T. Ahsan and R. M. Nerem, Fluid shear stress promotes an endothelial-like phenotype during the early differentiation of embryonic stem cells, Tissue Eng. A, vol.16, pp.3547-3553, 2010.

J. Ando and K. Yamamoto, Vascular mechanobiology: endothelial cell responses to fluid shear stress, Circ. J. Off. J. Jpn. Circ. Soc, vol.73, 1983.

K. Bai, Y. Huang, X. Jia, Y. Fan, and W. Wang, Endothelium oriented differentiation of bone marrow mesenchymal stem cells under chemical and mechanical stimulations, J. Biomech, vol.43, pp.1176-1181, 2010.

A. M. Barradas, V. Monticone, M. Hulsman, C. Danoux, H. Fernandes et al., Molecular mechanisms of biomaterial-driven osteogenic differentiation in human mesenchymal stromal cells, Integr. Biol, vol.5, pp.920-931, 2013.

V. Bassaneze, V. G. Barauna, C. Lavini-ramos, J. Kalil, I. T. Schettert et al., Shear stress induces nitric oxidemediated vascular endothelial growth factor production in human adipose tissue mesenchymal stem cells, Stem Cells Dev, vol.19, pp.371-378, 2010.

D. A. Bruzewicz, A. P. Mcguigan, and G. M. Whitesides, Fabrication of a modular tissue construct in a microfluidic chip, Lab. Chip, vol.8, pp.663-671, 2008.

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, p.67675, 2013.

P. F. Davies, Hemodynamic shear stress and the endothelium in cardiovascular pathophysiology, Nat. Clin. Pract. Cardiovasc. Med, vol.6, pp.16-26, 2009.

J. Dong, Y. Gu, C. Li, C. Wang, Z. Feng et al., Response of mesenchymal stem cells to shear stress in tissue-engineered vascular grafts, Acta Pharmacol. Sin, vol.30, pp.530-536, 2009.

Y. Du, M. Ghodousi, H. Qi, N. Haas, W. Xiao et al., Sequential assembly of cell-laden hydrogel constructs to engineer vascular-like microchannels, Biotechnol. Bioeng, vol.108, pp.1693-1703, 2011.

R. El-omar, J. Beroud, J. Stoltz, P. Menu, E. Velot et al., Umbilical cord mesenchymal stem cells: the new gold standard for mesenchymal stem cell-based therapies?, Tissue Eng. B Rev, vol.20, pp.523-544, 2014.
URL : https://hal.archives-ouvertes.fr/hal-01777817

G. C. Engelmayr, . Jr, V. L. Sales, J. E. Mayer, . Jr et al., Cyclic flexure and laminar flow synergistically accelerate mesenchymal stem cellmediated engineered tissue formation: Implications for engineered heart valve tissues, Biomaterials, vol.27, pp.6083-6095, 2006.

L. S. Ferreira, S. Gerecht, H. F. Shieh, N. Watson, M. A. Rupnick et al., Vascular progenitor cells isolated from human embryonic stem cells give rise to endothelial and smooth muscle like cells and form vascular networks in vivo, Circ. Res, vol.101, pp.286-294, 2007.

L. J. Fischer, S. Mcilhenny, T. Tulenko, N. Golesorkhi, P. Zhang et al., Endothelial differentiation of adipose-derived stem cells: effects of endothelial cell growth supplement and shear force, J. Surg. Res, vol.152, pp.157-166, 2009.

J. Folkman and C. Haudenschild, Angiogenesis in vitro, Nature, vol.288, pp.551-556, 1980.

S. Ghazanfari, M. Tafazzoli-shadpour, and M. A. Shokrgozar, Effects of cyclic stretch on proliferation of mesenchymal stem cells and their differentiation to smooth muscle cells, Biochem. Biophys. Res. Commun, vol.388, pp.601-605, 2009.

D. P. Giddens, C. K. Zarins, and S. Glagov, The role of fluid mechanics in the localization and detection of atherosclerosis, J. Biomech. Eng, vol.115, pp.588-594, 1993.

Z. Gong and L. E. Niklason, Small-diameter human vessel wall engineered from bone marrow-derived mesenchymal stem cells (hMSCs), FASEB J, vol.22, pp.1635-1648, 2008.

N. Haghighipour, S. Heidarian, M. A. Shokrgozar, and N. Amirizadeh, Differential effects of cyclic uniaxial stretch on human mesenchymal stem cell into skeletal muscle cell, Cell Biol. Int, vol.36, pp.669-675, 2012.

D. W. Hamilton, T. M. Maul, and D. A. Vorp, Characterization of the response of bone marrow-derived progenitor cells to cyclic strain: implications for vascular tissue-engineering applications, Tissue Eng, vol.10, pp.361-369, 2004.

D. A. Hoey, S. Tormey, S. Ramcharan, F. J. O'brien, and C. R. Jacobs, Primary cilia-mediated mechanotransduction in human mesenchymal stem cells, Stem Cells, vol.30, pp.2561-2570, 2012.

B. D. Hoffman, C. Grashoff, and M. A. Schwartz, Dynamic molecular processes mediate cellular mechanotransduction, Nature, vol.475, pp.316-323, 2011.

N. F. Huang and S. Li, Mesenchymal stem cells for vascular regeneration, Regen. Med, vol.3, pp.877-892, 2008.

A. H. Huang and L. E. Niklason, Engineering of arteries in vitro, Cell. Mol. Life Sci, vol.71, pp.2103-2118, 2014.

Y. Huang, X. Jia, K. Bai, X. Gong, and Y. Fan, Effect of fluid shear stress on cardiomyogenic differentiation of rat bone marrow mesenchymal stem cells, Arch. Med. Res, vol.41, pp.497-505, 2010.

F. Huang, Z. Fang, X. Hu, L. Tang, S. Zhou et al., Overexpression of miR-126 promotes the differentiation of mesenchymal stem cells toward endothelial cells via activation of PI3K/Akt and MAPK/ERK pathways and release of paracrine factors, Biol. Chem, vol.394, pp.1223-1233, 2013.

A. H. Huang, Y. Lee, E. A. Calle, M. Boyle, B. C. Starcher et al., Design and use of a novel bioreactor for regeneration of biaxially stretched tissue-engineered vessels, Tissue Eng. Part C Methods, 2015.

J. Jang, S. W. Lee, S. H. Park, J. W. Shin, C. Mun et al., Combined effects of surface morphology and mechanical straining magnitudes on the differentiation of mesenchymal stem cells without using biochemical reagents, J. Biomed. Biotechnol, p.860652, 2011.

P. A. Janmey and C. A. Mcculloch, Cell mechanics: integrating cell responses to mechanical stimuli, Annu. Rev. Biomed. Eng, vol.9, pp.1-34, 2007.

K. Kaneko, M. Ito, Y. Naoe, A. Lacy-hulbert, and K. Ikeda, Integrin ?v in the mechanical response of osteoblast lineage cells, Biochem. Biophys. Res. Commun, vol.447, pp.352-357, 2014.

O. F. Khan and M. V. Sefton, Perfusion and characterization of an endothelial cell-seeded modular tissue engineered construct formed in a microfluidic remodeling chamber, Biomaterials, vol.31, pp.8254-8261, 2010.

O. F. Khan, M. D. Chamberlain, and M. V. Sefton, Toward an in vitro vasculature: differentiation of mesenchymal stromal cells within an endothelial cell-seeded modular construct in a microfluidic flow chamber, Tissue Eng. A, vol.18, pp.744-756, 2012.

D. H. Kim, S. Heo, S. Kim, J. W. Shin, S. H. Park et al., Shear stress magnitude is critical in regulating the differentiation of mesenchymal stem cells even with endothelial growth medium, Biotechnol. Lett, vol.33, pp.2351-2359, 2011.

N. Kobayashi, T. Yasu, H. Ueba, M. Sata, S. Hashimoto et al., Mechanical stress promotes the expression of smooth muscle-like properties in marrow stromal cells, Exp. Hematol, vol.32, pp.1238-1245, 2004.

N. Koike, D. Fukumura, O. Gralla, P. Au, J. S. Schechner et al., Tissue engineering: creation of long-lasting blood vessels, Nature, vol.428, pp.138-139, 2004.

M. R. Kreke, W. R. Huckle, and A. S. Goldstein, Fluid flow stimulates expression of osteopontin and bone sialoprotein by bone marrow stromal cells in a temporally dependent manner, Bone, vol.36, pp.1047-1055, 2005.

M. R. Kreke, L. A. Sharp, Y. Woo-lee, and A. S. Goldstein, Effect of intermittent shear stress on mechanotransductive signaling and osteoblastic differentiation of bone marrow stromal cells, Tissue Eng. A, vol.14, pp.529-537, 2008.

K. Kurpinski, J. Chu, D. Wang, and S. Li, Proteomic profiling of mesenchymal stem cell responses to mechanical strain and TGF-beta1, Cell. Mol. Bioeng, vol.2, pp.606-614, 2009.

K. Kurpinski, J. Park, R. G. Thakar, and S. Li, Regulation of vascular smooth muscle cells and mesenchymal stem cells by mechanical strain, Mol. Cell. Biomech, vol.3, pp.21-34, 2006.

F. Le-noble, D. Moyon, L. Pardanaud, L. Yuan, V. Djonov et al., Flow regulates arterial-venous differentiation in the chick embryo yolk sac, Development, vol.131, pp.361-375, 2004.

W. C. Lee, T. M. Maul, D. A. Vorp, J. P. Rubin, and K. G. Marra, Effects of uniaxial cyclic strain on adipose-derived stem cell morphology, proliferation, and differentiation, Biomech. Model. Mechanobiol, vol.6, pp.265-273, 2007.

W. S. Leong, S. C. Wu, M. Pal, C. Y. Tay, H. Yu et al., Cyclic tensile loading regulates human mesenchymal stem cell differentiation into neuron-like phenotype, J. Tissue Eng. Regen. Med, vol.6, issue.3, pp.68-79, 2012.

N. L'heureux, S. Pa?quet, R. Labbe, L. Germain, and F. A. Auger, A completely biological tissue-engineered human blood vessel, FASEB J, vol.12, pp.47-56, 1998.

N. L'heureux, N. Dusserre, G. Konig, B. Victor, P. Keire et al., Human tissue-engineered blood vessels for adult arterial revascularization, Nat. Med, vol.12, pp.361-365, 2006.

N. L'heureux, N. Dusserre, A. Marini, S. Garrido, L. De-la-fuente et al., Technology Insight: the evolution of tissue-engineered vascular grafts-from research to clinical practice, Nat. Clin. Pract. Cardiovasc. Med, vol.4, pp.389-395, 2007.

J. Y. Lim, A. E. Loiselle, J. S. Lee, Y. Zhang, J. D. Salvi et al., Optimizing the osteogenic potential of adult stem cells for skeletal regeneration, J. Orthop. Res, vol.29, pp.1627-1633, 2011.

Y. Liu and O. K. Lee, In search of the pivot point of mechanotransduction: mechanosensing of stem cells, Cell Transplant, vol.23, pp.1-11, 2014.

T. M. Maul, D. W. Chew, A. Nieponice, and D. A. Vorp, Mechanical stimuli differentially control stem cell behavior: morphology, proliferation, and differentiation, Biomech. Model. Mechanobiol, vol.10, pp.939-953, 2011.

S. H. Mcbride, T. Falls, and M. L. Tate, Modulation of stem cell shape and fate B: mechanical modulation of cell shape and gene expression, Tissue Eng. A, vol.14, pp.1573-1580, 2008.

V. Mironov, V. Kasyanov, and R. R. Markwald, Organ printing: from bioprinter to organ biofabrication line, Curr. Opin. Biotechnol, vol.22, pp.667-673, 2011.

A. Nieponice, T. M. Maul, J. M. Cumer, L. Soletti, and D. A. Vorp, Mechanical stimulation induces morphological and phenotypic changes in bone marrow-derived progenitor cells within a three-dimensional fibrin matrix, J. Biomed. Mater. Res. A, vol.81, pp.523-530, 2007.

E. D. O'cearbhaill, M. A. Punchard, M. Murphy, F. P. Barry, P. E. Mchugh et al., Response of mesenchymal stem cells to the biomechanical environment of the endothelium on a flexible tubular silicone substrate, Biomaterials, vol.29, pp.1610-1619, 2008.

E. D. O'cearbhaill, M. Murphy, F. Barry, P. E. Mchugh, and V. Barron, Behavior of human mesenchymal stem cells in fibrin-based vascular tissue engineering constructs, Ann. Biomed. Eng, vol.38, pp.649-657, 2010.

J. Oswald, S. Boxberger, B. Jørgensen, S. Feldmann, G. Ehninger et al., Mesenchymal stem cells can be differentiated into endothelial cells in vitro, Stem Cells, vol.22, pp.377-384, 2004.

G. K. Owens, Regulation of differentiation of vascular smooth muscle cells, Physiol. Rev, vol.75, pp.487-517, 1995.

J. S. Park, J. S. Chu, C. Cheng, F. Chen, D. Chen et al., Differential effects of equiaxial and uniaxial strain on mesenchymal stem cells, Biotechnol. Bioeng, vol.88, pp.359-368, 2004.

J. S. Park, N. F. Huang, K. T. Kurpinski, S. Patel, S. Hsu et al., Mechanobiology of mesenchymal stem cells and their use in cardiovascular repair, Front. Biosci, vol.12, pp.5098-5116, 2007.

J. J. Paszkowiak and A. Dardik, Arterial wall shear stress: observations from the bench to the bedside, Vasc. Endovascular Surg, vol.37, pp.47-57, 2003.

H. A. Praetorius and K. R. Spring, A physiological view of the primary cilium, Annu. Rev. Physiol, vol.67, pp.515-529, 2005.

G. M. Riha, X. Wang, H. Wang, H. Chai, H. Mu et al., Cyclic strain induces vascular smooth muscle cell differentiation from murine embryonic mesenchymal progenitor cells, Surgery, vol.141, pp.394-402, 2007.

P. Satir, L. B. Pedersen, and S. T. Christensen, The primary cilium at a glance, J. Cell Sci, vol.123, pp.499-503, 2010.

M. A. Schwartz and R. K. Assoian, Integrins and cell proliferation: regulation of cyclin-dependent kinases via cytoplasmic signaling pathways, J. Cell Sci, vol.114, pp.2553-2560, 2001.

D. G. Seifu, A. Purnama, K. Mequanint, and D. Mantovani, Smalldiameter vascular tissue engineering, Nat. Rev. Cardiol, vol.10, pp.410-421, 2013.

N. Shah, Y. Morsi, and R. Manasseh, From mechanical stimulation to biological pathways in the regulation of stem cell fate, Cell Biochem. Funct, vol.32, pp.309-325, 2014.

T. Shin'oka, Y. Imai, and Y. Ikada, Transplantation of a tissue-engineered pulmonary artery, N. Engl. J. Med, vol.344, pp.532-533, 2001.

S. Shojaei, M. Tafazzoli-shahdpour, M. A. Shokrgozar, and N. Haghighipour, Effects of mechanical and chemical stimuli on differentiation of human adipose-derived stem cells into endothelial cells, Int. J. Artif. Organs, vol.36, pp.663-673, 2013.

J. Y. Shyy and S. Chien, Role of integrins in endothelial mechanosensing of shear stress, Circ. Res, vol.91, pp.769-775, 2002.

Q. Smith and S. Gerecht, Going with the flow: microfluidic platforms in vascular tissue engineering, Curr. Opin. Chem. Eng, vol.3, pp.42-50, 2014.

S. Stolberg and K. E. Mccloskey, Can shear stress direct stem cell fate?, Biotechnol. Prog, vol.25, pp.10-19, 2009.

B. V. Udelsman, R. Khosravi, K. S. Miller, E. W. Dean, M. R. Bersi et al., Characterization of evolving biomechanical properties of tissue engineered vascular grafts in the arterial circulation, J. Biomech, vol.47, pp.2070-2079, 2014.

N. E. Vlahakis, B. A. Young, A. Atakilit, A. E. Hawkridge, R. B. Issaka et al., Integrin alpha9beta1 directly binds to vascular endothelial growth factor (VEGF)-A and contributes to VEGF-A-induced angiogenesis, J. Biol. Chem, vol.282, pp.15187-15196, 2007.

H. Wang, G. M. Riha, S. Yan, M. Li, H. Chai et al., Shear stress induces endothelial differentiation from a murine embryonic mesenchymal progenitor cell line, Arterioscler. Thromb. Vasc. Biol, vol.25, pp.1817-1823, 2005.

H. Wang, M. Li, P. H. Lin, Q. Yao, and C. Chen, Fluid shear stress regulates the expression of TGF-beta1 and its signaling molecules in mouse embryo mesenchymal progenitor cells, J. Surg. Res, vol.150, pp.266-270, 2008.

C. Wang, L. Cen, S. Yin, Q. Liu, W. Liu et al., A small diameter elastic blood vessel wall prepared under pulsatile conditions from polyglycolic acid mesh and smooth muscle cells differentiated from adiposederived stem cells, Biomaterials, vol.31, pp.621-630, 2010.

P. D. Weinberg, R. Ethier, and C. , Twenty-fold difference in hemodynamic wall shear stress between murine and human aortas, J. Biomech, vol.40, pp.1594-1598, 2007.

Z. Wu, K. Wong, M. Glogauer, R. P. Ellen, and C. A. Mcculloch, Regulation of stretch-activated intracellular calcium transients by actin filaments, Biochem. Biophys. Res. Commun, vol.261, pp.419-425, 1999.

C. Wu, Y. Chao, C. Chen, S. Chien, Y. Chen et al., Synergism of biochemical and mechanical stimuli in the differentiation of human placenta-derived multipotent cells into endothelial cells, J. Biomech, vol.41, pp.813-821, 2008.

K. Yamamoto, T. Sokabe, T. Watabe, K. Miyazono, J. K. Yamashita et al., Fluid shear stress induces differentiation of Flk-1-positive embryonic stem cells into vascular endothelial cells in vitro, Am. J. Physiol. Heart Circ. Physiol, vol.288, pp.1915-1924, 2005.

H. Yamawaki, S. Pan, R. T. Lee, and B. C. Berk, Fluid shear stress inhibits vascular inflammation by decreasing thioredoxin-interacting protein in endothelial cells, J. Clin. Invest, vol.115, pp.733-738, 2005.

R. Yao and J. Y. Wong, The effects of mechanical stimulation on controlling and maintaining marrow stromal cell differentiation into vascular smooth muscle cells, J. Biomech. Eng, vol.137, p.20907, 2015.

A. B. Yeatts, E. M. Geibel, F. F. Fears, and J. P. Fisher, Human mesenchymal stem cell position within scaffolds influences cell fate during dynamic culture, Biotechnol. Bioeng, vol.109, pp.2381-2391, 2012.

L. Yuan, N. Sakamoto, G. Song, and M. Sato, High-level shear stress stimulates endothelial differentiation and VEGF secretion by human mesenchymal stem cells, Cell. Mol. Bioeng, vol.6, pp.220-229, 2013.

P. Zhang, J. Baxter, K. Vinod, T. N. Tulenko, D. Muzio et al., Endothelial differentiation of amniotic fluid-derived stem cells: synergism of biochemical and shear force stimuli, Stem Cells Dev, vol.18, pp.1299-1308, 2009.

P. Zhang, N. Moudgill, E. Hager, N. Tarola, C. Dimatteo et al., Endothelial differentiation of adiposederived stem cells from elderly patients with cardiovascular disease, Stem Cells Dev, vol.20, pp.977-988, 2011.

Y. Zhao, S. Zhang, J. Zhou, J. Wang, M. Zhen et al., The development of a tissue-engineered artery using decellularized scaffold and autologous ovine mesenchymal stem cells, Biomaterials, vol.31, pp.296-307, 2010.

W. Zheng, Y. Xie, W. Zhang, D. Wang, W. Ma et al., Fluid flow stress induced contraction and re-spread of mesenchymal stem cells: a microfluidic study, Integr. Biol, vol.4, pp.1102-1111, 2012.

J. Zhou and L. E. Niklason, Microfluidic artificial "vessels" for dynamic mechanical stimulation of mesenchymal stem cells, Integr. Biol, vol.4, p.1487, 2012.

R. Kuang, Z. Zhang, X. Jin, J. Hu, S. Shi et al., Nanofibrous spongy microspheres for the delivery of hypoxia-primed human dental pulp stem cells to regenerate vascularized dental pulp, Acta Biomater, vol.33, pp.225-234, 2016.

P. S. Briquez, L. E. Clegg, M. M. Martino, F. Gabhann, . Mac et al., Design principles for therapeutic angiogenic materials, Nat. Rev. Mater, 2016.

E. Dondossola, B. Holzapfel, S. Alexander, S. Filippini, D. Hutmacher et al., Examination of the foreign body response to biomaterials by nonlinear intravital microscopy, Nat. Biomed. Eng

C. D. Mccaig, B. Song, and A. M. Rajnicek, Electrical dimensions in cell science, J. Cell Sci, vol.122, pp.4267-4276, 2009.

C. D. Mccaig, A. M. Rajnicek, B. Song, and M. Zhao, Controlling cell behavior electrically: current views and future potential, Physiol. Rev, vol.85, pp.943-978, 2005.

N. Weber, Y. S. Lee, S. Shanmugasundaram, M. Jaffe, and T. L. Arinzeh, Characterization and in vitro cytocompatibility of piezoelectric electrospun scaffolds, Acta Biomater, vol.6, pp.3550-3556, 2010.

Z. Hu, M. Tian, B. Nysten, and A. M. Jonas, Regular arrays of highly ordered ferroelectric polymer nanostructures for non-volatile low-voltage memories, Nat. Mater, vol.8, pp.62-67, 2009.

S. Huang, W. A. Ye-e, W. C. Tjiu, Y. Liu, M. Kotaki et al., Electrospinning of polyvinylidene difluoride with carbon nanotubes: synergistic effects of extensional force and interfacial interaction on crystalline structures, Langmuir ACS J. surfaces colloids, vol.24, pp.13621-13626, 2008.

M. Li, H. J. Wondergem, M. J. Spijkman, K. Asadi, I. Katsouras et al., Revisiting the ?-phase of poly(vinylidene fluoride) for solution-processed ferroelectric thin films, Nat. Mater, vol.12, pp.433-438, 2013.

Y. Liu, J. Lu, H. Li, J. Wei, and X. Li, Engineering blood vessels through micropatterned co-culture of vascular endothelial and smooth muscle cells on bilayered electrospun fibrous mats with pDNA inoculation, Acta Biomater, vol.11, pp.114-125, 2015.

E. G. Fine, R. F. Valentini, R. Bellamkonda, and P. Aebischer,

R. E. Omar, Y. Xiong, G. Dostert, H. Louis, M. Gentils et al., Immunomodulation of endothelial differentiated mesenchymal stromal cells: impact on T and NK cells, Immunol. Cell Biol, vol.94, pp.342-356, 2016.
URL : https://hal.archives-ouvertes.fr/hal-01451665

J. A. Ankrum, J. F. Ong, and J. M. Karp, Mesenchymal stem cells: immune evasive, not immune privileged, Nat. Biotechnol, vol.32, pp.252-260, 2014.

F. Guilak, D. M. Cohen, B. T. Estes, J. M. Gimble, W. Liedtke et al., Control of stem cell fate by physical interactions with the extracellular matrix, Cell Stem Cell, vol.5, pp.17-26, 2009.

M. M. Stevens and J. H. George, Exploring and engineering the cell surface interface, Science, vol.310, pp.1135-1138, 2005.

J. Ringe and M. Sittinger, Regenerative medicine: selecting the right biological scaffold for tissue engineering, Nat. Rev. Rheumatol, vol.10, pp.388-389, 2014.

K. Mark, J. Park, S. Bauer, and P. Schmuki, Nanoscale engineering of biomimetic surfaces: cues from the extracellular matrix, Cell Tissue Res, vol.339, pp.131-153, 2010.

B. M. Gillette, J. A. Jensen, M. Wang, J. Tchao, and S. K. Sia, Dynamic hydrogels: switching of 3D microenvironments using two-component naturally derived extracellular matrices, Adv. Mater, vol.22, pp.686-691, 2010.

E. S. Place, N. D. Evans, and M. M. Stevens, Complexity in biomaterials for tissue engineering, vol.8, pp.457-470, 2009.

M. C. Moore, V. Pandolfi, and P. S. Mcfetridge, Novel human-derived extracellular matrix induces in vitro and in vivo vascularization and inhibits fibrosis, Biomaterials, vol.49, pp.37-46, 2015.

A. Can and S. Karahuseyinoglu, Concise review: human umbilical cord stroma with regard to the source of fetus-derived stem cells, Stem Cells, vol.25, pp.2886-2895, 2007.

H. Hao, G. Chen, J. Liu, D. Ti, Y. Zhao et al., Culturing on Wharton's jelly extract delays mesenchymal stem cell senescence through p53 and p16INK4a/pRb pathways, PLoS One, vol.8, p.58314, 2013.

N. Berthelemy, H. Kerdjoudj, C. Gaucher, P. Schaaf, J. Stoltz et al.,

P. Voegel and . Menu, Polyelectrolyte films boost progenitor cell differentiation into endothelium-like monolayers, Adv. Mater. Deerfield Beach Fla, vol.20, pp.2674-2678, 2008.
URL : https://hal.archives-ouvertes.fr/hal-02164025

I. N. Sneddon, The relation between load and penetration in the axi-symmetric Boussinesq problem for a punch of arbitrary profile, Int. J. Eng. Sci, vol.3, pp.47-57, 1965.

N. Gavara and R. S. Chadwick, Determination of the elastic moduli of thin samples and adherent cells using conical atomic force microscope tips, Nat. Nanotechnol, vol.7, pp.733-736, 2012.

P. Polyakov, C. Soussen, J. Duan, J. F. Duval, D. Brie et al., Automated force volume image processing for biological samples, PLoS One, vol.6, p.18887, 2011.
URL : https://hal.archives-ouvertes.fr/hal-00584565

R. Rago, J. Mitchen, and G. Wilding, DNA fluorometric assay in 96-well tissue culture plates using Hoechst 33258 after cell lysis by freezing in distilled water, Anal. Biochem, vol.191, pp.31-34, 1990.

C. M. Frendl, S. M. Tucker, N. A. Khan, M. B. Esch, S. Kanduru et al., Endothelial retention and phenotype on carbonized cardiovascular implant surfaces, Biomaterials, vol.35, pp.7714-7723, 2014.

N. Rajan, J. Habermehl, M. Coté, C. J. Doillon, and D. Mantovani, Preparation of ready-to-use, storable and reconstituted type I collagen from rat tail tendon for tissue engineering applications, Nat. Protoc, vol.1, pp.2753-2758, 2006.

Y. Han, R. Tao, T. Sun, J. Chai, G. Xu et al., Optimization of human umbilical cord mesenchymal stem cell isolation and culture methods, Cytotechnology, vol.65, pp.819-827, 2013.

K. Sobolewski, A. Ma?kowski, E. Ba?-kowski, and S. Jaworski, Wharton's jelly as a reservoir of peptide growth factors, Placenta, vol.26, pp.747-752, 2005.

W. L. Murphy, T. C. Mcdevitt, and A. J. Engler, Materials as stem cell regulators, Nat. Mater, vol.13, pp.547-557, 2014.

K. Wingate, W. Bonani, Y. Tan, S. J. Bryant, and W. Tan, Compressive elasticity of three-dimensional nanofiber matrix directs mesenchymal stem cell differentiation to vascular cells with endothelial or smooth muscle cell markers, Acta Biomater, vol.8, pp.1440-1449, 2012.

T. D. Ross, B. G. Coon, S. Yun, N. Baeyens, K. Tanaka et al., Integrins in mechanotransduction, Curr. Opin. Cell Biol, vol.25, pp.613-618, 2013.

D. Docheva, C. Popov, W. Mutschler, and M. Schieker, Human mesenchymal stem cells in contact with their environment: surface characteristics and the integrin system, J. Cell Mol. Med, vol.11, pp.21-38, 2007.

N. J. Turner, M. O. Murphy, C. M. Kielty, C. A. Shuttleworth, R. A. Black et al., Alpha2(VIII) collagen substrata enhance endothelial cell retention under acute shear stress flow via an alpha2beta1 integrin-dependent mechanism: an in vitro and in vivo study, Circulation, vol.114, pp.820-829, 2006.

K. Warstat, D. Meckbach, M. Weis-klemm, A. Hack, G. Klein et al., TGF-beta enhances the integrin alpha2beta1-mediated attachment of mesenchymal stem cells to type I collagen, Stem Cells Dev, vol.19, pp.645-656, 2010.

D. Mozaffarian, E. J. Benjamin, A. S. Go, D. K. Arnett, M. J. Blaha et al., American heart association statistics committee and stroke statistics subcommittee, Circulation, vol.133, pp.38-360, 2016.

E. Benrashid, C. C. Mccoy, L. M. Youngwirth, J. Kim, R. J. Manson et al., Tissue engineered vascular grafts: origins, development, and current strategies for clinical application, Methods San Diego Calif, vol.99, pp.13-19, 2016.

D. G. Seifu, A. Purnama, K. Mequanint, and D. Mantovani, Small-diameter vascular tissue engineering, Nat. Rev. Cardiol, vol.10, pp.410-421, 2013.

J. T. Krawiec and D. A. Vorp, Adult stem cell-based tissue engineered blood vessels: a review, Biomaterials, vol.33, pp.3388-3400, 2012.

Y. Zhao, S. Zhang, J. Zhou, J. Wang, M. Zhen et al., The development of a tissue-engineered artery using decellularized scaffold and autologous ovine mesenchymal stem cells, Biomaterials, vol.31, pp.296-307, 2010.

M. T. Koobatian, S. Row, R. J. Smith, C. Koenigsknecht, S. T. Andreadis et al., Successful endothelialization and remodeling of a cell-free smalldiameter arterial graft in a large animal model, Biomaterials, vol.76, pp.344-358, 2016.

M. M. Martino, S. Brkic, E. Bovo, M. Burger, D. J. Schaefer et al., Extracellular matrix and growth factor engineering for controlled angiogenesis in regenerative medicine, Front. Bioeng. Biotechnol, vol.3, p.45, 2015.

M. J. Sherratt, D. V. Bax, S. S. Chaudhry, N. Hodson, J. R. Lu et al., Substrate chemistry influences the morphology and biological function of adsorbed extracellular matrix assemblies, Biomaterials, vol.26, pp.7192-7206, 2005.

D. N. Coakley, F. M. Shaikh, K. O'sullivan, E. G. Kavanagh, P. A. Grace et al., Comparing the endothelialisation of extracellular matrix bioscaffolds with coated synthetic vascular graft materials, Int. J. Surg. Lond. Engl, vol.25, pp.31-37, 2016.

A. D. Doyle, N. Carvajal, A. Jin, K. Matsumoto, and K. M. Yamada, Local 3D matrix microenvironment regulates cell migration through spatiotemporal dynamics of contractility-dependent adhesions, Nat. Commun, vol.6, p.8720, 2015.

Y. Han, Q. Xu, Z. Lu, and J. Wang, Cell adhesion on zein films under shear stress field, Colloids Surf. B Biointerfaces, vol.111, pp.479-485, 2013.

X. Gong, H. Liu, X. Ding, M. Liu, X. Li et al., Physiological pulsatile flow culture conditions to generate functional endothelium on a sulfated silk fibroin nanofibrous scaffold, Biomaterials, vol.35, pp.4782-4791, 2014.

S. E. Mcilhenny, E. S. Hager, D. J. Grabo, C. Dimatteo, I. M. Shapiro et al., Linear shear conditioning improves vascular graft retention of adipose-derived stem cells by upregulation of the alpha5beta1 integrin, Tissue Eng. Part A, vol.16, pp.245-255, 2010.

F. M. Watt and W. T. Huck, Role of the extracellular matrix in regulating stem cell fate, Nat. Rev. Mol. Cell Biol, vol.14, pp.467-473, 2013.

E. Tzima, M. Irani-tehrani, W. B. Kiosses, E. Dejana, D. A. Schultz et al., A mechanosensory complex that mediates the endothelial cell response to fluid shear stress, Nature, vol.437, pp.426-431, 2005.

J. Chlupac, E. Filova, J. Havlikova, R. Matejka, T. Riedel et al., The gene expression of human endothelial cells is modulated by subendothelial extracellular matrix proteins: short-term response to laminar shear stress, Tissue Eng. Part A, vol.20, pp.2253-2264, 2014.

N. Huebsch and D. J. Mooney, Inspiration and application in the evolution of biomaterials, Nature, vol.462, pp.426-432, 2009.

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

N. Rajan, J. Habermehl, M. Coté, C. J. Doillon, and D. Mantovani, Preparation of ready-to-use, storable and reconstituted type I collagen from rat tail tendon for tissue engineering applications, Nat. Protoc, vol.1, pp.2753-2758, 2006.

P. S. Amenta, S. Gay, A. Vaheri, and A. Martinez-hernandez, The extracellular matrix is an integrated unit: Ultrastructural localization of collagen types I, Coll. Relat. Res, vol.6, issue.86, pp.80021-80029, 1986.

M. C. Moore, V. Pandolfi, and P. S. Mcfetridge, Novel human-derived extracellular matrix induces in vitro and in vivo vascularization and inhibits fibrosis, Biomaterials, vol.49, pp.37-46, 2015.

G. Astori, E. Amati, F. Bambi, M. Bernardi, K. Chieregato et al., Platelet lysate as a substitute for animal serum for the ex-vivo expansion of mesenchymal stem/stromal cells: Present and future, Stem Cell Res. Ther, vol.7, p.93, 2016.

A. Can and S. Karahuseyinoglu, Concise review: Human umbilical cord stroma with regard to the source of fetus-derived stem cells, Stem Cells, vol.25, pp.2886-2895, 2007.

R. E. Omar, Y. Xiong, G. Dostert, H. Louis, M. Gentils et al., Immunomodulation of endothelial differentiated mesenchymal stromal cells: Impact on T and NK cells, 2015.
URL : https://hal.archives-ouvertes.fr/hal-01451665

H. Rammal, J. Beroud, M. Gentils, P. Labrude, P. Menu et al., Reversing charges or how to improve Wharton's jelly mesenchymal stem cells culture on polyelectrolyte multilayer films, Biomed. Mater. Eng, vol.23, pp.299-309, 2013.

J. Ringe and M. Sittinger, Regenerative medicine: Selecting the right biological scaffold for tissue engineering, Nat. Rev. Rheumatol, vol.10, pp.388-389, 2014.

, BME

D. Mozaffarian, E. J. Benjamin, and A. S. Go, Heart Disease and Stroke Statistics-2016 Update: A Report From the American Heart Association, Circulation, vol.133, pp.38-360, 2016.

D. G. Seifu, A. Purnama, K. Mequanint, and D. Mantovani, Small-diameter vascular tissue engineering, Nat. Rev. Cardiol, vol.10, pp.410-421, 2013.

S. Ravi and E. L. Chaikof, Biomaterials for vascular tissue engineering, Regen. Med, vol.5, p.107, 2010.

T. Sugiura, S. Tara, and H. Nakayama, Novel Bioresorbable Vascular Graft With Sponge-Type Scaffold as a Small-Diameter Arterial Graft, Ann. Thorac. Surg, vol.102, pp.720-727, 2016.

N. L'heureux, S. Pâquet, R. Labbé, L. Germain, and F. A. Auger, A completely biological tissueengineered human blood vessel, FASEB J, vol.12, pp.47-56, 1998.

N. L'heureux, N. Dusserre, and G. Konig, Human tissue-engineered blood vessels for adult arterial revascularization, Nat. Med, vol.12, pp.361-365, 2006.

A. H. Huang and L. E. Niklason, Engineering of arteries in vitro, Cell. Mol. Life Sci. CMLS, vol.71, pp.2103-2118, 2014.

C. Quint, Y. Kondo, R. J. Manson, J. H. Lawson, A. Dardik et al., Decellularized tissueengineered blood vessel as an arterial conduit, Proc. Natl. Acad. Sci, vol.108, pp.9214-9219, 2011.

H. Kerdjoudj, N. Berthelemy, and S. Rinckenbach, Small Vessel Replacement by Human Umbilical Arteries With Polyelectrolyte Film-Treated ArteriesIn Vivo Behavior, J. Am. Coll. Cardiol, vol.52, pp.1589-1597, 2008.

M. T. Koobatian, S. Row, S. Jr, R. J. Koenigsknecht, C. Andreadis et al., Successful endothelialization and remodeling of a cell-free small-diameter arterial graft in a large animal model, Biomaterials, vol.76, pp.344-358, 2016.

P. A. Cahill and E. M. Redmond, Vascular endothelium -Gatekeeper of vessel health, Atherosclerosis, vol.248, pp.97-109, 2016.

D. Wion, T. Christen, E. L. Barbier, and J. A. Coles, PO2 Matters in Stem Cell Culture, Cell Stem Cell, vol.5, pp.242-243, 2009.
URL : https://hal.archives-ouvertes.fr/inserm-00527773

N. Dahan, U. Sarig, and T. Bronshtein, Dynamic Autologous Reendothelialization of Small-Caliber Arterial Extracellular Matrix: A Preclinical Large Animal Study, Tissue Eng. Part A, vol.23, pp.69-79, 2017.

D. Mohebbi-kalhori, M. Rukhlova, A. Ajji, M. Bureau, and M. J. Moreno, A novel automated cellseeding device for tissue engineering of tubular scaffolds: design and functional validation, J. Tissue Eng. Regen. Med, vol.6, pp.710-720, 2012.

A. Mahara, S. Somekawa, and N. Kobayashi, Tissue-engineered acellular small diameter long-bypass grafts with neointima-inducing activity, Biomaterials, vol.58, pp.54-62, 2015.

P. Dan, É. Velot, G. Francius, P. Menu, and V. Decot, Human-derived extracellular matrix from Wharton's jelly: An untapped substrate to build up a standardized and homogeneous coating for vascular engineering, Acta Biomater, 2016.
URL : https://hal.archives-ouvertes.fr/hal-01691349

R. El-omar, Y. Xiong, and G. Dostert, Immunomodulation of endothelial differentiated mesenchymal stromal cells: impact on T and NK cells, Immunol. Cell Biol, 2015.
URL : https://hal.archives-ouvertes.fr/hal-01451665

H. Inoguchi, T. Tanaka, Y. Maehara, and T. Matsuda, The effect of gradually graded shear stress on the morphological integrity of a huvec-seeded compliant small-diameter vascular graft, Biomaterials, vol.28, pp.486-495, 2007.

J. T. Krawiec and D. A. Vorp, Adult stem cell-based tissue engineered blood vessels: A review, Biomaterials, vol.33, pp.3388-3400, 2012.

G. Fercana, D. Bowser, and M. Portilla, Platform Technologies for Decellularization, Tunic-Specific Cell Seeding, and In Vitro Conditioning of Extended Length, Small Diameter Vascular Grafts, Tissue Eng. Part C Methods, vol.20, pp.1016-1027, 2014.

S. De-valence, J. Tille, and D. Mugnai, Long term performance of polycaprolactone vascular grafts in a rat abdominal aorta replacement model, Biomaterials, vol.33, pp.38-47, 2012.

M. Ahmed, G. Hamilton, and A. M. Seifalian, The performance of a small-calibre graft for vascular reconstructions in a senescent sheep model, Biomaterials, vol.35, pp.9033-9040, 2014.

T. N. Mcallister, M. Maruszewski, and S. A. Garrido, Effectiveness of haemodialysis access with an autologous tissue-engineered vascular graft: a multicentre cohort study, Lancet Lond. Engl, vol.373, pp.1440-1446, 2009.

T. Fukunishi, C. A. Best, and T. Sugiura, :e0158555. lumière pour une implantation in vivo. Grace à cette stratégie on peut envisager de développer une banque tissulaire avec les artères ombilicales richement entourées de WJ-natif. Ces artères peuvent être conservées au moment de la naissance afin d'être disponibles pour chacun si nécessaire. Cependant, des études plus approfondies sont nécessaires pour caractériser les comportements cellulaires (adhésion, prolifération, alignement et différenciation) sur la WJnatif. Et la technique de conserver WJ-natif avec une épaisseur désirée est aussi importante afin de contrôler cette surface. En conclusion, la technique de retournement donne la possibilité d'utiliser divers coatings dans l'ITV, Tissue-Engineered Small Diameter Arterial Vascular Grafts from Cell-Free Nanofiber PCL/Chitosan Scaffolds in a Sheep Model, vol.11, 2016.

N. J. Turner, M. O. Murphy, C. M. Kielty, C. A. Shuttleworth, R. A. Black et al., Alpha2(VIII) collagen substrata enhance endothelial cell retention under acute shear stress flow via an alpha2beta1 integrin-dependent mechanism: an in vitro and in vivo study, Circulation, vol.114, pp.820-829, 2006.

J. Chlupá?, E. Filová, T. Riedel, M. Houska, E. Bry-da et al., Atta hment of human endothelial cells to polyester vascular grafts: pre-coating with adhesive protein assemblies and resistance to short-term shear stress, Physiol. Res. Acad. Sci. Bohemoslov, vol.63, pp.167-177, 2014.

G. Chen, Y. Lv, P. Guo, C. Lin, X. Zhang et al., Matrix mechanics and fluid shear stress control stem cells fate in three dimensional microenvironment, Curr. Stem Cell Res. Ther, vol.8, pp.313-323, 2013.

E. D. O'cear-haill, M. Murphy, F. Barry, P. E. Hugh, and V. Barro, Behavior of hu a mesenchymal stem cells in fibrin-based vascular tissue engineering constructs, Ann. Biomed. Eng, vol.38, pp.649-657, 2010.

D. D. Swartz and S. T. Andreadis, Animal models for vascular tissue-engineering, Curr. Opin. Biotechnol, vol.24, pp.916-925, 2013.

H. Kerdjoudj, N. Berthelemy, S. Rinckenbach, A. Kearney-schwartz, K. Montagne et al.,

P. Schaaf, J. Lacolley, J. Stoltz, P. Voegel, and . Menu, Small Vessel Replacement by Human Umbilical Arteries With Polyelectrolyte Film-Treated ArteriesIn Vivo Behavior, J. Am. Coll. Cardiol, vol.52, pp.1589-1597, 2008.

S. S. White, C. K. Zarins, D. P. Giddens, H. Bassiouny, F. Loth et al., Hemodynamic Patterns in Two Models of End-to-Side Vascular Graft Anastomoses: Effects of Pulsatility, Flow Division, Reynolds Number, and Hood Length, J. Biomech. Eng, vol.115, pp.104-111, 1993.

A. Mahara, S. Somekawa, N. Kobayashi, Y. Hirano, Y. Kimura et al., Tissue-engineered acellular small diameter long-bypass grafts with neointima-inducing activity, Biomaterials, vol.58, pp.54-62, 2015.

M. T. Koobatian, S. Row, R. J. Smith, C. Koenigsknecht, S. T. Andreadis et al., Successful endothelialization and remodeling of a cell-free small-diameter arterial graft in a large animal model, Biomaterials, vol.76, pp.344-358, 2016.

D. Mozaffarian, E. J. Benjamin, A. S. Go, D. K. Arnett, M. J. Blaha et al.,

B. M. Judd, J. H. Kissela, L. D. Lichtman, S. Lisabeth, R. H. Liu et al., American Heart Association Statistics Committee and Stroke Statistics Subcommittee, 2016.

, Update: A Report From the American Heart Association, vol.133, pp.38-360, 2016.

G. A. Roth, M. H. Forouzanfar, A. E. Moran, R. Barber, G. Nguyen et al., Demographic and Epidemiologic Drivers of Global Cardiovascular Mortality, N. Engl. J. Med, vol.372, pp.1333-1341, 2015.

A. Carrel and V. , On the Experimental Surgery of the Thoracic Aorta and Heart, Ann. Surg, vol.52, pp.83-95, 1910.

F. Sellke and P. J. Del-nido, Sabiston and Spencer Surgery of the Chest, Sabiston Spencer Surg. Chest

M. Nichols, N. Townsend, P. Scarborough, and M. Rayner, Cardiovascular disease in Europe 2014: epidemiological update, Eur. Heart J, vol.35, pp.2950-2959, 2014.

S. Yusuf, D. Wood, J. Ralston, and K. S. Reddy, The World Heart Federation's vision for worldwide cardiovascular disease prevention, The Lancet, vol.386, pp.60265-60268, 2015.

H. C. Stary, Natural History and Histological Classification of Atherosclerotic Lesions An Update, Arterioscler. Thromb. Vasc. Biol, vol.20, pp.1177-1178, 2000.

S. Glagov, E. Weisenberg, C. K. Zarins, R. Stankunavicius, and G. J. Kolettis, Compensatory Enlargement of Human Atherosclerotic Coronary Arteries, N. Engl. J. Med, vol.316, pp.1371-1375, 1987.

A. R. Grüntzig, Å. Senning, and W. E. Siegenthaler, Nonoperative Dilatation of Coronary-Artery Stenosis, N. Engl. J. Med, vol.301, pp.61-68, 1979.

G. N. Levine, E. R. Bates, J. C. Blankenship, S. R. Bailey, J. A. Bittl et al., ACCF/AHA/SCAI Guideline for Percutaneous Coronary Intervention: a report of the American College of Cardiology Foundation/American Heart Association Task Force on Practice Guidelines and the Society for Cardiovascular Angiography and Interventions, Circulation, vol.124, pp.574-651, 2011.

B. R. Shah, L. A. Mccoy, J. J. Federspiel, D. Mudrick, P. A. Cowper et al., Use of Stress Testing and Diagnostic Catheterization After Coronary StentingAssociation of Site-Level Patterns With Patient Characteristics and Outcomes, vol.247, p.52

, Medicare Beneficiaries, vol.62, pp.439-446, 2013.

E. Kedhi, K. S. Joesoef, E. Mcfadden, J. Wassing, C. Van-mieghem et al., Second-generation everolimus-eluting and paclitaxel-eluting stents in real-life practice (COMPARE): a randomised trial, Lancet Lond. Engl, vol.375, pp.62127-62136, 2010.

T. Palmerini, G. Biondi-zoccai, D. D. Riva, A. Mariani, P. Genereux et al.,

. Stone, Stent thrombosis with drug-eluting stents: is the paradigm shifting?, J. Am. Coll. Cardiol, vol.62, pp.1915-1921, 2013.

L. Qi-hua, Z. Qi, Z. Yu, L. Xiao-long, J. Hai-gang et al., Long-term effect of second-generation drug-eluting stents for coronary artery disease, everolimus-eluting versus zotarolimus-eluting stents: a meta-analysis, Artery Dis, vol.26, pp.259-265, 2015.

P. W. Serruys, M. Morice, A. P. Kappetein, A. Colombo, D. R. Holmes et al.,

T. E. Ståhle, M. Feldman, E. J. Van-den-brand, N. Bass, K. Van-dyck et al.,

S. Mohr and . Investigators, Percutaneous coronary intervention versus coronary-artery bypass grafting for severe coronary artery disease, N. Engl. J. Med, vol.360, pp.961-972, 2009.

M. E. Farkouh, M. Domanski, L. A. Sleeper, F. S. Siami, G. Dangas et al.,

Y. Cohen, S. D. Rosenberg, A. S. Solomon, B. J. Desai, E. A. Gersh et al., FREEDOM Trial Investigators, Strategies for multivessel revascularization in patients with diabetes, vol.367, pp.2375-2384, 2012.

A. Kapur, R. J. Hall, I. S. Malik, A. C. Qureshi, J. Butts et al., Randomized comparison of percutaneous coronary intervention with coronary artery bypass grafting in diabetic patients. 1-year results of the CARDia (Coronary Artery Revascularization in Diabetes) trial, J. Am. Coll. Cardiol, vol.55, pp.432-440, 2010.

A. P. Kappetein, T. E. Feldman, M. J. Mack, M. Morice, D. R. Holmes et al.,

F. W. Dawkins, P. W. Mohr, A. Serruys, and . Colombo, Comparison of coronary bypass surgery with drugeluting stenting for the treatment of left main and/or three-vessel disease: 3-year follow-up of the SYNTAX trial, Eur. Heart J, vol.32, pp.2125-2134, 2011.

F. D. Loop, B. W. Lytle, D. M. Cosgrove, R. W. Stewart, M. Goormastic et al., Influence of the internal-mammary-artery graft on 10-year survival and other cardiac events, N. Engl. J. Med, vol.314, pp.1-6, 1986.

N. D. Desai, E. A. Cohen, C. D. Naylor, and S. E. Fremes, Radial Artery Patency Study Investigators, A randomized comparison of radial-artery and saphenous-vein coronary bypass grafts, N. Engl. J. Med, vol.351, pp.2302-2309, 2004.

S. Goldman, G. K. Sethi, W. Holman, H. Thai, E. Mcfalls et al.,

M. K. Pierce, S. Kreamer, M. Shih, and K. Lee, Radial artery grafts vs saphenous vein grafts in coronary artery bypass surgery: a randomized trial, JAMA, vol.305, pp.167-174, 2011.

H. Hirose, A. Amano, S. Takanashi, and A. Takahashi, Coronary artery bypass grafting using the gastroepiploic artery in 1,000 patients, Ann. Thorac. Surg, vol.73, pp.1371-1379, 2002.

Y. Gao, F. Liu, L. Zhang, X. Su, J. Y. Liu et al., Acellular blood vessels combined human hair follicle mesenchymal stem cells for engineering of functional arterial grafts, Ann. Biomed. Eng, vol.42, pp.2177-2189, 2014.

N. Heureux, N. Dusserre, G. Konig, B. Victor, P. Keire et al.,

C. R. Kyles, G. Gregory, R. C. Hoyt, T. N. Robbins, and . Mcallister, Human tissue-engineered blood vessels for adult arterial revascularization, Nat. Med, vol.12, pp.361-365, 2006.

C. Michiels, Endothelial cell functions, J. Cell. Physiol, vol.196, pp.430-443, 2003.

J. S. Pober and W. C. Sessa, Evolving functions of endothelial cells in inflammation, Nat. Rev. Immunol, vol.7, pp.803-815, 2007.

M. T. Koobatian, S. Row, R. J. Smith, C. Koenigsknecht, S. T. Andreadis et al., Successful endothelialization and remodeling of a cell-free small-diameter arterial graft in a large animal model, Biomaterials, vol.76, pp.344-358, 2016.

D. G. Seifu, A. Purnama, K. Mequanint, and D. Mantovani, Small-diameter vascular tissue engineering, Nat. Rev. Cardiol, vol.10, pp.410-421, 2013.

H. Rammal, C. Harmouch, J. Lataillade, D. Laurent-maquin, P. Labrude et al., Stem cells: a promising source for vascular regenerative medicine, Stem Cells Dev, vol.23, pp.2931-2949, 2014.
URL : https://hal.archives-ouvertes.fr/hal-01453191

R. E. Omar, J. Beroud, J. Stoltz, P. Menu, E. Velot et al., Umbilical Cord Mesenchymal Stem Cells: The New Gold Standard for Mesenchymal Stem Cell-Based Therapies?, Tissue Eng. Part B Rev, 2014.
URL : https://hal.archives-ouvertes.fr/hal-01777817

L. J. Fischer, S. Mcilhenny, T. Tulenko, N. Golesorkhi, P. Zhang et al.,

P. J. Shapiro and . Dimuzio, Endothelial differentiation of adipose-derived stem cells: effects of endothelial cell growth supplement and shear force, J. Surg. Res, vol.152, pp.157-166, 2009.

M. Chen, P. Lie, Z. Li, and X. Wei, Endothelial differentiation of Wharton's jellyderived mesenchymal stem cells in comparison with bone marrow-derived mesenchymal stem cells, Exp. Hematol, vol.37, pp.629-640, 2009.

T. Ahsan and R. M. Nerem, Fluid shear stress promotes an endothelial-like phenotype during the early differentiation of embryonic stem cells, Tissue Eng. Part A, vol.16, pp.3547-3553, 2010.

G. Chen, Y. Lv, P. Guo, C. Lin, X. Zhang et al., Matrix mechanics and fluid shear stress control stem cells fate in three dimensional microenvironment, Curr. Stem Cell Res. Ther, vol.8, pp.313-323, 2013.

W. L. Murphy, T. C. Mcdevitt, and A. J. Engler, Materials as stem cell regulators, Nat. Mater, vol.13, pp.547-557, 2014.

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

J. Ringe and M. Sittinger, Regenerative medicine: Selecting the right biological scaffold for tissue engineering, Nat. Rev. Rheumatol, vol.10, pp.388-389, 2014.

S. J. Lee, J. Liu, S. H. Oh, S. Soker, A. Atala et al., Development of a composite vascular scaffolding system that withstands physiological vascular conditions, Biomaterials, vol.29, pp.2891-2898, 2008.

F. Ahmed, N. K. Dutta, A. Zannettino, K. Vandyke, and N. R. Choudhury, Engineering interaction between bone marrow derived endothelial cells and electrospun surfaces for artificial vascular graft applications, Biomacromolecules, vol.15, pp.1276-1287, 2014.

D. D. Swartz, J. A. Russell, and S. T. Andreadis, Engineering of fibrin-based functional and implantable small-diameter blood vessels, Am. J. Physiol. Heart Circ. Physiol, vol.288, pp.1451-1460, 2005.

C. B. Weinberg and E. Bell, A blood vessel model constructed from collagen and cultured vascular cells, Science, vol.231, pp.397-400, 1986.

L. E. Niklason, J. Gao, W. M. Abbott, K. K. Hirschi, S. Houser et al., Functional arteries grown in vitro, Science, vol.284, pp.489-493, 1999.

S. S. Silva, J. F. Mano, and R. L. Reis, Potential applications of natural origin polymer-based systems in soft tissue regeneration, Crit. Rev. Biotechnol, vol.30, pp.200-221, 2010.

Y. Zhao, S. Zhang, J. Zhou, J. Wang, M. Zhen et al., The development of a tissue-engineered artery using decellularized scaffold and autologous ovine mesenchymal stem cells, Biomaterials, vol.31, pp.296-307, 2010.

M. P. Lutolf, P. M. Gilbert, and H. M. Blau, Designing materials to direct stem-cell fate, Nature, vol.462, pp.433-441, 2009.

R. A. Pérez, J. Won, J. C. Knowles, and H. Kim, Naturally and synthetic smart composite biomaterials for tissue regeneration, Adv. Drug Deliv. Rev, vol.65, pp.471-496, 2013.

S. Gomes, I. B. Leonor, J. F. Mano, R. L. Reis, and D. L. Kaplan, Natural and Genetically Engineered Proteins for Tissue Engineering, Prog. Polym. Sci, vol.37, pp.1-17, 2012.

J. F. Mano, G. A. Silva, H. S. Azevedo, P. B. Malafaya, R. A. Sousa et al., Natural origin biodegradable systems in tissue engineering and regenerative medicine: present status and some moving trends, J. R. Soc. Interface R. Soc, vol.4, pp.999-1030, 2007.

P. Chandra and S. J. Lee, Synthetic Extracellular Microenvironment for Modulating Stem Cell Behaviors, Biomark. Insights, vol.10, pp.105-116, 2015.

D. E. Discher, P. Janmey, and Y. Wang, Tissue Cells Feel and Respond to the Stiffness of Their Substrate, Science, vol.310, pp.1139-1143, 2005.

W. Liu, K. Merrett, M. Griffith, P. Fagerholm, S. Dravida et al., Biomaterials, vol.29, pp.1147-1158, 2008.

F. Guilak, D. M. Cohen, B. T. Estes, J. M. Gimble, W. Liedtke et al., Control of Stem Cell Fate by Physical Interactions with the Extracellular Matrix, Cell Stem Cell, vol.5, pp.17-26, 2009.

N. Berthelemy, H. Kerdjoudj, C. Gaucher, P. Schaaf, J. Stoltz et al., Polyelectrolyte Films Boost Progenitor Cell Differentiation into Endothelium-like Monolayers, Adv. Mater. Deerfield Beach Fla, vol.20, pp.2674-2678, 2008.
URL : https://hal.archives-ouvertes.fr/hal-02164025

T. Boudou, T. Crouzier, K. Ren, G. Blin, and C. Picart, Multiple functionalities of polyelectrolyte multilayer films: new biomedical applications, Adv. Mater. Deerfield Beach Fla, vol.22, pp.441-467, 2010.
URL : https://hal.archives-ouvertes.fr/hal-00670215

H. Kerdjoudj, N. Berthelemy, S. Rinckenbach, A. Kearney-schwartz, K. Montagne et al., Small Vessel Replacement by Human Umbilical Arteries With Polyelectrolyte Film-Treated ArteriesIn Vivo Behavior, J. Am. Coll. Cardiol, vol.52, pp.1589-1597, 2008.

A. J. Melchiorri, L. G. Bracaglia, L. Kimerer, N. Hibino, and J. P. Fisher, In Vitro Endothelialization of Biodegradable Vascular Grafts via Endothelial Progenitor Cell Seeding and Maturation in a Tubular Perfusion System Bioreactor, Tissue Eng. Part C Methods, 2016.

G. A. Villalona, B. Udelsman, D. R. Duncan, E. Mcgillicuddy, R. F. Sawh-martinez et al.,

C. Hibino, T. Painter, B. Mirensky, T. Erickson, C. K. Shinoka et al., Cell-Seeding Techniques in Vascular Tissue Engineering, vol.16, pp.341-350, 2010.

C. C. Anamelechi, E. C. Clermont, M. T. Novak, and W. M. Reichert, Dynamic seeding of perfusing human umbilical vein endothelial cells (HUVECs) onto dual-function cell adhesion ligands: Arg-Gly-Asp (RGD)-streptavidin and biotinylated fibronectin, Langmuir ACS J. Surf. Colloids, vol.25, pp.5725-5730, 2009.

L. A. Solchaga, E. Tognana, K. Penick, H. Baskaran, V. M. Goldberg et al., A Rapid Seeding Technique for the Assembly of Large Cell/Scaffold Composite Constructs, Tissue Eng, vol.12, pp.1851-1863, 2006.

T. Kitagawa, T. Yamaoka, R. Iwase, and A. Murakami, Three-dimensional cell seeding and growth in radial-flow perfusion bioreactor for in vitro tissue reconstruction, Biotechnol. Bioeng, vol.93, pp.947-954, 2006.

W. T. Godbey, B. S. Stacey-hindy, M. E. Sherman, and A. Atala, A novel use of centrifugal force for cell seeding into porous scaffolds, Biomaterials, vol.25, pp.2799-2805, 2004.

C. Quint, Y. Kondo, R. J. Manson, J. H. Lawson, A. Dardik et al., Decellularized tissue-engineered blood vessel as an arterial conduit, Proc. Natl. Acad. Sci, vol.108, pp.9214-9219, 2011.

D. Wion, T. Christen, E. L. Barbier, and J. A. Coles, PO2 Matters in Stem Cell Culture, Cell Stem Cell, vol.5, pp.242-243, 2009.
URL : https://hal.archives-ouvertes.fr/inserm-00527773

S. Hsu, I. Tsai, D. Lin, and D. C. Chen, The effect of dynamic culture conditions on endothelial cell seeding and retention on small diameter polyurethane vascular grafts, Med. Eng. Phys, vol.27, pp.267-272, 2005.

G. Shen, H. C. Tsung, C. F. Wu, X. Y. Liu, X. Wang et al., Tissue engineering of blood vessels with endothelial cells differentiated from mouse embryonic stem cells, Cell Res, vol.13, pp.335-341, 2003.

H. Kurobe, M. W. Maxfield, C. K. Breuer, and T. Shinoka, Concise Review: Tissue-Engineered Vascular Grafts for Cardiac Surgery: Past, Present, and Future, Stem Cells Transl. Med, vol.1, pp.566-571, 2012.

J. A. Ankrum, J. F. Ong, and J. M. Karp, Mesenchymal stem cells: immune evasive, not immune privileged, Nat. Biotechnol, 2014.

R. J. Galas and J. C. Liu, Vascular endothelial growth factor does not accelerate endothelial differentiation of human mesenchymal stem cells, J. Cell. Physiol, vol.229, pp.90-96, 2014.

M. B. Chan-park, J. Y. Shen, Y. Cao, Y. Xiong, Y. Liu et al.,

. Greisler, Biomimetic control of vascular smooth muscle cell morphology and phenotype for functional tissue-engineered small-diameter blood vessels, J. Biomed. Mater. Res. A, vol.88, pp.1104-1121, 2009.

N. Koike, D. Fukumura, O. Gralla, P. Au, J. S. Schechner et al., Tissue engineering: creation of long-lasting blood vessels, Nature, vol.428, pp.138-139, 2004.

P. Dan, É. Velot, V. Decot, and P. Menu, The role of mechanical stimuli in the vascular differentiation of mesenchymal stem cells, J. Cell Sci, vol.128, pp.2415-2422, 2015.
URL : https://hal.archives-ouvertes.fr/hal-01465904

C. Wang, L. Cen, S. Yin, Q. Liu, W. Liu et al., A small diameter elastic blood vessel wall prepared under pulsatile conditions from polyglycolic acid mesh and smooth muscle cells differentiated from adipose-derived stem cells, Biomaterials, vol.31, pp.621-630, 2010.

E. S. Place, N. D. Evans, and M. M. Stevens, Complexity in biomaterials for tissue engineering, vol.8, pp.457-470, 2009.

T. Okoshi, H. Chen, G. Soldani, P. M. Galletti, and M. Goddard, Microporous small diameter PVDF-TrFE vascular grafts fabricated by a spray phase inversion technique, ASAIO J. Am. Soc. Artif. Intern. Organs, vol.38, pp.201-206, 1992.

A. Huttenlocher and A. R. Horwitz, Wound healing with electric potential, N. Engl. J. Med, vol.356, pp.303-304, 2007.

D. N. Coakley, F. M. Shaikh, K. O'sullivan, E. G. Kavanagh, P. A. Grace et al.,

. Mcgloughlin, Comparing the endothelialisation of extracellular matrix bioscaffolds with coated synthetic vascular graft materials, Int. J. Surg. Lond. Engl, vol.25, pp.31-37, 2016.

H. Kobayashi, D. Terada, Y. Yokoyama, D. W. Moon, Y. Yasuda et al., Vascular-inducing poly(glycolic acid)-collagen nanocomposite-fiber scaffold, J. Biomed. Nanotechnol, vol.9, pp.1318-1326, 2013.

W. S. Choi, Y. K. Joung, Y. Lee, J. W. Bae, H. K. Park et al., Enhanced Patency and Endothelialization of Small-Caliber Vascular Grafts Fabricated by Coimmobilization of Heparin and Cell-Adhesive Peptides, ACS Appl. Mater. Interfaces, vol.8, pp.4336-4346, 2016.

X. Yao, R. Peng, and J. Ding, Cell-material interactions revealed via material techniques of surface patterning, Adv. Mater. Deerfield Beach Fla, vol.25, pp.5257-5286, 2013.

W. Gong, D. Lei, S. Li, P. Huang, Q. Qi et al., Hybrid small-diameter vascular grafts: Anti-expansion effect of electrospun poly ?-caprolactone on heparin-coated decellularized matrices, Biomaterials, vol.76, pp.359-370, 2016.

J. M. Aamodt and D. W. Grainger, Extracellular matrix-based biomaterial scaffolds and the host response, Biomaterials, vol.86, pp.68-82, 2016.

L. Amadori, N. Rajan, S. Vesentini, and D. Mantovani, Atomic Force and Confocal Microscopic Studies of Collagen-Cell-Based Scaffolds for Vascular Tissue Engineering, Adv. Mater. Res, pp.83-88, 2007.

P. M. Crapo, T. W. Gilbert, and S. F. Badylak, An overview of tissue and whole organ decellularization processes, Biomaterials, vol.32, pp.3233-3243, 2011.

L. Gui, A. Muto, S. A. Chan, C. K. Breuer, and L. E. Niklason, Development of Decellularized Human Umbilical Arteries as Small-Diameter Vascular Grafts, Tissue Eng. Part A, vol.15, pp.2665-2676, 2009.

J. Chlupá?, E. Filová, T. Riedel, M. Houska, E. Brynda et al.,

R. Fernandez, C. Daculsi, L. Bourget, L. Bordenave, and . Ba?áková, Attachment of human endothelial cells to polyester vascular grafts: pre-coating with adhesive protein assemblies and resistance to shortterm shear stress, Physiol. Res. Acad. Sci. Bohemoslov, vol.63, pp.167-177, 2014.

M. M. Stevens and J. H. George, Exploring and engineering the cell surface interface, Science, vol.310, pp.1135-1138, 2005.

N. Huebsch and D. J. Mooney, Inspiration and application in the evolution of biomaterials, Nature, vol.462, pp.426-432, 2009.

A. Can and S. Karahuseyinoglu, Concise Review: Human Umbilical Cord Stroma with Regard to the Source of Fetus-Derived Stem Cells, STEM CELLS, vol.25, pp.2886-2895, 2007.

M. Köckritz-blickwede, O. A. Chow, and V. Nizet, Fetal calf serum contains heat-stable nucleases that degrade neutrophil extracellular traps, Blood, vol.114, pp.5245-5246, 2009.

G. Fercana, D. Bowser, M. Portilla, E. M. Langan, C. G. Carsten et al.,

. Simionescu, Platform Technologies for Decellularization, Tunic-Specific Cell Seeding, and In Vitro Conditioning of Extended Length, Small Diameter Vascular Grafts, Tissue Eng. Part C Methods, vol.20, pp.1016-1027, 2014.

A. Mahara, S. Somekawa, N. Kobayashi, Y. Hirano, Y. Kimura et al., Tissue-engineered acellular small diameter long-bypass grafts with neointima-inducing activity, Biomaterials, vol.58, pp.54-62, 2015.

R. Y. Kannan, H. J. Salacinski, P. E. Butler, G. Hamilton, and A. M. Seifalian, Current status of prosthetic bypass grafts: A review, J. Biomed. Mater. Res. B Appl. Biomater, vol.74, pp.570-581, 2005.