, biomarkers for chronic rejection of renal allografts, Transplantation, vol.87, pp.1505-1513, 2009.

J. Gralla, S. Tong, and A. C. Wiseman, The impact of human leukocyte antigen mismatching on sensitization rates and subsequent retransplantation after first graft failure in pediatric renal transplant recipients, Transplantation, vol.95, pp.1218-1224, 2013.

D. P. Lucas, M. S. Leffell, and A. A. Zachary, Differences in immunogenicity of HLA antigens and the impact of cross-reactivity on the humoral response, Transplantation, vol.99, pp.77-85, 2015.

G. Opelz and T. Wujciak, The influence of HLA compatibility on graft survival after heart transplantation. The Collaborative Transplant Study, N Engl J Med, vol.330, pp.816-819, 1994.

G. Opelz, J. Mytilineos, and S. Scherer, Survival of DNA HLA-DR typed and matched cadaver kidney transplants. The Collaborative Transplant Study, Lancet, vol.338, pp.461-463, 1991.

S. Yamanaka, Induced pluripotent stem cells: past, present, and future, Cell Stem Cell, vol.10, pp.678-684, 2012.

F. Sampaziotis, C. De-brito, M. Madrigal, and P. , Cholangiocytes derived from human induced pluripotent stem cells for disease modeling and drug validation, Nat Biotechnol, vol.33, pp.845-852, 2015.

N. R. Hannan, F. Sampaziotis, and C. P. Segeritz, Generation of distal airway epithelium from multipotent human foregut stem cells, Stem Cells Dev, vol.24, pp.1680-1690, 2015.

M. Baxter, S. Withey, and S. Harrison, Phenotypic and functional analyses show stem cell-derived hepatocyte-like cells better mimic fetal rather than adult hepatocytes, J Hepatol, vol.62, pp.581-589, 2015.
DOI : 10.1016/j.jhep.2014.10.016

URL : https://doi.org/10.1016/j.jhep.2014.10.016

I. Cebola, S. A. Rodriguez-segui, and C. H. Cho, TEAD and YAP regulate the enhancer network of human embryonic pancreatic progenitors, Nat Cell Biol, vol.17, pp.615-626, 2015.

N. Dianat, C. Steichen, and L. Vallier, Human pluripotent stem cells for modelling human liver diseases and cell therapy, Curr Gene Ther, vol.13, pp.120-132, 2013.

T. Takebe, K. Sekine, and M. Enomura, Vascularized and functional human liver from an iPSC-derived organ bud transplant, Nature, vol.499, pp.481-484, 2013.

T. Takebe, R. R. Zhang, and H. Koike, Generation of a vascularized and functional human liver from an iPSC-derived organ bud transplant, Nat Protoc, vol.9, pp.396-409, 2014.

A. O. Cramer and R. E. Maclaren, Translating induced pluripotent stem cells from bench to bedside: application to retinal diseases, Curr Gene Ther, vol.13, pp.139-151, 2013.

W. K. Song, K. M. Park, and H. J. Kim, Treatment of macular degeneration using embryonic stem cell-derived retinal pigment epithelium: preliminary results in Asian patients, Stem cell reports, vol.4, pp.860-872, 2015.

S. D. Schwartz, C. D. Regillo, and B. L. Lam, Human embryonic stem cell-derived retinal pigment epithelium in patients with age-related macular degeneration and Stargardt's macular dystrophy: follow-up of two open-label phase 1/2 studies, Lancet, vol.385, pp.509-516, 2015.

G. F. Rousseau, M. C. Giarratana, and L. Douay, Large-scale production of red blood cells from stem cells: what are the technical challenges ahead?, Biotechnol J, vol.9, pp.28-38, 2014.

T. M. Neildez-nguyen, H. Wajcman, and M. C. Marden, Human erythroid cells produced ex vivo at large scale differentiate into red blood cells in vivo, Nat Biotechnol, vol.20, pp.467-472, 2002.

M. C. Giarratana, L. Kobari, and H. Lapillonne, Ex vivo generation of fully mature human red blood cells from hematopoietic stem cells, Nat Biotechnol, vol.23, pp.69-74, 2005.

M. C. Giarratana, H. Rouard, and A. Dumont, Proof of principle for transfusion of in vitro-generated red blood cells, Blood, vol.118, pp.5071-5079, 2011.
URL : https://hal.archives-ouvertes.fr/hal-00692474

D. Darghouth, M. C. Giarratana, and L. Oliveira, Bio-engineered and native red blood cells from cord blood exhibit the same metabolomic profile, Haematologica, vol.101, pp.220-222, 2016.

G. Q. Daley, Deriving blood stem cells from pluripotent stem cells for research and therapy, Best Pract Res Clin Haematol, vol.27, pp.293-297, 2014.

M. Ackermann, S. Liebhaber, and J. H. Klusmann, Lost in translation: pluripotent stem cell-derived hematopoiesis, EMBO Mol Med, vol.7, pp.1388-1402, 2015.

H. Lapillonne, L. Kobari, and C. Mazurier, Red blood cell generation from human induced pluripotent stem cells: perspectives for transfusion medicine, Haematologica, vol.95, pp.1651-1659, 2010.

L. Kobari, F. Yates, and N. Oudrhiri, Human induced pluripotent stem cells can reach complete terminal maturation: in vivo and in vitro evidence in the erythropoietic differentiation model, Haematologica, vol.97, pp.1795-1803, 2012.

S. Rouzbeh, L. Kobari, and M. Cambot, Molecular signature of erythroblast enucleation in human embryonic stem cells, Stem Cells, vol.33, pp.2431-2441, 2015.
URL : https://hal.archives-ouvertes.fr/hal-01515305

S. Doulatov, L. T. Vo, and S. S. Chou, Induction of multipotential hematopoietic progenitors from human pluripotent stem cells via respecification of lineage-restricted precursors, Cell Stem Cell, vol.13, pp.459-470, 2013.

V. M. Sandler, R. Lis, and Y. Liu, Reprogramming human endothelial cells to haematopoietic cells requires vascular induction, Nature, vol.511, pp.312-318, 2014.
DOI : 10.1038/nature13547

URL : http://europepmc.org/articles/pmc4159670?pdf=render

P. J. Fairchild, A. Leishman, and P. Sachamitr, Dendritic cells and pluripotency: unlikely allies in the pursuit of immunotherapy, Regen Med, vol.10, pp.275-286, 2015.

K. M. Silk, J. D. Silk, and N. Ichiryu, Cross-presentation of tumour antigens by human induced pluripotent stem cell-derived CD141(+)XCR1+ dendritic cells, Gene Ther, vol.19, pp.1035-1040, 2012.

A. Bachem, S. Guttler, and E. Hartung, Superior antigen crosspresentation and XCR1 expression define human CD11c + CD141+ cells as homologues of mouse CD8+ dendritic cells, J Exp Med, vol.207, pp.1273-1281, 2010.

M. Themeli, I. Riviere, and M. Sadelain, New cell sources for T cell engineering and adoptive immunotherapy, Cell Stem Cell, vol.16, pp.357-366, 2015.

M. Themeli, C. C. Kloss, and G. Ciriello, Generation of tumor-targeted human T lymphocytes from induced pluripotent stem cells for cancer therapy, Nat Biotechnol, vol.31, pp.928-933, 2013.

P. D. Hsu, E. S. Lander, and F. Zhang, Development and applications of CRISPRCas9 for genome engineering, Cell, vol.157, pp.1262-1278

P. S. Woll, B. Grzywacz, and X. Tian, Human embryonic stem cells differentiate into a homogeneous population of natural killer cells with potent in vivo antitumor activity, Blood, vol.113, pp.6094-6101, 2009.

Z. Ni, D. A. Knorr, and C. L. Clouser, Human pluripotent stem cells produce natural killer cells that mediate anti-HIV-1 activity by utilizing diverse cellular mechanisms, J Virol, vol.85, pp.43-50, 2011.
DOI : 10.1128/jvi.01774-10

URL : https://jvi.asm.org/content/85/1/43.full.pdf

D. A. Knorr, Z. Ni, and D. Hermanson, Clinical-scale derivation of natural killer cells from human pluripotent stem cells for cancer therapy, Stem Cells Transl Med, vol.2, pp.274-283, 2013.

D. L. Hermanson, L. Bendzick, and L. Pribyl, Induced pluripotent stem cellderived natural killer cells for treatment of ovarian cancer, Stem Cells, vol.34, pp.93-101, 2016.

A. Dorronsoro, I. Ferrin, and J. M. Salcedo, Human mesenchymal stromal cells modulate T-cell responses through TNF-?-mediated activation of NF-kB, Eur J Immunol, vol.44, pp.480-488, 2014.

L. Tejedor, G. Mausset-bonnefont, and A. L. , Glucocorticoidinduced leucine zipper governs the therapeutic potential of mesenchymal stem cells by inducing a switch from pathogenic to regulatory Th17 cells in a mouse model of collagen-induced arthritis, Arthritis Rheumatol, vol.67, pp.1514-1524, 2015.
URL : https://hal.archives-ouvertes.fr/hal-01834130

L. Ipseiz, N. Espinosa-carrasco, and G. , PPAR?/? directs the therapeutic potential of mesenchymal stem cells in arthritis, Ann Rheum Dis, vol.75, pp.2166-2174, 2016.

W. Shan, P. S. Palkar, and I. A. Murray, Ligand activation of peroxisome proliferator-activated receptor beta/delta (PPARbeta/delta) attenuates carbon tetrachloride hepatotoxicity by downregulating proinflammatory gene expression, Toxicol Sci, vol.105, pp.418-428, 2008.

R. A. Daynes and D. C. Jones, Emerging roles of PPARs in inflammation and immunity, Nat Rev Immunol, vol.2, pp.748-759, 2002.

Y. Rival, N. Beneteau, and T. Taillandier, PPARalpha and PPARdelta activators inhibit cytokine-induced nuclear translocation of NF-kappaB and expression of VCAM-1 in EAhy926 endothelial cells, Eur J Pharmacol, vol.435, pp.143-151, 2002.

D. Guijarro, M. Lebrin, and O. Lairez, Intramyocardial transplantation of mesenchymal stromal cells for chronic myocardial ischemia and impaired left ventricular function: Results of the MESAMI 1 pilot trial, Int J Cardiol, vol.209, pp.258-265, 2016.
URL : https://hal.archives-ouvertes.fr/inserm-01269644

X. P. Huang, Z. Sun, and Y. Miyagi, Differentiation of allogeneic mesenchymal stem cells induces immunogenicity and limits their long-term benefits for myocardial repair, Circulation, vol.122, pp.2419-2429, 2010.

M. J. Crop, S. S. Korevaar, and R. De-kuiper, Human mesenchymal stem cells are susceptible to lysis by CD8(+) T cells and NK cells, Cell Transplant, vol.20, pp.1547-1559, 2011.

W. R. Prather, A. Toren, and M. Meiron, The role of placental-derived adherent stromal cell (PLX-PAD) in the treatment of critical limb ischemia, Cytotherapy, vol.11, pp.427-434, 2009.

C. Consentius, P. Reinke, and H. D. Volk, Immunogenicity of allogeneic mesenchymal stromal cells: what has been seen in vitro and in vivo, Regen Med, vol.10, pp.305-315, 2015.

L. M. Ball, M. E. Bernardo, and H. Roelofs, Multiple infusions of mesenchymal stromal cells induce sustained remission in children with steroidrefractory, grade III-IV acute graft-versus-host disease, Br J Haematol, vol.163, pp.501-509, 2013.

J. Galipeau, The mesenchymal stromal cells dilemma-does a negative phase III trial of random donor mesenchymal stromal cells in steroid-resistant graftversus-host disease represent a death knell or a bump in the road?, Cytotherapy, vol.15, pp.2-8, 2013.

L. M. Tobin, M. E. Healy, and K. English, Human mesenchymal stem cells suppress donor CD4(+) T cell proliferation and reduce pathology in a humanized mouse model of acute graft-versus-host disease, Clin Exp Immunol, vol.172, pp.333-348, 2013.

J. W. Streilein, Ocular immune privilege: therapeutic opportunities from an experiment of nature, Nat Rev Immunol, vol.3, pp.879-889, 2003.

U. Pleyer and S. Schlickeiser, The taming of the shrew? The immunology of corneal transplantation, Acta Ophthalmol, vol.87, pp.488-497, 2009.

O. Treacy, L. O'flynn, and A. E. Ryan, Mesenchymal stem cell therapy promotes corneal allograft survival in rats by local and systemic immunomodulation, Am J Transplant, vol.14, pp.2023-2036, 2014.

Z. Jia, C. Jiao, and S. Zhao, Immunomodulatory effects of mesenchymal stem cells in a rat corneal allograft rejection model, Exp Eye Res, vol.102, pp.44-49, 2012.

M. Omoto, K. R. Katikireddy, and A. Rezazadeh, Mesenchymal stem cells home to inflamed ocular surface and suppress allosensitization in corneal transplantation, Invest Ophthalmol Vis Sci, vol.55, pp.6631-6638, 2014.

J. H. Ko, H. J. Lee, and H. J. Jeong, Mesenchymal stem/stromal cells precondition lung monocytes/macrophages to produce tolerance against allo-and autoimmunity in the eye, Proc Natl Acad Sci, vol.113, pp.158-163, 2016.

J. Y. Oh, R. H. Lee, and J. M. Yu, Intravenous mesenchymal stem cells prevented rejection of allogeneic corneal transplants by aborting the early inflammatory response, Mol Ther, vol.20, pp.2143-2152, 2012.

Z. Rong, M. Wang, and Z. Hu, An effective approach to prevent immune rejection of human ESC-derived allografts, Cell Stem Cell, vol.14, pp.121-130, 2014.

T. Zhao, Z. N. Zhang, and Z. Rong, Immunogenicity of induced pluripotent stem cells, Nature, vol.474, pp.212-215, 2011.

Q. Lu, M. Yu, and C. Shen, Negligible immunogenicity of induced pluripotent stem cells derived from human skin fibroblasts, PLoS One, vol.9, p.114949, 2014.

R. Araki, M. Uda, and Y. Hoki, Negligible immunogenicity of terminally differentiated cells derived from induced pluripotent or embryonic stem cells, Nature, vol.494, pp.100-104, 2013.

P. Guha, J. W. Morgan, and G. Mostoslavsky, Lack of immune response to differentiated cells derived from syngeneic induced pluripotent stem cells, Cell Stem Cell, vol.12, pp.407-412, 2013.

S. Kaneko and S. Yamanaka, To be immunogenic, or not to be: that's the iPSC question, Cell Stem Cell, vol.12, pp.385-386, 2013.

T. Zhao, Z. N. Zhang, and P. D. Westenskow, Humanized mice reveal differential immunogenicity of cells derived from autologous induced pluripotent stem cells, Cell Stem Cell, vol.17, pp.353-359, 2015.

H. Song, S. K. Chung, and Y. Xu, Modeling disease in human ESCs using an efficient BAC-based homologous recombination system, Cell Stem Cell, vol.6, pp.80-89, 2010.

J. Park and J. E. Babensee, Differential functional effects of biomaterials on dendritic cell maturation, Acta Biomater, vol.8, pp.3606-3617, 2012.

J. Park, M. H. Gerber, and J. E. Babensee, Phenotype and polarization of autologous T cells by biomaterial-treated dendritic cells, J Biomed Mater Res A, vol.103, pp.170-184, 2015.

S. Ville, N. Poirier, and G. Blancho, Co-stimulatory blockade of the CD28/ CD80-86/CTLA-4 balance in transplantation: impact on memory T cells?, Front Immunol, vol.6, p.411, 2015.
URL : https://hal.archives-ouvertes.fr/inserm-02148498

N. Poirier, M. Chevalier, and C. Mary, Selective CD28 antagonist blunts memory immune responses and promotes long-term control of skin inflammation in nonhuman primates, J Immunol, vol.196, pp.274-283, 2016.
URL : https://hal.archives-ouvertes.fr/inserm-02150299

M. P. Vierboom, E. Breedveld, and Y. S. Kap, Clinical efficacy of a new CD28-targeting antagonist of Tcell co-stimulation in a non-human primate model of collagen-induced arthritis, Clin Exp Immunol, vol.183, pp.405-418, 2016.

K. G. Haanstra, K. Dijkman, and N. Bashir, Selective blockade of CD28-mediated T cell costimulation protects rhesus monkeys against acute fatal experimental autoimmune encephalomyelitis, J Immunol, vol.194, pp.1454-1466, 2015.
URL : https://hal.archives-ouvertes.fr/inserm-02148517

N. Poirier, N. Dilek, and C. Mary, FR104, an antagonist anti-CD28 monovalent fab' antibody, prevents alloimmunization and allows calcineurin inhibitor minimization in nonhuman primate renal allograft, Am J Transplant, vol.15, pp.88-100, 2015.
URL : https://hal.archives-ouvertes.fr/inserm-02148443

N. Poirier, T. Haudebourg, and C. Brignone, Antibody-mediated depletion of lymphocyte-activation gene-3 (LAG-3(+))-activated T lymphocytes prevents delayed-type hypersensitivity in non-human primates, Clin Exp Immunol, vol.164, pp.265-274, 2011.