulating phenotype endothelial progenitor cells, Am J Physiol-Cell Physiol, vol.303, issue.6, pp.589-91, 2012.

M. G. Angelos, M. A. Brown, L. L. Satterwhite, V. W. Levering, N. T. Shaked et al., Dynamic Adhesion of Umbilical Cord Blood Endothelial Progenitor Cells under Laminar Shear Stress, Biophys J, vol.99, issue.11, pp.3545-54, 2010.

M. Cheng, X. Guan, H. Li, X. Cui, X. Zhang et al., Shear Stress Regulates Late EPC Differentiation via Mechanosensitive Molecule-Mediated Cytoskeletal Rearrangement, PLoS ONE, vol.8, issue.7, p.67675, 2013.

D. M. Smadja, I. Bièche, D. Helley, I. Laurendeau, G. Simonin et al.,

, Increased VEGFR2 expression during human late endothelial progenitor cells expansion enhances in vitro angiogenesis with up-regulation of integrin ? 6, J Cell Mol Med, vol.11, issue.5, pp.1149-61, 2007.

P. De-la-puente, B. Muz, F. Azab, A. Azab, T. Kokubo et al., Integrin alpha(v)beta 3 as a target in the prevention of neointimal hyperplasia, Clin Cancer Res, vol.19, issue.13, pp.33-38, 2007.

S. Obi, K. Yamamoto, N. Shimizu, S. Kumagaya, T. Masumura et al., Fluid shear stress induces arterial differentiation of endothelial progenitor cells, J Appl Physiol Bethesda Md, vol.106, issue.1, p.203, 1985.

I. Petit, D. Jin, and S. Rafii, The SDF-1-CXCR4 signaling pathway: a molecular hub modulating neo-angiogenesis, Trends Immunol, vol.28, issue.7, pp.299-307, 2007.

S. Chua, J. Sheu, Y. Chen, L. Chang, C. Sun et al., Sitagliptin therapy enhances the number of circulating angiogenic cells and angiogenesis-evaluations in vitro and in the rat critical limb ischemia model, Cytotherapy, vol.15, issue.9, pp.1148-63, 2013.

I. Adipurnama, M. Yang, T. Ciach, and B. Butruk-raszeja, Surface modification and endothelialization of polyurethane for vascular tissue engineering applications: a review, Biomater Sci, vol.5, issue.1, pp.22-37, 2017.

T. Aper, A. Haverich, and O. Teebken, New developments in tissue engineering of vascular prosthetic grafts, VASA-J Vasc Dis, vol.38, issue.2, pp.99-122, 2009.

P. Zilla, D. Bezuidenhout, P. Human, E. Goh, E. Wong et al., Current concepts and new developments for autologous in vivo endothelialisation of biomaterials for intravascular applications, Avci-Adali M, vol.28, pp.157-176, 2007.

S. E. Greenwald and C. L. Berry, Improving vascular grafts: the importance of mechanical and haemodynamic properties, J Pathol, vol.190, issue.3, pp.292-301, 2000.

J. Eggermann, Endothelial progenitor cell culture and differentiation in vitro: a methodological comparison using human umbilical cord blood, Cardiovasc Res, vol.58, issue.2, pp.478-86, 2003.

K. Dheda, J. F. Huggett, S. A. Bustin, M. A. Johnson, G. Rook et al., Determination of stable housekeeping genes, differentially regulated target genes and sample integrity: BestKeeper-Excel-based tool using pair-wise correlations, Biotechnol Lett, vol.37, issue.1, pp.509-515, 2004.

L. Ponsonnet, K. Reybier, N. Jaffrezic, V. Comte, C. Lagneau et al., Relationship between surface properties (roughness, wettability) of titanium and titanium alloys and cell behaviour, Mater Sci Eng C, vol.23, issue.4, pp.551-60, 2003.

M. Dettin, A. Zamuner, M. Roso, G. Iucci, V. Samouillan et al.,

, Facile and selective covalent grafting of an RGD-peptide to electrospun scaffolds improves HUVEC adhesion: FACILE AND SELECTIVE COVALENT GRAFTING, J Pept Sci, vol.21, issue.10, pp.786-95, 2015.

R. Sinha, L. Gac, S. Verdonschot, N. Van-den-berg, A. Koopman et al., Endothelial cell alignment as a result of anisotropic strain and flow induced shear stress combinations. Sci Rep, vol.6, p.29510, 2016.

Y. Suzuki, K. Yamamoto, J. Ando, K. Matsumoto, T. Matsuda et al., Arterial shear stress augments the differentiation of endothelial progenitor cells adhered to VEGF-bound surfaces, Biochem Biophys Res Commun, vol.423, issue.1, 2012.

, RT-PCR versus immunohistochemistry for correlation and quantification of ERCC1, BRCA1, TUBB3 and RRM1 in NSCLC. Lung Cancer, vol.75, pp.306-318, 2012.

H. Ohnishi, H. Sasaki, Y. Nakamura, S. Kato, K. Ando et al., Regulation of cell shape and adhesion by CD34, Cell Adhes Migr, vol.7, issue.5, pp.426-459, 2013.

K. Vallières, É. Petitclerc, and G. Laroche, On the ability of imatinib mesylate to inhibit smooth muscle cell proliferation without delaying endothelialization: An in vitro study, Vascul Pharmacol, vol.51, issue.1, pp.50-56, 2009.

Y. J. Park, S. Min, I. M. Jung, T. Lee, J. Ha et al., Different Responses of Neointimal Cells to Imatinib Mesylate and Rapamycin Compared with Normal Vascular Smooth Muscle Cells. Vasc Spec Int, vol.30, pp.5-10, 2014.

C. Royer, A. Bégin, L. Plawinski, M. -. , C. Durrieu et al., Differentiation of Mononuclear Cells from Cord Blood in Endothelial Progenitors Cells onto Bioactive Poly (ethylene terephthalate) Film, 28th European Conference on Biomaterials, 2017.

C. Royer, L. Plawinski, C. Ayela, G. Laroche, and M. Durrieu, Impact of peptides micro-patterning on Endothelial Progenitors and Endothelial Mature Cells

C. Royer, P. Chevallier, L. Plawinski, G. Laroche, and M. Durrieu, Grafting of bioactive molecules on PET to recruit endothelials progenitor cells and enhance adhesion of endothelial cells, European Symposium on Vascular Biomaterials, pp.16-17, 2015.

C. Royer, L. Plawinski, G. Laroche, and M. Durrieu, Grafting of bioactive molecules on PET to recruit endothelials progenitor cells and enhance adhesion of endothelial cells, Cracovie, Pologne] Contribution dans des conférences nationales 1. Seconde place au concours Ma thèse en 180 secondes à la faculté de Sciences et Génie de l'Université Laval, 2015.

C. Royer, A. Bégin, L. P. , C. Chanseau, M. -. et al., Differentiation of Mononuclear Cells from Cord Blood in Endothelial Progenitor Cells onto Bioactive Poly (ethylene terephthalate) Film, vol.17

C. Royer, L. Plawinski, C. Chanseau, M. -. , C. Durrieu et al., Two-dimensional micropatterning of bioactive molecules to recruit and differentiate endothelial cell progenitors: Potential for endothelialization of poly (ethylene terephthalate)

C. Royer, L. Plawinski, C. Ayela, G. Laroche, and M. Durrieu, Fabrication of innovative vascular biomaterials for endothelial progenitor cells recruitment and differentiation, 2016.

C. Royer, L. Plawinski, C. Chanseau, L. Bordenave, G. Laroche et al., Spatially controlled simultaneous patterning of multiple biomolecules to guide endothelial progenitor cells adhesion and differentiation, Communication orale, Journée scientifique de la FR TecSan, 2016.

C. Royer, L. Plawinski, G. Laroche, and M. Durrieu, Surface modification of polymer with innovative biomolecules to improve vascular prostheses hemocompatibility, 2016.

C. Royer, P. Chevallier, L. Plawinski, G. Laroche, and M. Durrieu, Impact of micropatterned PET with peptides on Endothelial Progenitors differentiation and Endothelial Cells adhesion, 2016.

C. Royer, P. Chevallier, L. Plawinski, G. Laroche, and M. Durrieu, Endothelialization of PET: Grafting of bioactive molecules to recruit endothelials progenitor cells and enhance adhesion of endothelial cells