A. Islam, Haemopoietic stem cell: A new concept, Leukemia Research, vol.9, issue.11, pp.1415-1432, 1985.
DOI : 10.1016/0145-2126(85)90130-4

M. Tavassoli and A. Friedenstein, Hemopoietic stromal microenvironment, American Journal of Hematology, vol.302, issue.2, pp.195-203, 1983.
DOI : 10.1002/ajh.2830150211

E. D. Thomas, H. L. Jr, J. H. Cannon, O. D. Sahler, and J. W. Ferrebee, SUPRALETHAL WHOLE BODY IRRADIATION AND ISOLOGOUS MARROW TRANSPLANTATION IN MAN*???, Journal of Clinical Investigation, vol.38, issue.10 Pt 1-2, pp.1709-1716, 1959.
DOI : 10.1172/JCI103949

URL : http://www.ncbi.nlm.nih.gov/pmc/articles/PMC444138

R. A. Gatti, H. J. Meuwissen, H. D. Allen, R. Hong, and R. A. Good, IMMUNOLOGICAL RECONSTITUTION OF SEX-LINKED LYMPHOPENIC IMMUNOLOGICAL DEFICIENCY, The Lancet, vol.292, issue.7583, pp.1366-1369, 1968.
DOI : 10.1016/S0140-6736(68)92673-1

E. D. Thomas, A history of haemopoietic cell transplantation, British Journal of Haematology, vol.300, issue.2, pp.330-339, 1999.
DOI : 10.1016/0145-2126(77)90065-0

M. J. Evans and M. H. Kaufman, Establishment in culture of pluripotential cells from mouse embryos, Nature, vol.131, issue.5819, pp.154-156, 1981.
DOI : 10.1038/292154a0

G. R. Martin, Isolation of a pluripotent cell line from early mouse embryos cultured in medium conditioned by teratocarcinoma stem cells., Proceedings of the National Academy of Sciences, vol.78, issue.12, pp.7634-7638, 1981.
DOI : 10.1073/pnas.78.12.7634

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.663-676, 2006.
DOI : 10.1016/j.cell.2006.07.024

K. Takahashi, K. Tanabe, and M. Ohnuki, Induction of Pluripotent Stem Cells from Adult Human Fibroblasts by Defined Factors, Cell, vol.131, issue.5, pp.861-872, 2007.
DOI : 10.1016/j.cell.2007.11.019

E. Galende, I. Karakikes, and L. Edelmann, Amniotic Fluid Cells Are More Efficiently Reprogrammed to Pluripotency Than Adult Cells, Cellular Reprogramming (Formerly "Cloning and Stem Cells"), vol.12, issue.2, pp.117-125, 2010.
DOI : 10.1089/cell.2009.0077

URL : http://www.ncbi.nlm.nih.gov/pmc/articles/PMC2998987

C. Magnon, D. Lucas, and P. Frenette, Trafficking of Stem Cells, Stem Cell Migration: Methods and Protocols, pp.3-24, 2011.
DOI : 10.1007/978-1-61779-145-1_1

T. Zhao, Z. Zhang, Z. Rong, and Y. Xu, Immunogenicity of induced pluripotent stem cells, Nature, vol.6, issue.7350, pp.212-215, 2011.
DOI : 10.1038/nature10135

C. Campagnoli, I. A. Roberts, S. Kumar, P. R. Bennett, I. Bellantuono et al., Identification of mesenchymal stem/progenitor cells in human first-trimester fetal blood, liver, and bone marrow, Blood, vol.98, issue.8, pp.2396-2402, 2001.
DOI : 10.1182/blood.V98.8.2396

V. Dexheimer, S. Mueller, F. Braatz, and W. Richter, Reduced Reactivation from Dormancy but Maintained Lineage Choice of Human Mesenchymal Stem Cells with Donor Age, PLoS ONE, vol.21, issue.8, 2011.
DOI : 10.1371/journal.pone.0022980.t002

Y. Fukuchi, H. Nakajima, D. Sugiyama, I. Hirose, T. Kitamura et al., Human Placenta-Derived Cells Have Mesenchymal Stem/Progenitor Cell Potential, Stem Cells, vol.98, issue.5, pp.649-658, 2004.
DOI : 10.1634/stemcells.22-5-649

C. A. Roufosse, N. C. Direkze, W. R. Otto, and N. A. Wright, Circulating mesenchymal stem cells, The International Journal of Biochemistry & Cell Biology, vol.36, issue.4, pp.585-597, 2004.
DOI : 10.1016/j.biocel.2003.10.007

D. Suva, G. Garavaglia, and J. Menetrey, Non-hematopoietic human bone marrow contains long-lasting, pluripotential mesenchymal stem cells, Journal of Cellular Physiology, vol.13, issue.1, pp.110-118, 2004.
DOI : 10.1002/jcp.10396

W. Wagner, F. Wein, and A. Seckinger, Comparative characteristics of mesenchymal stem cells from human bone marrow, adipose tissue, and umbilical cord blood, Experimental Hematology, vol.33, issue.11, pp.1402-1416, 2005.
DOI : 10.1016/j.exphem.2005.07.003

H. Wang, S. Hung, and S. Peng, Mesenchymal Stem Cells in the Wharton's Jelly of the Human Umbilical Cord, Stem Cells, vol.74, issue.7, pp.1330-1337, 2004.
DOI : 10.1634/stemcells.2004-0013

B. L. Yen, H. Huang, and C. Chien, Isolation of Multipotent Cells from Human Term Placenta, Stem Cells, vol.265, issue.1, pp.3-9, 2005.
DOI : 10.1634/stemcells.2004-0098

G. Yu, X. Wu, and M. A. Dietrich, Yield and characterization of subcutaneous human adipose-derived stem cells by flow cytometric and adipogenic mRNA analyzes, Cytotherapy, vol.12, issue.4, pp.538-546, 2010.
DOI : 10.3109/14653241003649528

B. M. Abdallah and M. Kassem, Human mesenchymal stem cells: from basic biology to clinical applications, Gene Therapy, vol.137, issue.2, pp.109-116, 2008.
DOI : 10.1002/(SICI)1097-4636(20000305)49:3<328::AID-JBM5>3.0.CO;2-Q

S. Aggarwal and M. F. Pittenger, Human mesenchymal stem cells modulate allogeneic immune cell responses, Blood, vol.105, issue.4, pp.1815-1822, 2005.
DOI : 10.1182/blood-2004-04-1559

N. A. Kia, A. R. Bahrami, and M. Ebrahimi, Comparative analysis of chemokine receptor's expression in mesenchymal stem cells derived from human bone marrow and adipose tissue, Journal of Molecular Neuroscience, vol.44, issue.3, pp.178-185, 2011.

A. Can and S. Karahuseyinoglu, Concise Review: Human Umbilical Cord Stroma with Regard to the Source of Fetus-Derived Stem Cells, Stem Cells, vol.142, issue.11, pp.2886-2895, 2007.
DOI : 10.1634/stemcells.2007-0417

M. Dominici, K. L. Blanc, and I. Mueller, Minimal criteria for defining multipotent mesenchymal stromal cells. The International Society for Cellular Therapy position statement, Cytotherapy, vol.8, issue.4, pp.315-317, 2006.
DOI : 10.1080/14653240600855905

J. A. Efe, S. Hilcove, and J. Kim, Conversion of mouse fibroblasts into cardiomyocytes using a direct reprogramming strategy, Nature Cell Biology, vol.280, issue.3, pp.215-222, 2011.
DOI : 10.1038/ncb2164

A. Mojallal, C. Lequeux, and C. Shipkov, Influence of Age and Body Mass Index on the Yield and Proliferation Capacity of Adipose-Derived Stem Cells, Aesthetic Plastic Surgery, vol.64, issue.6, pp.1097-1105, 2011.
DOI : 10.1007/s00266-011-9743-7

L. V. Rodríguez, Z. Alfonso, R. Zhang, J. Leung, B. Wu et al., Clonogenic multipotent stem cells in human adipose tissue differentiate into functional smooth muscle cells, Proceedings of the National Academy of Sciences, vol.103, issue.32, pp.12167-12172, 2006.
DOI : 10.1073/pnas.0604850103

R. Sarugaser, D. Lickorish, D. Baksh, M. M. Hosseini, and J. E. Davies, Human Umbilical Cord Perivascular (HUCPV) Cells: A Source of Mesenchymal Progenitors, Stem Cells, vol.414, issue.31, pp.220-229, 2005.
DOI : 10.1634/stemcells.2004-0166

J. Yang, N. De-isla, and C. Huselstein, Evaluation of human MSCs cell cycle, viability and differentiation in micromass culture, Biorheology, vol.43, pp.3-4, 2006.

M. Malinowski, K. Pietraszek, and C. Perreau, Effect of Lumican on the Migration of Human Mesenchymal Stem Cells and Endothelial Progenitor Cells: Involvement of Matrix Metalloproteinase-14, PLoS ONE, vol.30, issue.19, 2012.
DOI : 10.1371/journal.pone.0050709.s009

URL : https://hal.archives-ouvertes.fr/hal-00772843

J. Oswald, S. Boxberger, and B. Jørgensen, Mesenchymal Stem Cells Can Be Differentiated Into Endothelial Cells In Vitro, Stem Cells, vol.9, issue.3, pp.377-384, 2004.
DOI : 10.1634/stemcells.22-3-377

M. F. Pittenger, A. M. Mackay, and S. C. Beck, Multilineage Potential of Adult Human Mesenchymal Stem Cells, Science, vol.284, issue.5411, pp.143-147, 1999.
DOI : 10.1126/science.284.5411.143

V. Planat-benard, J. Silvestre, and B. Cousin, Plasticity of Human Adipose Lineage Cells Toward Endothelial Cells: Physiological and Therapeutic Perspectives, Circulation, vol.109, issue.5, pp.656-663, 2004.
DOI : 10.1161/01.CIR.0000114522.38265.61

URL : https://hal.archives-ouvertes.fr/hal-00409575

N. Salmon, E. Paternotte, V. Decot, J. Stoltz, P. Menu et al., Polyelectrolyte multilayer films promote human cord blood stem cells differentiation into mature endothelial cells exhibiting a stable phenotype, Bio-Medical Materials and Engineering, vol.19, pp.4-5, 2009.

A. Allameh, H. Ahmadi-ashtiani, M. S. Emami-aleagha, and H. Rastegar, The metabolic function of hepatocytes differentiated from human mesenchymal stem cells is inversely related to cellular glutathione levels, Cell Biochemistry and Function, vol.719, issue.1, pp.194-200, 2014.
DOI : 10.1002/cbf.2994

L. Zhang, Y. Zhao, Z. Guan, J. Ye, N. De-isla et al., Application potential of mesenchymal stem cells derived from Wharton's jelly in liver tissue engineering, Bio-Medical Materials and Engineering, vol.25, issue.1, pp.137-143, 2015.

J. Ye, X. Su, J. Stoltz, N. De-isla, and L. Zhang, Signalling pathways involved in the process of mesenchymal stem cells differentiating into hepatocytes, Cell Proliferation, vol.330, issue.2, pp.157-165, 2015.
DOI : 10.1111/cpr.12165

URL : https://hal.archives-ouvertes.fr/hal-01451903

N. Feng, Q. Han, and J. Li, Activation, Stem Cells and Development, vol.23, issue.5, pp.515-529, 2014.
DOI : 10.1089/scd.2013.0263

URL : https://hal.archives-ouvertes.fr/hal-01399119

M. M. Martini, T. D. Jeremias, M. C. Kohler, L. L. Marostica, A. G. Trentin et al., Human Placenta-Derived Mesenchymal Stem Cells Acquire Neural Phenotype Under the Appropriate Niche Conditions, DNA and Cell Biology, vol.32, issue.2, pp.58-65, 2013.
DOI : 10.1089/dna.2012.1807

K. Ma, L. Fox, and G. Shi, Generation of neural stem cell-like cells from bone marrow-derived human mesenchymal stem cells, Neurological Research, vol.13, issue.10, pp.1083-1093, 2011.
DOI : 10.1089/scd.2007.0212

G. Chamberlain, J. Fox, B. Ashton, and J. Middleton, Concise Review: Mesenchymal Stem Cells: Their Phenotype, Differentiation Capacity, Immunological Features, and Potential for Homing, Stem Cells, vol.354, issue.151, pp.2739-2749, 2007.
DOI : 10.1634/stemcells.2007-0197

K. Chen, D. Wang, and W. T. Du, Human umbilical cord mesenchymal stem cells hUC-MSCs exert immunosuppressive activities through a PGE2-dependent mechanism, Clinical Immunology, vol.135, issue.3, pp.448-458, 2010.
DOI : 10.1016/j.clim.2010.01.015

A. Corcione, F. Benvenuto, and E. Ferretti, Human mesenchymal stem cells modulate B-cell functions, Blood, vol.107, issue.1, pp.367-372, 2006.
DOI : 10.1182/blood-2005-07-2657

L. Reppel, T. Margossian, and L. Yaghi, Hypoxic Culture Conditions for Mesenchymal Stromal/Stem Cells from Wharton???s Jelly: A Critical Parameter to Consider in a Therapeutic Context, Current Stem Cell Research & Therapy, vol.9, issue.4, pp.306-318, 2014.
DOI : 10.2174/1574888X09666140213204850

D. E. Wright, A. J. Wagers, A. P. Gulati, F. L. Johnson, and I. L. Weissman, Physiological Migration of Hematopoietic Stem and Progenitor Cells, Science, vol.294, issue.5548, pp.1933-1936, 2001.
DOI : 10.1126/science.1064081

H. H. Chen, V. Decot, J. P. Ouyang, J. F. Stoltz, D. Bensoussan et al., In vitro initial expansion of mesenchymal stem cells is influenced by the culture parameters used in the isolation process, Bio-Medical Materials and Engineering, vol.19, pp.4-5, 2009.

E. U. Alt, C. Senst, and S. N. Murthy, Aging alters tissue resident mesenchymal stem cell properties, Stem Cell Research, vol.8, issue.2, pp.215-225, 2012.
DOI : 10.1016/j.scr.2011.11.002

URL : http://doi.org/10.1016/j.scr.2011.11.002

M. S. Rao and M. P. Mattson, Stem cells and aging: expanding the possibilities, Mechanisms of Ageing and Development, vol.122, issue.7, pp.713-734, 2001.
DOI : 10.1016/S0047-6374(01)00224-X

B. M. Schipper, K. G. Marra, W. Zhang, A. D. Donnenberg, and J. P. Rubin, Regional Anatomic and Age Effects on Cell Function of Human Adipose-Derived Stem Cells, Annals of Plastic Surgery, vol.60, issue.5, pp.538-544, 2008.
DOI : 10.1097/SAP.0b013e3181723bbe

S. Zhou, J. S. Greenberger, and M. W. Epperly, Age-related intrinsic changes in human bone-marrow-derived mesenchymal stem cells and their differentiation to osteoblasts, Aging Cell, vol.78, issue.3, pp.335-343, 2008.
DOI : 10.1074/jbc.M412953200

M. Zhu, E. Kohan, J. Bradley, M. Hedrick, P. Benhaim et al., The effect of age on osteogenic, adipogenic and proliferative potential of female adipose-derived stem cells, Journal of Tissue Engineering and Regenerative Medicine, vol.7, issue.2, pt 1, pp.290-301, 2009.
DOI : 10.1002/term.165

Y. Li, N. Charif, D. Mainard, D. Bensoussan, J. Stoltz et al., Donor's age dependent proliferation decrease of human bone marrow mesenchymal stem cells is linked to diminished clonogenicity, Bio-Medical Materials and Engineering, vol.24, issue.1, pp.47-52, 2014.

G. Brooke, H. Tong, J. Levesque, and K. Atkinson, Molecular Trafficking Mechanisms of Multipotent Mesenchymal Stem Cells Derived from Human Bone Marrow and Placenta, Stem Cells and Development, vol.17, issue.5, pp.929-940, 2008.
DOI : 10.1089/scd.2007.0156

J. W. Goodman and G. S. Hodgson, Evidence for stem cells in the peripheral blood of mice, Blood, vol.19, pp.702-714, 1962.

Y. Jiang, B. Vaessen, T. Lenvik, M. Blackstad, M. Reyes et al., Multipotent progenitor cells can be isolated from postnatal murine bone marrow, muscle, and brain, Experimental Hematology, vol.30, issue.8, pp.896-904, 2002.
DOI : 10.1016/S0301-472X(02)00869-X

J. Tolar, M. J. Shaughnessy, and A. Panoskaltsis-mortari, Host factors that impact the biodistribution and persistence of multipotent adult progenitor cells, Blood, vol.107, issue.10, pp.4182-4188, 2006.
DOI : 10.1182/blood-2005-08-3289

M. Kastrinaki, I. Andreakou, P. Charbord, and H. A. Papadaki, Isolation of Human Bone Marrow Mesenchymal Stem Cells Using Different Membrane Markers: Comparison of Colony/Cloning Efficiency, Differentiation Potential, and Molecular Profile, Tissue Engineering Part C: Methods, vol.14, issue.4, pp.333-339, 2008.
DOI : 10.1089/ten.tec.2008.0173

M. F. Pera, Stem cells: The dark side of induced pluripotency, Nature, vol.25, issue.7336, pp.46-47, 2011.
DOI : 10.1038/471046a

G. Brooke, M. Cook, and C. Blair, Therapeutic applications of mesenchymal stromal cells, Seminars in Cell & Developmental Biology, vol.18, issue.6, pp.846-858, 2007.
DOI : 10.1016/j.semcdb.2007.09.012

A. Giordano, U. Galderisi, and I. R. Marino, From the laboratory bench to the patient's bedside: An update on clinical trials with mesenchymal stem cells, Journal of Cellular Physiology, vol.174, issue.1, pp.27-35, 2007.
DOI : 10.1002/jcp.20959

K. Tarte, J. Gaillard, and J. Lataillade, Clinical-grade production of human mesenchymal stromal cells: occurrence of aneuploidy without transformation, Blood, vol.115, issue.8, pp.1549-1553, 2010.
DOI : 10.1182/blood-2009-05-219907

URL : https://hal.archives-ouvertes.fr/hal-00744192

G. Brooke, T. Rossetti, and R. Pelekanos, Manufacturing of human placenta-derived mesenchymal stem cells for clinical trials, British Journal of Haematology, vol.208, issue.4, pp.571-579, 2009.
DOI : 10.1111/j.1365-2141.2008.07492.x

J. F. Stoltz, D. Dumas, and X. Wang, Influence of mechanical forces on cells and tissues A cell initiating human acute myeloid leukaemia after transplantation into SCID mice, Biorheology Nature, vol.37, issue.367 6464, pp.3-14, 1994.

M. Al-hajj, M. S. Wicha, A. Benito-hernandez, S. J. Morrison, and M. F. Clarke, Prospective identification of tumorigenic breast cancer cells, Proceedings of the National Academy of Sciences, vol.100, issue.7, pp.3983-3988, 2003.
DOI : 10.1073/pnas.0530291100

S. K. Singh, C. Hawkins, and I. D. Clarke, Identification of human brain tumour initiating cells, Nature, vol.64, issue.7015, pp.396-401, 2004.
DOI : 10.1038/nature03128

A. T. Collins, P. A. Berry, C. Hyde, M. J. Stower, and N. J. Maitland, Prospective Identification of Tumorigenic Prostate Cancer Stem Cells, Cancer Research, vol.65, issue.23, pp.10946-10951, 2005.
DOI : 10.1158/0008-5472.CAN-05-2018

C. Li, D. G. Heidt, and P. Dalerba, Identification of Pancreatic Cancer Stem Cells, Cancer Research, vol.67, issue.3, pp.1030-1037, 2007.
DOI : 10.1158/0008-5472.CAN-06-2030

P. Dalerba, R. W. Cho, and M. F. Clarke, Cancer Stem Cells: Models and Concepts, Annual Review of Medicine, vol.58, issue.1, pp.267-284, 2007.
DOI : 10.1146/annurev.med.58.062105.204854

W. Liao, Y. Ye, Y. Deng, X. Bian, and Y. Ding, Metastatic cancer stem cells: from the concept to therapeutics, American Journal of Stem Cells, vol.3, issue.2, pp.46-62, 2014.

A. D. Kim, D. L. Stachura, and D. Traver, Cell signaling pathways involved in hematopoietic stem cell specification, Experimental Cell Research, vol.329, issue.2, pp.227-233, 2014.
DOI : 10.1016/j.yexcr.2014.10.011

Y. Wu, Z. Wang, and Y. Cao, Cotransplantation of haploidentical hematopoietic and umbilical cord mesenchymal stem cells with a myeloablative regimen for refractory/relapsed hematologic malignancy, Annals of Hematology, vol.9, issue.12, pp.1675-1684, 2013.
DOI : 10.1007/s00277-013-1831-0

Y. Wu, Y. Cao, and X. Li, Cotransplantation of haploidentical hematopoietic and umbilical cord mesenchymal stem cells for severe aplastic anemia: Successful engraftment and mild GVHD, Stem Cell Research, vol.12, issue.1, pp.132-138, 2014.
DOI : 10.1016/j.scr.2013.10.001

J. J. Auletta, S. K. Eid, and P. Wuttisarnwattana, Human Mesenchymal Stromal Cells Attenuate Graft-Versus-Host Disease and Maintain Graft-Versus-Leukemia Activity Following Experimental Allogeneic Bone Marrow Transplantation, STEM CELLS, vol.120, issue.suppl 6, pp.601-614, 2015.
DOI : 10.1002/stem.1867

URL : http://www.ncbi.nlm.nih.gov/pmc/articles/PMC4304927

M. S. Cairo and J. E. Wagner, Placental and/or umbilical cord blood: an alternative source of hematopoietic stem cells for transplantation, Blood, vol.90, issue.12, pp.4665-4678, 1997.

J. Cany, H. Dolstra, and N. Shah, Umbilical cord blood???derived cellular products for cancer immunotherapy, Cytotherapy, vol.17, issue.6, pp.739-748, 2015.
DOI : 10.1016/j.jcyt.2015.03.005

V. Rocha, C. Chastang, and G. Souillet, Related cord blood transplants: the Eurocord experience from 78 transplants, Bone Marrow Transplantation, vol.21, pp.59-62, 1998.

E. Gluckman, Ten years of cord blood transplantation: from bench to bedside, British Journal of Haematology, vol.137, issue.2, pp.192-199, 2009.
DOI : 10.1111/j.1365-2141.2009.07780.x

M. E. Horwitz and F. Frassoni, Improving the outcome of umbilical cord blood transplantation through ex??vivo expansion or graft manipulation, Cytotherapy, vol.17, issue.6, pp.730-738, 2015.
DOI : 10.1016/j.jcyt.2015.02.004

K. K. Ballen, E. Gluckman, and H. E. Broxmeyer, Umbilical cord blood transplantation: the first 25 years and beyond, Blood, vol.122, issue.4, pp.491-498, 2013.
DOI : 10.1182/blood-2013-02-453175

URL : http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3952633

M. Gigante and E. Ranieri, Research Highlights, Immunotherapy, vol.2, issue.5, pp.607-608, 2010.
DOI : 10.2217/imt.10.54

G. Zhang, H. Zhao, and J. Wu, Adoptive immunotherapy for non-small cell lung cancer by NK and cytotoxic T lymphocytes mixed effector cells: Retrospective clinical observation, International Immunopharmacology, vol.21, issue.2, pp.396-405, 2014.
DOI : 10.1016/j.intimp.2014.04.026

V. Decot, L. Voillard, and V. Latger-cannard, Natural-killer cell amplification for adoptive leukemia relapse immunotherapy: Comparison of three cytokines, IL-2, IL-15, or IL-7 and impact on NKG2D, KIR2DL1, and KIR2DL2 expression, Experimental Hematology, vol.38, issue.5, pp.351-362, 2010.
DOI : 10.1016/j.exphem.2010.02.006

M. Cheng, Y. Chen, W. Xiao, R. Sun, and Z. Tian, NK cell-based immunotherapy for malignant diseases, Cellular and Molecular Immunology, vol.8, issue.3, pp.230-252, 2013.
DOI : 10.3109/14653249.2010.515582

URL : http://www.ncbi.nlm.nih.gov/pmc/articles/PMC4076738

C. Eguizabal, O. Zenarruzabeitia, and J. Monge, Natural Killer Cells for Cancer Immunotherapy: Pluripotent Stem Cells-Derived NK Cells as an Immunotherapeutic Perspective, Frontiers in Immunology, vol.2, issue.74, 2014.
DOI : 10.1371/journal.pone.0000232

Y. Zhang, L. Wang, D. Li, and N. Li, Taming regulatory T cells by autologous T cell immunization: A potential new strategy for cancer immune therapy, International Immunopharmacology, vol.9, issue.5, pp.593-595, 2009.
DOI : 10.1016/j.intimp.2009.01.026

L. Xu, W. Xu, Z. Jiang, F. Zhang, Y. Chu et al., Depletion of CD4+CD25high regulatory T cells from tumor infiltrating lymphocytes predominantly induces Th1 type immune response in vivo which inhibits tumor growth in adoptive immunotherapy, Cancer Biology & Therapy, vol.8, issue.1, pp.66-72, 2009.
DOI : 10.4161/cbt.8.1.7131

F. Dazzi and F. M. Marelli-berg, Mesenchymal stem cells for graft-versus-host disease: Close encounters with T cells, European Journal of Immunology, vol.19, issue.6, pp.1479-1482, 2008.
DOI : 10.1002/eji.200838433

K. , L. Blanc, I. Rasmusson, and B. Sundberg, Treatment of severe acute graft-versus-host disease with third party haploidentical mesenchymal stem cells, The Lancet, vol.363, issue.9419, pp.1439-1441, 2004.

N. Kim, K. Im, and J. Lim, Mesenchymal stem cells for the treatment and prevention of graft-versus-host disease: experiments and practice, Annals of Hematology, vol.114, issue.3, pp.1295-1308, 2013.
DOI : 10.1007/s00277-013-1796-z

J. D. Glenn and K. A. Whartenby, Mesenchymal stem cells: Emerging mechanisms of immunomodulation and therapy, World Journal of Stem Cells, vol.6, issue.5, pp.526-539, 2014.
DOI : 10.4252/wjsc.v6.i5.526

M. Introna and A. Rambaldi, Mesenchymal stromal cells for prevention and treatment of graft-versus-host disease, Current Opinion in Organ Transplantation, vol.20, issue.1, pp.72-78, 2015.
DOI : 10.1097/MOT.0000000000000158

N. Amariglio, A. Hirshberg, and B. W. Scheithauer, Donorderived brain tumor following neural stem cell transplantation in an ataxia telangiectasia patient, PLoS Medicine, vol.6, issue.2, 2009.

J. Tritz-schiavi, N. Charif, and C. Henrionnet, Original approach for cartilage tissue engineering with mesenchymal stem cells, Bio-Medical Materials and Engineering, vol.20, issue.3, pp.167-174, 2010.

A. I. Caplan and S. P. Bruder, Mesenchymal stem cells: building blocks for molecular medicine in the 21st century, Trends in Molecular Medicine, vol.7, issue.6, pp.259-264, 2001.
DOI : 10.1016/S1471-4914(01)02016-0

A. Schäffler and C. Büchler, Concise Review: Adipose Tissue-Derived Stromal Cells-Basic and Clinical Implications for Novel Cell-Based Therapies, Stem Cells, vol.24, issue.4, pp.818-827, 2007.
DOI : 10.1634/stemcells.2006-0589

A. I. Caplan, Adult mesenchymal stem cells for tissue engineering versus regenerative medicine, Journal of Cellular Physiology, vol.7, issue.8, pp.341-347, 2007.
DOI : 10.1002/jcp.21200

H. Gai, E. L. Leung, and P. D. Costantino, Generation and characterization of functional cardiomyocytes using induced pluripotent stem cells derived from human fibroblasts, Cell Biology International, vol.33, issue.11, pp.1184-1193, 2009.
DOI : 10.1016/j.cellbi.2009.08.008

R. E. Bittner, C. Schöfer, and K. Weipoltshammer, Recruitment of bone-marrow-derived cells by skeletal and cardiac muscle in adult dystrophic mdx mice, Anatomy and Embryology, vol.199, issue.5, pp.391-396, 1999.
DOI : 10.1007/s004290050237

A. Deb, S. Wang, K. A. Skelding, D. Miller, D. Simper et al., Bone Marrow-Derived Cardiomyocytes Are Present in Adult Human Heart: A Study of Gender-Mismatched Bone Marrow Transplantation Patients, Circulation, vol.107, issue.9, pp.1247-1249, 2003.
DOI : 10.1161/01.CIR.0000061910.39145.F0

S. Itescu, A. A. Kocher, and M. D. Schuster, Myocardial neovascularization by adult bone marrow-derived angioblasts: strategies for improvement of cardiomyocyte function, Heart Failure Reviews, vol.8, issue.3, pp.253-258, 2003.
DOI : 10.1023/A:1024721717926

S. Kang, Y. J. Yang, C. J. Li, and R. L. Gao, Effects of intracoronary autologous bone marrow cells on left ventricular function in acute myocardial infarction: a systematic review and meta-analysis for randomized controlled trials, Coronary Artery Disease, vol.19, issue.5, pp.327-335, 2008.
DOI : 10.1097/MCA.0b013e328300dbd3

M. A. Laflamme, D. Myerson, J. E. Saffitz, and C. E. Murry, Evidence for Cardiomyocyte Repopulation by Extracardiac Progenitors in Transplanted Human Hearts, Circulation Research, vol.90, issue.6, pp.634-640, 2002.
DOI : 10.1161/01.RES.0000014822.62629.EB

R. Swijnenburg, M. Tanaka, and H. Vogel, Embryonic stem cell immunogenicity increases upon differentiation after transplantation into ischemic myocardium, Circulation, vol.112, issue.9, pp.166-172, 2005.

J. Tang, Q. Xie, G. Pan, J. Wang, and M. Wang, Mesenchymal stem cells participate in angiogenesis and improve heart function in rat model of myocardial ischemia with reperfusion, European Journal of Cardio-Thoracic Surgery, vol.30, issue.2, pp.353-361, 2006.
DOI : 10.1016/j.ejcts.2006.02.070

C. Toma, M. F. Pittenger, K. S. Cahill, B. J. Byrne, and P. D. Kessler, Human Mesenchymal Stem Cells Differentiate to a Cardiomyocyte Phenotype in the Adult Murine Heart, Circulation, vol.105, issue.1, pp.93-98, 2002.
DOI : 10.1161/hc0102.101442

A. P. Gee, S. Richman, and A. Durett, Multicenter cell processing for cardiovascular regenerative medicine applications: the Cardiovascular Cell Therapy Research Network (CCTRN) experience, Cytotherapy, vol.12, issue.5, pp.684-691, 2010.
DOI : 10.3109/14653249.2010.487900

B. Trachtenberg, D. L. Velazquez, and A. R. Williams, Rationale and design of the Transendocardial Injection of Autologous Human Cells (bone marrow or mesenchymal) in Chronic Ischemic Left Ventricular Dysfunction and Heart Failure Secondary to Myocardial Infarction (TAC-HFT) trial: A randomized, double-blind, placebo-controlled study of safety and efficacy, American Heart Journal, vol.161, issue.3, pp.487-493, 2011.
DOI : 10.1016/j.ahj.2010.11.024

J. M. Hare, J. H. Traverse, and T. D. Henry, A Randomized, Double-Blind, Placebo-Controlled, Dose-Escalation Study of Intravenous Adult Human Mesenchymal Stem Cells (Prochymal) After Acute Myocardial Infarction, Journal of the American College of Cardiology, vol.54, issue.24, pp.2277-2286, 2009.
DOI : 10.1016/j.jacc.2009.06.055

S. Chen, W. Fang, and F. Ye, Effect on left ventricular function of intracoronary transplantation of autologous bone marrow mesenchymal stem cell in patients with acute myocardial infarction, The American Journal of Cardiology, vol.94, issue.1, pp.92-95, 2004.
DOI : 10.1016/j.amjcard.2004.03.034

K. H. Wu, B. Zhou, and X. M. Mo, Therapeutic Potential of Human Umbilical Cord???Derived Stem Cells in Ischemic Diseases, Transplantation Proceedings, vol.39, issue.5, pp.1620-1622, 2007.
DOI : 10.1016/j.transproceed.2006.12.041

N. Berthelemy, H. Kerdjoudj, and P. Schaaf, O2 Level Controls Hematopoietic Circulating Progenitor Cells Differentiation into Endothelial or Smooth Muscle Cells, PLoS ONE, vol.12, issue.5, 2009.
DOI : 10.1371/journal.pone.0005514.g005

URL : https://hal.archives-ouvertes.fr/hal-00430681

M. Faustini, M. Bucco, and T. Chlapanidas, Nonexpanded Mesenchymal Stem Cells for Regenerative Medicine: Yield in Stromal Vascular Fraction from Adipose Tissues, Tissue Engineering Part C: Methods, vol.16, issue.6, pp.1515-1521, 2010.
DOI : 10.1089/ten.tec.2010.0214

S. Kachgal and A. J. Putnam, Mesenchymal stem cells from adipose and bone marrow promote angiogenesis via distinct cytokine and protease expression mechanisms, Angiogenesis, vol.100, issue.9, pp.47-59, 2011.
DOI : 10.1007/s10456-010-9194-9

URL : http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3369878

D. Kaigler, P. H. Krebsbach, P. J. Polverini, and D. J. Mooney, Role of Vascular Endothelial Growth Factor in Bone Marrow Stromal Cell Modulation of Endothelial Cells, Tissue Engineering, vol.9, issue.1, pp.95-103, 2003.
DOI : 10.1089/107632703762687573

C. Sengenès, A. Miranville, M. Maumus, S. De-barros, R. Busse et al., Progenitor Cells: Role of Stromal Derived Factor-1 Released by Adipose Tissue Capillary Endothelial Cells, STEM CELLS, vol.56, issue.9, pp.2269-2276, 2007.
DOI : 10.1634/stemcells.2007-0180

F. Zhang, S. Tsai, and K. Kato, Transforming Growth Factor-?? Promotes Recruitment of Bone Marrow Cells and Bone Marrow-derived Mesenchymal Stem Cells through Stimulation of MCP-1 Production in Vascular Smooth Muscle Cells, Journal of Biological Chemistry, vol.284, issue.26, pp.17564-17574, 2009.
DOI : 10.1074/jbc.M109.013987

U. Riegelsberger, A. Deten, and C. , Intravenous human umbilical cord blood transplantation for stroke: Impact on infarct volume and caspase-3-dependent cell death in spontaneously hypertensive rats, Experimental Neurology, vol.227, issue.1, pp.218-223, 2011.
DOI : 10.1016/j.expneurol.2010.11.008

J. S. Lee, J. M. Hong, G. J. Moon, P. H. Lee, Y. H. Ahn et al., A Long-Term Follow-Up Study of Intravenous Autologous Mesenchymal Stem Cell Transplantation in Patients With Ischemic Stroke, STEM CELLS, vol.25, issue.Suppl, pp.1099-1106, 2010.
DOI : 10.1002/stem.430

Y. Jiang, W. Zhu, J. Zhu, L. Wu, G. Xu et al., Feasibility of Delivering Mesenchymal Stem Cells Via Catheter to the Proximal End of the Lesion Artery in Patients With Stroke in the Territory of the Middle Cerebral Artery, Cell Transplantation, vol.22, issue.12, pp.2291-2298, 2013.
DOI : 10.3727/096368912X658818

D. C. Hess, C. A. Sila, A. J. Furlan, L. R. Wechsler, J. A. Switzer et al., A Double-Blind Placebo-Controlled Clinical Evaluation of Multistem for the Treatment of Ischemic Stroke, International Journal of Stroke, vol.42, issue.3, pp.381-386, 2014.
DOI : 10.1002/ana.20501

S. Lee, K. Chu, and K. Jung, Slowed progression in models of huntington disease by adipose stem cell transplantation, Annals of Neurology, vol.110, issue.5, pp.671-681, 2009.
DOI : 10.1002/ana.21788

T. Lopatina, N. Kalinina, and M. Karagyaur, Adipose-derived stem cells stimulate regeneration of peripheral nerves: BDNF secreted by these cells promotes nerve healing and axon growth de Novo Geron gets green light for human trial of ES cellderived product, PLoS ONE Nature Biotechnology, vol.6, issue.27 3, pp.213-214, 2009.

A. R. Chapman and C. C. Scala, Evaluating the First-in-Human Clinical Trial of a Human Embryonic Stem Cell-Based Therapy, Kennedy Institute of Ethics Journal, vol.22, issue.3, pp.243-261, 2012.
DOI : 10.1353/ken.2012.0013

P. Bigini, P. Veglianese, and G. Andriolo, Intracerebroventricular Administration of Human Umbilical Cord Blood Cells Delays Disease Progression in Two Murine Models of Motor Neuron Degeneration, Rejuvenation Research, vol.14, issue.6, pp.623-639, 2011.
DOI : 10.1089/rej.2011.1197

N. K. Venkataramana, S. K. Kumar, and S. Balaraju, Open-labeled study of unilateral autologous bone-marrow-derived mesenchymal stem cell transplantation in Parkinson's disease, Translational Research, vol.155, issue.2, pp.62-70, 2010.
DOI : 10.1016/j.trsl.2009.07.006

J. Y. Shin, H. J. Park, and H. N. Kim, Mesenchymal stem cells enhance autophagy and increase ??-amyloid clearance in Alzheimer disease models, Autophagy, vol.223, issue.1, pp.32-44, 2014.
DOI : 10.1002/ar.1092310411

C. M. Lewis and M. Suzuki, Therapeutic applications of mesenchymal stem cells for amyotrophic lateral sclerosis, Stem Cell Research & Therapy, vol.5, issue.2, 2014.
DOI : 10.1186/scrt421

N. De-isla, C. Huseltein, and N. , Introduction to tissue engineering and application for cartilage engineering, Bio- Medical Materials and Engineering, vol.20, pp.3-4, 2010.

F. Mao, W. Xu, and H. Qian, Immunosuppressive effects of mesenchymal stem cells in collagen-induced mouse arthritis, Inflammation Research, vol.27, issue.3, pp.219-225, 2010.
DOI : 10.1007/s00011-009-0090-y

J. F. Stoltz and E. , Mechanobiology: Cartilage and Chondrocyte, 2006.

A. Arthur, A. Zannettino, and S. Gronthos, The therapeutic applications of multipotential mesenchymal/stromal stem cells in skeletal tissue repair, Journal of Cellular Physiology, vol.12, issue.2, pp.237-245, 2009.
DOI : 10.1002/jcp.21592

S. P. Bruder, A. A. Kurth, M. Shea, W. C. Hayes, N. Jaiswal et al., Bone regeneration by implantation of purified, culture-expanded human mesenchymal stem cells, Journal of Orthopaedic Research, vol.22, issue.2, pp.155-162, 1998.
DOI : 10.1002/jor.1100160202

S. Wakitani, T. Mitsuoka, N. Nakamura, Y. Toritsuka, Y. Nakamura et al., Autologous Bone Marrow Stromal Cell Transplantation for Repair of Full-Thickness Articular Cartilage Defects in Human Patellae: Two Case Reports, Cell Transplantation, vol.13, issue.5, pp.595-600, 2004.
DOI : 10.3727/000000004783983747

C. Chen, D. T. Tambe, L. Deng, and L. Yang, Biomechanical properties and mechanobiology of the articular chondrocyte, AJP: Cell Physiology, vol.305, issue.12, pp.1202-1208, 2013.
DOI : 10.1152/ajpcell.00242.2013

R. E. Wilusz, J. Sanchez-adams, and F. Guilak, The structure and function of the pericellular matrix of articular cartilage, Matrix Biology, vol.39, pp.25-32, 2014.
DOI : 10.1016/j.matbio.2014.08.009

H. A. Breinan, T. Minas, H. Hsu, S. Nehrer, C. B. Sledge et al., Effect of Cultured Autologous Chondrocytes on Repair of Chondral Defects in a Canine Model*, The Journal of Bone & Joint Surgery, vol.79, issue.10, pp.1439-1451, 1997.
DOI : 10.2106/00004623-199710000-00001

S. Wakitani, K. Imoto, T. Yamamoto, M. Saito, N. Murata et al., Human autologous culture expanded bone marrow mesenchymal cell transplantation for repair of cartilage defects in osteoarthritic knees, Osteoarthritis and Cartilage, vol.10, issue.3, pp.199-206, 2002.
DOI : 10.1053/joca.2001.0504

M. Brittberg, A. Lindahl, A. Nilsson, C. Ohlsson, O. Isaksson et al., Treatment of Deep Cartilage Defects in the Knee with Autologous Chondrocyte Transplantation, New England Journal of Medicine, vol.331, issue.14, pp.889-895, 1994.
DOI : 10.1056/NEJM199410063311401

C. Huselstein, P. Netter, and N. De-isla, Mechanobiology, chondrocyte and cartilage, Bio-Medical Materials and Engineering, vol.18, pp.4-5, 2008.

P. Angele, J. U. Yoo, and C. Smith, Cyclic hydrostatic pressure enhances the chondrogenic phenotype of human mesenchymal progenitor cells differentiated in vitro, Journal of Orthopaedic Research, vol.80, issue.3, pp.451-457, 2003.
DOI : 10.1016/S0736-0266(02)00230-9

F. Barry, R. E. Boynton, B. Liu, and J. M. Murphy, Chondrogenic Differentiation of Mesenchymal Stem Cells from Bone Marrow: Differentiation-Dependent Gene Expression of Matrix Components, Experimental Cell Research, vol.268, issue.2, pp.189-200, 2001.
DOI : 10.1006/excr.2001.5278

J. J. Campbell, D. A. Lee, and D. L. Bader, Dynamic compressive strain influences chondrogenic gene expression in human mesenchymal stem cells, Biorheology, vol.43, pp.3-4, 2006.

L. De-girolamo, S. Lopa, E. Arrigoni, M. F. Sartori, F. W. Preis et al., Human adipose-derived stem cells isolated from young and elderly women: their differentiation potential and scaffold interaction during in vitro osteoblastic differentiation, Cytotherapy, vol.11, issue.6, pp.793-803, 2009.
DOI : 10.3109/14653240903079393

K. Hiraoka, S. Grogan, T. Olee, and M. Lotz, Mesenchymal progenitor cells in adult human articular cartilage, Biorheology, vol.43, pp.3-4, 2006.

J. I. Huang, N. Kazmi, M. M. Durbhakula, T. M. Hering, J. U. Yoo et al., Chondrogenic potential of progenitor cells derived from human bone marrow and adipose tissue: A patient-matched comparison, Journal of Orthopaedic Research, vol.23, issue.6, pp.1383-1389, 2005.
DOI : 10.1016/j.orthres.2005.03.018

J. Stoltz, C. Huselstein, and J. Schiavi, Human Stem Cells and Articular Cartilage Tissue Engineering, Current Pharmaceutical Biotechnology, vol.13, issue.15, pp.2682-2691, 2012.
DOI : 10.2174/138920112804724846

C. Fong, A. Subramanian, and K. Gauthaman, Human Umbilical Cord Wharton???s Jelly Stem Cells Undergo Enhanced Chondrogenic Differentiation when Grown on Nanofibrous Scaffolds and in a Sequential Two-stage Culture Medium Environment, Stem Cell Reviews and Reports, vol.203, issue.2, pp.195-209, 2012.
DOI : 10.1007/s12015-011-9289-8

X. Chen, F. Zhang, and X. He, Chondrogenic differentiation of umbilical cord-derived mesenchymal stem cells in type I collagen-hydrogel for cartilage engineering, Injury, vol.44, issue.4, pp.540-549, 2013.
DOI : 10.1016/j.injury.2012.09.024

S. Liu, K. D. Hou, and M. Yuan, Characteristics of mesenchymal stem cells derived from Wharton's jelly of human umbilical cord and for fabrication of non-scaffold tissue-engineered cartilage, Journal of Bioscience and Bioengineering, vol.117, issue.2, pp.229-235, 2014.
DOI : 10.1016/j.jbiosc.2013.07.001

S. Ciavarella, F. Dammacco, M. De-matteo, G. Loverro, F. Silvestris et al., Umbilical Cord Mesenchymal Stem Cells: Role of Regulatory Genes in Their Differentiation to Osteoblasts, Stem Cells and Development, vol.18, issue.8, pp.1211-1220, 2009.
DOI : 10.1089/scd.2008.0340

C. H. Jo, Y. G. Lee, and W. H. Shin, Intra-Articular Injection of Mesenchymal Stem Cells for the Treatment of Osteoarthritis of the Knee: A Proof-of-Concept Clinical Trial, STEM CELLS, vol.1, issue.5, pp.1254-1266, 2014.
DOI : 10.1002/stem.1634

A. Vega, M. A. Martín-ferrero, and F. D. Canto, Treatment of Knee Osteoarthritis With Allogeneic Bone Marrow Mesenchymal Stem Cells, Transplantation, vol.99, issue.8, 2015.
DOI : 10.1097/TP.0000000000000678

M. Dubsk´ydubsk´y, A. Jirkovská, and R. Bem, Comparison of the effect of stem cell therapy and percutaneous transluminal angioplasty on diabetic foot disease in patients with critical limb ischemia, Cytotherapy, vol.16, issue.12, pp.1733-1738, 2014.
DOI : 10.1016/j.jcyt.2014.08.010

R. Subrammaniyan, J. Amalorpavanathan, and R. Shankar, Our experience of application of Autologous Bone Marrow Stem Cells in critical limb ischemia in six diabetic patients? a five-year follow-up, Journal of Stem cells & Regenerative Medicine, vol.7, issue.2, p.97, 2011.

V. Sordi, M. L. Malosio, and F. Marchesi, Bone marrow mesenchymal stem cells express a restricted set of functionally active chemokine receptors capable of promoting migration to pancreatic islets, Blood, vol.106, issue.2, pp.419-427, 2005.
DOI : 10.1182/blood-2004-09-3507

A. G. Tzakis, C. Ricordi, and R. , Pancreatic islet transplantation after upper abdominal exeriteration and liver replacement, The Lancet, vol.336, issue.8712, pp.402-405, 1990.
DOI : 10.1016/0140-6736(90)91946-8

A. M. Shapiro, J. R. Lakey, and E. A. Ryan, Islet Transplantation in Seven Patients with Type 1 Diabetes Mellitus Using a Glucocorticoid-Free Immunosuppressive Regimen, New England Journal of Medicine, vol.343, issue.4, pp.230-238, 2000.
DOI : 10.1056/NEJM200007273430401

N. C. Close, B. J. Hering, and T. L. Eggerman, Results From the Inaugural Year of the Collaborative Islet Transplant Registry, Transplantation Proceedings, vol.37, issue.2, pp.1305-1308, 2005.
DOI : 10.1016/j.transproceed.2004.12.117

A. M. Shapiro, J. R. Lakey, B. W. Paty, P. A. Senior, D. L. Bigam et al., Strategic Opportunities in Clinical Islet Transplantation, Transplantation, vol.79, issue.10, pp.1304-1307, 2005.
DOI : 10.1097/01.TP.0000157300.53976.2A

P. Czubak, A. Bojarska-junak, J. Tabarkiewicz, and L. Putowski, A Modified Method of Insulin Producing Cells??? Generation from Bone Marrow-Derived Mesenchymal Stem Cells, Journal of Diabetes Research, vol.5, issue.1, 2014.
DOI : 10.1007/s11427-013-4469-1

L. Khorsandi, F. Nejad-dehbashi, A. Ahangarpour, and M. Hashemitabar, Three-dimensional differentiation of bone marrow-derived mesenchymal stem cells into insulin-producing cells, Tissue and Cell, vol.47, issue.1, pp.66-72, 2015.
DOI : 10.1016/j.tice.2014.11.005

S. Kadam, S. Muthyala, P. Nair, and R. Bhonde, Human Placenta-Derived Mesenchymal Stem Cells and Islet-Like Cell Clusters Generated From These Cells as a Novel Source for Stem Cell Therapy in Diabetes, The Review of Diabetic Studies, vol.7, issue.2, pp.168-182, 2010.
DOI : 10.1900/RDS.2010.7.168

H. Qu, X. Liu, and Y. Ni, Laminin 411 acts as a potent inducer of umbilical cord mesenchymal stem cell differentiation into insulin-producing cells, Journal of Translational Medicine, vol.12, issue.1, 2014.
DOI : 10.1016/j.cell.2008.02.008

P. Tsai, H. Wang, and G. Lin, Undifferentiated Wharton's jelly mesenchymal stem cell transplantation induces 16 Stem Cells International insulin-producing cell differentiation and suppression of T cell-mediated autoimmunity in non-obese diabetic mice, Cell Transplantation, 2014.

K. A. D-'amour, A. D. Agulnick, S. Eliazer, O. G. Kelly, E. Kroon et al., Efficient differentiation of human embryonic stem cells to definitive endoderm, Nature Biotechnology, vol.132, issue.12, pp.1534-1541, 2005.
DOI : 10.1038/nbt1163

S. Chen, M. Borowiak, and J. L. Fox, A small molecule that directs differentiation of human ESCs into the pancreatic lineage, Nature Chemical Biology, vol.644, issue.4, pp.258-265, 2009.
DOI : 10.1038/nchembio.154

E. Kroon, L. A. Martinson, and K. Kadoya, Pancreatic endoderm derived from human embryonic stem cells generates glucose-responsive insulin-secreting cells in vivo, Nature Biotechnology, vol.25, issue.4, pp.443-452, 2008.
DOI : 10.1038/nbt1393

K. Tateishi, J. He, O. Taranova, G. Liang, A. C. et al., Generation of Insulin-secreting Islet-like Clusters from Human Skin Fibroblasts, Journal of Biological Chemistry, vol.283, issue.46, pp.31601-31607, 2008.
DOI : 10.1074/jbc.M806597200

T. Thatava, T. J. Nelson, and R. Edukulla, Indolactam V/GLP-1-mediated differentiation of human iPS cells into glucose-responsive insulin-secreting progeny, Gene Therapy, vol.105, issue.3, pp.283-293, 2011.
DOI : 10.1038/gt.2010.145

R. Jiang, Z. Han, and G. Zhuo, Transplantation of placenta-derived mesenchymal stem cells in type 2 diabetes: a pilot study, Frontiers of Medicine, vol.11, issue.4, pp.94-100, 2011.
DOI : 10.1007/s11684-011-0116-z

J. Hu, X. Yu, and Z. Wang, Long term effects of the implantation of Wharton^|^rsquo;s jelly-derived mesenchymal stem cells from the umbilical cord for newly-onset type 1 diabetes mellitus, Endocrine Journal, vol.60, issue.3, pp.347-357, 2013.
DOI : 10.1507/endocrj.EJ12-0343

D. Kong, X. Zhuang, and D. Wang, Umbilical Cord Mesenchymal Stem Cell Transfusion Ameliorated Hyperglycemia in Patients with Type 2 Diabetes Mellitus, Clinical Laboratory, vol.60, issue.12/2014, pp.1969-1976, 2014.
DOI : 10.7754/Clin.Lab.2014.140305

P. Carlsson, E. Schwarcz, O. Korsgren, and K. L. Blanc, Preserved ??-Cell Function in Type 1 Diabetes by Mesenchymal Stromal Cells, Diabetes, vol.64, issue.2, pp.587-592, 2015.
DOI : 10.2337/db14-0656

S. T. Rashid, S. Corbineau, and N. Hannan, Modeling inherited metabolic disorders of the liver using human induced pluripotent stem cells, Journal of Clinical Investigation, vol.120, issue.9, pp.3127-3136, 2010.
DOI : 10.1172/JCI43122DS1

L. Zhang, J. Ye, V. Decot, J. Stoltz, and N. De-isla, Research on stem cells as candidates to be differentiated into hepatocytes, Bio-Medical Materials and Engineering, vol.22, pp.1-3, 2012.

Q. Zhao, H. Ren, and X. Li, Differentiation of human umbilical cord mesenchymal stromal cells into low immunogenic hepatocyte-like cells, Cytotherapy, vol.11, issue.4, pp.414-426, 2009.
DOI : 10.1080/14653240902849754

T. S. Ramasamy, J. S. Yu, C. Selden, H. Hodgson, and W. Cui, Application of Three-Dimensional Culture Conditions to Human Embryonic Stem Cell-Derived Definitive Endoderm Cells Enhances Hepatocyte Differentiation and Functionality, Tissue Engineering Part A, vol.19, issue.3-4, pp.3-4, 2013.
DOI : 10.1089/ten.tea.2012.0190

J. Jozefczuk, A. Prigione, L. Chavez, and J. Adjaye, Comparative Analysis of Human Embryonic Stem Cell and Induced Pluripotent Stem Cell-Derived Hepatocyte-Like Cells Reveals Current Drawbacks and Possible Strategies for Improved Differentiation, Stem Cells and Development, vol.20, issue.7, pp.1259-1275, 2011.
DOI : 10.1089/scd.2010.0361

K. Si-tayeb, F. K. Noto, and M. Nagaoka, Highly efficient generation of human hepatocyte-like cells from induced pluripotent stem cells, Hepatology, vol.26, issue.1, pp.297-305, 2010.
DOI : 10.1002/hep.23354

M. Mohamadnejad, K. Alimoghaddam, and M. Bagheri, Randomized placebo-controlled trial of mesenchymal stem cell transplantation in decompensated cirrhosis, Liver International, vol.3, issue.Suppl. 1, pp.1490-1496, 2013.
DOI : 10.1111/liv.12228

L. Wang, J. Li, and H. Liu, A pilot study of umbilical cord-derived mesenchymal stem cell transfusion in patients with primary biliary cirrhosis, Journal of Gastroenterology and Hepatology, vol.9, issue.Suppl. 1, pp.85-92, 2013.
DOI : 10.1111/jgh.12029

M. Shi, Z. Zhang, and R. Xu, Human Mesenchymal Stem Cell Transfusion Is Safe and Improves Liver Function in Acute-on-Chronic Liver Failure Patients, STEM CELLS Translational Medicine, vol.113, issue.suppl 2, pp.725-731, 2012.
DOI : 10.5966/sctm.2012-0034

M. M. Amer, S. Z. El-sayed, and W. A. , Clinical and laboratory evaluation of patients with end-stage liver cell failure injected with bone marrow-derived hepatocyte-like cells, European Journal of Gastroenterology & Hepatology, vol.23, issue.10, pp.936-941, 2011.
DOI : 10.1097/MEG.0b013e3283488b00

A. Atala, S. B. Bauer, S. Soker, J. J. Yoo, and A. B. Retik, Tissue-engineered autologous bladders for patients needing cystoplasty, The Lancet, vol.367, issue.9518, pp.1241-1246, 2006.
DOI : 10.1016/S0140-6736(06)68438-9

L. Ma, Y. Yang, and S. C. Sikka, Adipose tissue-derived stem cell-seeded small intestinal submucosa for tunica albuginea grafting and reconstruction, Proceedings of the National Academy of Sciences, vol.109, issue.6, pp.2090-2095, 2012.
DOI : 10.1073/pnas.1113810109

G. Nolazco, I. Kovanecz, and D. Vernet, Effect of muscle-derived stem cells on the restoration of corpora cavernosa smooth muscle and erectile function in the aged rat, BJU International, vol.268, issue.9, pp.1156-1164, 2008.
DOI : 10.1046/j.1464-410X.2003.04373.x

J. Y. Bahk, J. H. Jung, H. Han, S. K. Min, and Y. S. Lee, Treatment of diabetic impotence with umbilical cord blood stem cell intracavernosal transplant: preliminary report of 7 cases, Experimental and Clinical Transplantation, vol.8, issue.2, pp.150-160, 2010.

F. Castiglione, P. Hedlund, and F. Van-der-aa, Intratunical Injection of Human Adipose Tissue???derived Stem Cells Prevents Fibrosis and Is Associated with Improved Erectile Function in a Rat Model of Peyronie's Disease, European Urology, vol.63, issue.3, pp.551-560, 2013.
DOI : 10.1016/j.eururo.2012.09.034

T. K. Ng, V. R. Fortino, D. Pelaez, and H. S. Cheung, Progress of mesenchymal stem cell therapy for neural and retinal diseases, World Journal of Stem Cells, vol.6, issue.2, pp.111-119, 2014.
DOI : 10.4252/wjsc.v6.i2.111

A. Tzameret, I. Sher, and M. Belkin, Transplantation of human bone marrow mesenchymal stem cells as a thin subretinal layer ameliorates retinal degeneration in a rat model of retinal dystrophy, Experimental Eye Research, vol.118, pp.135-144, 2014.
DOI : 10.1016/j.exer.2013.10.023

Y. Hu, H. B. Tan, X. M. Wang, H. Rong, and H. P. Cui, Bone marrow mesenchymal stem cells protect against retinal ganglion cell loss in aged rats with glaucoma, Clinical Interventions in Aging, vol.8, pp.1467-1470, 2013.

L. Ronzoni, P. Bonara, D. Rusconi, C. Frugoni, I. Libani et al., Erythroid differentiation and maturation from peripheral CD34+ cells in liquid culture: Cellular and molecular characterization, Blood Cells, Molecules, and Diseases, vol.40, issue.2, pp.148-155, 2008.
DOI : 10.1016/j.bcmd.2007.07.006

E. Olivier, C. Qiu, and E. E. Bouhassira, Novel, High-Yield Red Blood Cell Production Methods from CD34-Positive Cells Derived from Human Embryonic Stem, Yolk Sac, Fetal Liver, Cord Blood, and Peripheral Blood, STEM CELLS Translational Medicine, vol.117, issue.8, pp.604-614, 2012.
DOI : 10.5966/sctm.2012-0059

URL : http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3659727

F. Ma, Y. Ebihara, and K. Umeda, Generation of functional erythrocytes from human embryonic stem cell-derived definitive hematopoiesis, Proceedings of the National Academy of Sciences, vol.105, issue.35, pp.13087-13092, 2008.
DOI : 10.1073/pnas.0802220105

Y. Ebihara, F. Ma, and K. Tsuji, Generation of red blood cells from human embryonic/induced pluripotent stem cells for blood transfusion, International Journal of Hematology, vol.6, issue.6, pp.610-616, 2012.
DOI : 10.1007/s12185-012-1107-9

C. Yang, A. French, and P. A. Goh, Human induced pluripotent stem cell derived erythroblasts can undergo definitive erythropoiesis and co-express gamma and beta globins, British Journal of Haematology, vol.6, issue.3, pp.435-448, 2014.
DOI : 10.1111/bjh.12910

URL : http://www.ncbi.nlm.nih.gov/pmc/articles/PMC4375519

A. Atala, Engineering organs, Current Opinion in Biotechnology, vol.20, issue.5, pp.575-592, 2009.
DOI : 10.1016/j.copbio.2009.10.003

G. Orlando, P. Baptista, and M. Birchall, Regenerative medicine as applied to solid organ transplantation: current status and future challenges, Transplant International, vol.33, issue.19 Suppl. 3, pp.223-232, 2011.
DOI : 10.1111/j.1432-2277.2010.01182.x

URL : http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3817209

S. F. Badylak, D. Taylor, and K. Uygun, Whole-Organ Tissue Engineering: Decellularization and Recellularization of Three-Dimensional Matrix Scaffolds, Annual Review of Biomedical Engineering, vol.13, issue.1, pp.27-53, 2011.
DOI : 10.1146/annurev-bioeng-071910-124743

P. M. Baptista, G. Orlando, S. Mirmalek-sani, M. Siddiqui, A. Atala et al., Whole organ decellularization - a tool for bioscaffold fabrication and organ bioengineering, 2009 Annual International Conference of the IEEE Engineering in Medicine and Biology Society, pp.6526-6529, 2009.
DOI : 10.1109/IEMBS.2009.5333145

S. V. Murphy and A. Atala, Organ engineering - combining stem cells, biomaterials, and bioreactors to produce bioengineered organs for transplantation, BioEssays, vol.329, issue.3, pp.163-172, 2013.
DOI : 10.1002/bies.201200062

M. E. Scarritt, N. C. Pashos, and B. A. Bunnell, A review of cellularization strategies for tissue engineering of whole organs, Frontiers in Bioengineering and Biotechnology, vol.3, issue.43, 2015.
DOI : 10.3389/fbioe.2015.00043

A. Mathur and J. F. Martin, Stem cells and repair of the heart, The Lancet, vol.364, issue.9429, pp.183-192, 2004.
DOI : 10.1016/S0140-6736(04)16632-4

H. C. Ott, T. S. Matthiesen, and S. Goh, Perfusion-decellularized matrix: using nature's platform to engineer a bioartificial heart, Nature Medicine, vol.57, issue.2, pp.213-221, 2008.
DOI : 10.1038/nm1684

A. A. Khan, S. K. Vishwakarma, A. Bardia, and J. Venkateshwarulu, Repopulation of decellularized whole organ scaffold using stem cells: an emerging technology for the development of neo-organ, Journal of Artificial Organs, vol.6, issue.4, pp.291-300, 2014.
DOI : 10.1007/s10047-014-0780-2

J. M. Singelyn and K. L. Christman, Injectable Materials for the Treatment of Myocardial Infarction and Heart Failure: The Promise of Decellularized Matrices, Journal of Cardiovascular Translational Research, vol.13, issue.9, pp.478-486, 2010.
DOI : 10.1007/s12265-010-9202-x

T. Ota, T. W. Gilbert, S. F. Badylak, D. Schwartzman, and M. A. Zenati, Electromechanical characterization of a tissue-engineered myocardial patch derived from extracellular matrix, The Journal of Thoracic and Cardiovascular Surgery, vol.133, issue.4, pp.979-985, 2007.
DOI : 10.1016/j.jtcvs.2006.11.035

I. A. Potapova, S. V. Doronin, and D. J. Kelly, Enhanced recovery of mechanical function in the canine heart by seeding an extracellular matrix patch with mesenchymal stem cells committed to a cardiac lineage, AJP: Heart and Circulatory Physiology, vol.295, issue.6, pp.2257-2263, 2008.
DOI : 10.1152/ajpheart.00219.2008

E. Martinod, A. Seguin, and M. Holder-espinasse, Tracheal Regeneration Following Tracheal Replacement With an Allogenic Aorta, The Annals of Thoracic Surgery, vol.79, issue.3, pp.942-948, 2005.
DOI : 10.1016/j.athoracsur.2004.08.035

D. M. Radu, A. Seguin, P. Bruneval, A. F. Legendre, A. Carpentier et al., Bronchial Replacement With Arterial Allografts, The Annals of Thoracic Surgery, vol.90, issue.1, pp.252-258, 2010.
DOI : 10.1016/j.athoracsur.2010.03.079

G. M. Roomans, Tissue engineering and the use of stem/progenitor cells for airway epithelium repair, European Cells and Materials, vol.19, pp.284-299, 2010.
DOI : 10.22203/eCM.v019a27

A. Seguin, D. Radu, and M. Holder-espinasse, Tracheal Replacement With Cryopreserved, Decellularized, or Glutaraldehyde-Treated Aortic Allografts, The Annals of Thoracic Surgery, vol.87, issue.3, pp.861-867, 2009.
DOI : 10.1016/j.athoracsur.2008.11.038

T. H. Petersen, E. A. Calle, and L. Zhao, Tissue-Engineered Lungs for in Vivo Implantation, Science, vol.329, issue.5991, pp.538-541, 2010.
DOI : 10.1126/science.1189345

D. A. Chistiakov, Endogenous and exogenous stem cells: a role in lung repair and use in airway tissue engineering and transplantation, Journal of Biomedical Science, vol.17, issue.1, 2010.
DOI : 10.1186/1423-0127-17-92

H. A. Chapman, Toward Lung Regeneration, New England Journal of Medicine, vol.364, issue.19, pp.1867-1868, 2011.
DOI : 10.1056/NEJMe1101800

J. Martin, K. Helm, P. Ruegg, M. Varella-garcia, E. Burnham et al., Adult lung side population cells have mesenchymal stem cell potential, Cytotherapy, vol.10, issue.2, pp.140-151, 2008.
DOI : 10.1080/14653240801895296

L. Jarvinen, L. Badri, and S. Wettlaufer, Lung Resident Mesenchymal Stem Cells Isolated from Human Lung Allografts Inhibit T Cell Proliferation via a Soluble Mediator, The Journal of Immunology, vol.181, issue.6, pp.4389-4396, 2008.
DOI : 10.4049/jimmunol.181.6.4389

X. Gong, Z. Sun, and D. Cui, Isolation and characterization of lung resident mesenchymal stem cells capable of differentiating into alveolar epithelial type II cells, Cell Biology International, vol.59, issue.4, pp.405-411, 2014.
DOI : 10.1002/cbin.10240

K. Chow, J. P. Fessel, and . Kaoriihida-stansbury, Dysfunctional resident lung mesenchymal stem cells contribute to pulmonary microvascular remodeling, Pulmonary Circulation, vol.3, issue.1, pp.31-49, 2013.
DOI : 10.4103/2045-8932.109912

URL : http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3641738

K. M. Antoniou, H. A. Papadaki, and G. Soufla, Investigation of bone marrow mesenchymal stem cells (BM MSCs) involvement in idiopathic pulmonary fibrosis (IPF), Respiratory Medicine, vol.104, issue.10, pp.1535-1542, 2010.
DOI : 10.1016/j.rmed.2010.04.015

Y. Zhang, S. Liao, and M. Yang, Improved Cell Survival and Paracrine Capacity of Human Embryonic Stem Cell-Derived Mesenchymal Stem Cells Promote Therapeutic Potential for Pulmonary Arterial Hypertension, Cell Transplantation, vol.21, issue.10, pp.2225-2239, 2012.
DOI : 10.3727/096368912X653020

K. D. Liu, J. G. Wilson, and H. Zhuo, Design and implementation of the START (STem cells for ARDS Treatment) trial, a phase 1/2 trial of human mesenchymal stem/stromal cells for the treatment of moderate-severe acute respiratory distress syndrome, Annals of Intensive Care, vol.131, issue.4, 2014.
DOI : 10.1186/s13613-014-0022-z

J. G. Wilson, K. D. Liu, and H. Zhuo, Mesenchymal stem (stromal) cells for treatment of ARDS: a phase 1 clinical trial, The Lancet Respiratory Medicine, vol.3, issue.1, pp.24-32, 2015.
DOI : 10.1016/S2213-2600(14)70291-7

H. C. Ott, B. Clippinger, and C. Conrad, Regeneration and orthotopic transplantation of a bioartificial lung, Nature Medicine, vol.70, issue.8, pp.927-933, 2010.
DOI : 10.1038/nm.2193

J. E. Nichols, J. Niles, and M. Riddle, Production and Assessment of Decellularized Pig and Human Lung Scaffolds, Tissue Engineering Part A, vol.19, issue.17-18, pp.17-18, 2013.
DOI : 10.1089/ten.tea.2012.0250

D. E. Wagner, N. R. Bonenfant, and D. Sokocevic, Three-dimensional scaffolds of acellular human and porcine lungs for high throughput studies of lung disease and regeneration, Biomaterials, vol.35, issue.9, pp.2664-2679, 2014.
DOI : 10.1016/j.biomaterials.2013.11.078

R. W. Bonvillain, S. Danchuk, and D. E. Sullivan, Recellularization with Mesenchymal Stem Cells, Tissue Engineering Part A, vol.18, issue.23-24, pp.23-24, 2012.
DOI : 10.1089/ten.tea.2011.0594

J. Cortiella, J. Niles, and A. Cantu, Influence of Acellular Natural Lung Matrix on Murine Embryonic Stem Cell Differentiation and Tissue Formation, Tissue Engineering Part A, vol.16, issue.8, pp.2565-2580, 2010.
DOI : 10.1089/ten.tea.2009.0730

S. Lecht, C. T. Stabler, and A. L. Rylander, Enhanced reseeding of decellularized rodent lungs with mouse embryonic stem cells, Biomaterials, vol.35, issue.10, pp.3252-3262, 2014.
DOI : 10.1016/j.biomaterials.2013.12.093

T. Tsuchiya, A. Sivarapatna, K. Rocco, A. Nanashima, T. Nagayasu et al., Future prospects for tissue engineered lung transplantation, Organogenesis, vol.1, issue.2, pp.196-207, 2014.
DOI : 10.1016/S0022-5223(98)70438-6

URL : http://www.ncbi.nlm.nih.gov/pmc/articles/PMC4154954

A. Soto-gutierrez, L. Zhang, and C. Medberry, A Whole-Organ Regenerative Medicine Approach for Liver Replacement, Tissue Engineering Part C: Methods, vol.17, issue.6, pp.677-686, 2011.
DOI : 10.1089/ten.tec.2010.0698

H. Yagi, K. Fukumitsu, and K. Fukuda, Human-Scale Whole-Organ Bioengineering for Liver Transplantation: A Regenerative Medicine Approach, Cell Transplantation, vol.22, issue.2, pp.231-242, 2013.
DOI : 10.3727/096368912X654939

J. Ye, J. Stoltz, N. De-isla, Y. Liu, Y. Yin et al., An approach to preparing decellularized whole liver organ scaffold in rat, Bio-Medical Materials and Engineering, vol.25, issue.1, pp.159-166, 2015.
URL : https://hal.archives-ouvertes.fr/hal-01452240

W. Jiang, Y. Cheng, M. Yen, Y. Chang, V. W. Yang et al., Cryo-chemical decellularization of the whole liver for mesenchymal stem cells-based functional hepatic tissue engineering, Biomaterials, vol.35, issue.11, pp.3607-3617, 2014.
DOI : 10.1016/j.biomaterials.2014.01.024

R. Ji, N. Zhang, and N. You, The differentiation of MSCs into functional hepatocyte-like cells in a liver biomatrix scaffold and their transplantation into liver-fibrotic mice, Biomaterials, vol.33, issue.35, pp.8995-9008, 2012.
DOI : 10.1016/j.biomaterials.2012.08.058

L. Behr, M. Hekmati, and A. Lucchini, Evaluation of the effect of autologous mesenchymal stem cell injection in a large-animal model of bilateral kidney ischaemia reperfusion injury, Cell Proliferation, vol.55, issue.Suppl. 1, pp.284-297, 2009.
DOI : 10.1111/j.1365-2184.2009.00591.x

L. Behr, M. Hekmati, and G. Fromont, Intra Renal Arterial Injection of Autologous Mesenchymal Stem Cells in an Ovine Model in the Postischemic Kidney, Nephron Physiology, vol.107, issue.3, pp.65-76, 2007.
DOI : 10.1159/000109821

S. K. Nigam, W. Wu, and K. T. Bush, Organogenesis forum lecture, Organogenesis, vol.298, issue.3, pp.137-143, 2008.
DOI : 10.1681/ASN.2005050544

E. Rosines, K. Johkura, and X. Zhang, Tissue Engineering of the Kidney, Tissue Engineering Part A, vol.16, issue.8, pp.2441-2455, 2009.
DOI : 10.1089/ten.tea.2009.0548

K. H. Nakayama, C. A. Batchelder, C. I. Lee, and A. F. , Decellularized Rhesus Monkey Kidney as a Three-Dimensional Scaffold for Renal Tissue Engineering, Tissue Engineering Part A, vol.16, issue.7, pp.2207-2216, 2010.
DOI : 10.1089/ten.tea.2009.0602

URL : http://www.ncbi.nlm.nih.gov/pmc/articles/PMC2947947

F. Tasnim, R. Deng, and M. Hu, Achievements and challenges in bioartificial kidney development, Fibrogenesis & Tissue Repair, vol.3, issue.1, 2010.
DOI : 10.1186/1755-1536-3-14

URL : http://www.ncbi.nlm.nih.gov/pmc/articles/PMC2925816

S. R. Baglio, D. M. Pegtel, and N. Baldini, Mesenchymal stem cell secreted vesicles provide novel opportunities in (stem) cellfree therapy, Frontiers in Physiology, vol.3, issue.359, 2012.

R. C. Lai, R. W. Yeo, and S. K. Lim, Mesenchymal stem cell exosomes, Mesenchymal stem cell exosomes, pp.82-88, 2015.
DOI : 10.1016/j.semcdb.2015.03.001

S. E. Haynesworth, M. A. Baber, and A. I. Caplan, Cytokine expression by human marrow-derived mesenchymal progenitor cells in vitro: Effects of dexamethasone and IL-1??, Journal of Cellular Physiology, vol.177, issue.3, pp.585-592, 1996.
DOI : 10.1002/(SICI)1097-4652(199603)166:3<585::AID-JCP13>3.0.CO;2-6

B. Yu, X. Zhang, and X. Li, Exosomes Derived from Mesenchymal Stem Cells, International Journal of Molecular Sciences, vol.15, issue.3, pp.4142-4157, 2014.
DOI : 10.3390/ijms15034142

J. R. Lavoie and M. Rosu-myles, Uncovering the secretes of mesenchymal stem cells, Biochimie, vol.95, issue.12, pp.2212-2221, 2013.
DOI : 10.1016/j.biochi.2013.06.017

K. C. Vallabhaneni, P. Penfornis, and S. Dhule, Extracellular vesicles from bone marrow mesenchymal stem/stromal cells transport tumor regulatory microRNA, proteins, and metabolites, Oncotarget, vol.6, issue.7, pp.4953-4967, 2015.
DOI : 10.18632/oncotarget.3211

URL : https://hal.archives-ouvertes.fr/inserm-01179843

T. Kinnaird, E. Stabile, and M. S. Burnett, Marrow-Derived Stromal Cells Express Genes Encoding a Broad Spectrum of Arteriogenic Cytokines and Promote In Vitro and In Vivo Arteriogenesis Through Paracrine Mechanisms, Circulation Research, vol.94, issue.5, pp.678-685, 2004.
DOI : 10.1161/01.RES.0000118601.37875.AC

M. Gnecchi, H. He, and N. Noiseux, Evidence supporting paracrine hypothesis for Akt-modified mesenchymal stem cell-mediated cardiac protection and functional improvement, The FASEB Journal, vol.20, issue.6, pp.661-669, 2006.
DOI : 10.1096/fj.05-5211com

A. Shabbir, A. Coz, L. Rodriguez, M. Salgado, and E. Badiavas, Mesenchymal Stem Cell Exosomes Induce Proliferation and Migration of Normal and Chronic Wound Fibroblasts, and Enhance Angiogenesis In Vitro, Stem Cells and Development, vol.24, issue.14, 2015.
DOI : 10.1089/scd.2014.0316

Y. Zhang, M. Chopp, and Y. Meng, Effect of exosomes derived from multipluripotent mesenchymal stromal cells on functional recovery and neurovascular plasticity in rats after traumatic brain injury, Journal of Neurosurgery, vol.122, issue.4, pp.856-867, 2015.
DOI : 10.3171/2014.11.JNS14770

Y. Zhu, X. Feng, and J. Abbott, Endotoxin-Induced Acute Lung Injury in Mice, STEM CELLS, vol.286, issue.1, pp.116-125, 2014.
DOI : 10.1002/stem.1504

K. Deng, D. L. Lin, and B. Hanzlicek, Mesenchymal stem cells and their secretome partially restore nerve and urethral function in a dual muscle and nerve injury stress urinary incontinence model, AJP: Renal Physiology, vol.308, issue.2, pp.92-100, 2015.
DOI : 10.1152/ajprenal.00510.2014

M. Ono, N. Kosaka, and N. Tominaga, Exosomes from bone marrow mesenchymal stem cells contain a microRNA that, Stem Cells International

J. Lee, S. Park, and B. Jung, Exosomes derived from mesenchymal stem cells suppress angiogenesis by downregulating VEGF expression in breast cancer cells, PLoS ONE, vol.8, issue.12, 2013.