N. Hwang, C. Zhang, Y. Hwang, and S. Varghese, Mesenchymal stem cell differentiation and roles in regenerative medicine, Wiley Interdisciplinary Reviews: Systems Biology and Medicine, vol.18, issue.suppl 1, pp.97-106, 2009.
DOI : 10.1002/wsbm.26

L. Ortiz, F. Gambelli, C. Mcbride, D. Gaupp, M. Baddoo et al., Mesenchymal stem cell engraftment in lung is enhanced in response to bleomycin exposure and ameliorates its fibrotic effects, Proceedings of the National Academy of Sciences, vol.100, issue.14, pp.8407-8411, 2003.
DOI : 10.1073/pnas.1432929100

F. Zhao, Y. Zhang, Y. Liu, J. Zhou, Z. Li et al., Therapeutic Effects of Bone Marrow-Derived Mesenchymal Stem Cells Engraftment on Bleomycin-Induced Lung Injury in Rats, Transplantation Proceedings, vol.40, issue.5, pp.1700-1705, 2008.
DOI : 10.1016/j.transproceed.2008.01.080

A. Croft and S. Przyborski, Formation of Neurons by Non-Neural Adult Stem Cells: Potential Mechanism Implicates an Artifact of Growth in Culture, Stem Cells, vol.384, issue.8, pp.1841-18512005, 2006.
DOI : 10.1634/stemcells.2005-0609

L. Von-bahr, I. Batsis, G. Moll, M. Hagg, A. Szakos et al., Analysis of Tissues Following Mesenchymal Stromal Cell Therapy in Humans Indicates Limited Long-Term Engraftment and No Ectopic Tissue Formation, STEM CELLS, vol.10, issue.7, pp.1575-1578, 1118.
DOI : 10.1002/stem.1118

K. Huang, X. Kang, X. Wang, S. Wu, X. J. Li et al., Conversion of bone marrow mesenchymal stem cells into type II alveolar epithelial cells reduces pulmonary fibrosis by decreasing oxidative stress in rats, Molecular Medicine Reports, vol.11, issue.3, pp.1685-1692, 2015.
DOI : 10.3892/mmr.2014.2981

S. Hashemi, S. Ghods, F. Kolodgie, K. Parcham-azad, M. Keane et al., A placebo controlled, dose-ranging, safety study of allogenic mesenchymal stem cells injected by endomyocardial delivery after an acute myocardial infarction, European Heart Journal, vol.29, issue.2, pp.251-259, 2008.
DOI : 10.1093/eurheartj/ehm559

A. Leroy, B. Nottelet, C. Bony, C. Pinese, C. B. Garric et al., PLA-poloxamer/poloxamine copolymers for ligament tissue engineering: sound macromolecular design for degradable scaffolds and MSC differentiation, Biomater. Sci., vol.9, issue.4, 2015.
DOI : 10.1039/C4BM00433G

URL : https://hal.archives-ouvertes.fr/hal-01369239

C. Bouffi, O. Thomas, C. Bony, A. Giteau, M. Venier-julienne et al., The role of pharmacologically active microcarriers releasing TGF-??3 in cartilage formation in vivo by mesenchymal stem cells, Biomaterials, vol.31, issue.25, pp.6485-6493, 2010.
DOI : 10.1016/j.biomaterials.2010.05.013

C. Jorgensen, J. Gordeladze, and D. Noel, Tissue engineering through autologous mesenchymal stem cells, Current Opinion in Biotechnology, vol.15, issue.5, pp.406-410, 2004.
DOI : 10.1016/j.copbio.2004.08.003

C. Vinatier, C. Bouffi, C. Merceron, J. Gordeladze, J. Brondello et al., Cartilage Tissue Engineering: Towards a Biomaterial-Assisted Mesenchymal Stem Cell Therapy, Current Stem Cell Research & Therapy, vol.4, issue.4, pp.318-329, 2009.
DOI : 10.2174/157488809789649205

URL : https://hal.archives-ouvertes.fr/inserm-00423696

M. Mathieu, S. Vigier, M. Labour, C. Jorgensen, E. Belamie et al., Induction of mesenchymal stem cell differentiation and cartilage formation by cross-linker-free collagen microspheres, European Cells and Materials, vol.28, pp.82-96, 2014.
DOI : 10.22203/eCM.v028a07

M. Morille, T. Van-thanh, X. Garric, J. Cayon, J. Coudane et al., New PLGA???P188???PLGA matrix enhances TGF-??3 release from pharmacologically active microcarriers and promotes chondrogenesis of mesenchymal stem cells, Journal of Controlled Release, vol.170, issue.1, pp.99-110, 2013.
DOI : 10.1016/j.jconrel.2013.04.017

URL : https://hal.archives-ouvertes.fr/hal-00881128

S. Portron, V. Hivernaud, C. Merceron, J. Lesoeur, M. Masson et al., Inverse Regulation of Early and Late Chondrogenic Differentiation by Oxygen Tension Provides Cues for Stem Cell-Based Cartilage Tissue Engineering, Cellular Physiology and Biochemistry, vol.35, issue.3, pp.841-85710000369742, 1159.
DOI : 10.1159/000369742

S. Eap, L. Keller, J. Schiavi, O. Huck, L. Jacomine et al., ) A living thick nanofibrous implant bifunctionalized with active growth factor and stem cells for bone regeneration, Int J Nanomedicine, vol.10, pp.1061-107510, 2015.

I. Kim, J. Ko, H. Lee, S. Do, and K. Park, Mesenchymal cells condensation-inducible mesh scaffolds for cartilage tissue engineering, Biomaterials, vol.85, pp.18-29, 2016.
DOI : 10.1016/j.biomaterials.2016.01.048

Y. Pers, M. Ruiz, D. Noel, and C. Jorgensen, Mesenchymal stem cells for the management of inflammation in osteoarthritis: state of the art and perspectives, Osteoarthritis and Cartilage, vol.23, issue.11, pp.2027-2035, 2015.
DOI : 10.1016/j.joca.2015.07.004

M. Maumus, C. Jorgensen, and D. Noel, Mesenchymal stem cells in regenerative medicine applied to??rheumatic diseases: Role of secretome and exosomes, Biochimie, vol.95, issue.12, 2013.
DOI : 10.1016/j.biochi.2013.04.017

URL : https://hal.archives-ouvertes.fr/inserm-00832507

K. Toupet, M. Maumus, P. Luz-crawford, E. Lombardo, J. Lopez-belmonte et al., Survival and Biodistribution of Xenogenic Adipose Mesenchymal Stem Cells Is Not Affected by the Degree of Inflammation in Arthritis, PLOS ONE, vol.5, issue.1, 2015.
DOI : 10.1371/journal.pone.0114962.g004

K. Toupet, M. Maumus, J. Peyrafitte, P. Bourin, P. Van-lent et al., Long-Term Detection of Human Adipose-Derived Mesenchymal Stem Cells After Intraarticular Injection in SCID Mice, Arthritis & Rheumatism, vol.46, issue.7, pp.1786-1794, 2013.
DOI : 10.1002/art.37960

A. Maria, K. Toupet, C. Bony, N. Pirot, M. Vozenin et al., Anti-fibrotic, anti-oxidant and immunomodulatory effects of mesenchymal stem cells in HOCl-induced systemic sclerosis, 2015.

T. Meyerrose, D. Ugarte, D. Hofling, A. Herrbrich, P. Cordonnier et al., In Vivo Distribution of Human Adipose-Derived Mesenchymal Stem Cells in Novel Xenotransplantation Models, STEM CELLS, vol.12, issue.1, pp.220-2272006, 2007.
DOI : 10.1634/stemcells.2006-0243

J. Gao, J. Dennis, R. Muzic, M. Lundberg, and A. Caplan, The Dynamic in vivo Distribution of Bone Marrow-Derived Mesenchymal Stem Cells after Infusion, Cells Tissues Organs, vol.169, issue.1, pp.12-2047856, 2001.
DOI : 10.1159/000047856

U. Fischer, M. Harting, F. Jimenez, W. Monzon-posadas, H. Xue et al., Pulmonary Passage is a Major Obstacle for Intravenous Stem Cell Delivery: The Pulmonary First-Pass Effect, Stem Cells and Development, vol.18, issue.5, pp.683-6920253, 2008.
DOI : 10.1089/scd.2008.0253

Y. Moodley, D. Atienza, U. Manuelpillai, C. Samuel, J. Tchongue et al., Human Umbilical Cord Mesenchymal Stem Cells Reduce Fibrosis of Bleomycin-Induced Lung Injury, The American Journal of Pathology, vol.175, issue.1, pp.303-313, 2009.
DOI : 10.2353/ajpath.2009.080629

D. Wolf, R. A. Seckinger, A. Katus, H. Kuecherer, H. Hansen et al., Dose-Dependent Effects of Intravenous Allogeneic Mesenchymal Stem Cells in the Infarcted Porcine Heart, Stem Cells and Development, vol.18, issue.2, pp.321-3290019, 2008.
DOI : 10.1089/scd.2008.0019

S. Joo, K. Cho, Y. Jung, H. Kim, S. Park et al., Mesenchymal stromal cells inhibit graft-versus-host disease of mice in a dose-dependent manner, Cytotherapy, vol.12, issue.3, pp.361-37010, 2010.
DOI : 10.3109/14653240903502712

G. Desando, C. Cavallo, F. Sartoni, L. Martini, A. Parrilli et al., Intra-articular delivery of adipose derived stromal cells attenuates osteoarthritis progression in an experimental rabbit model, Arthritis Research & Therapy, vol.15, issue.1, pp.22-32, 2013.
DOI : 10.1517/14712590903039684

M. Murphy, K. Moncivais, and A. Caplan, Mesenchymal stem cells: environmentally responsive therapeutics for regenerative medicine, Experimental & Molecular Medicine, vol.154, issue.11, 2013.
DOI : 10.3324/haematol.11869

L. Blanc, K. Mougiakakos, and D. , Multipotent mesenchymal stromal cells and the innate immune system, Nature Reviews Immunology, vol.107, issue.5, pp.383-396, 2012.
DOI : 10.1038/nri3209

G. Spaggiari, A. Capobianco, S. Becchetti, M. Mingari, and L. Moretta, Mesenchymal stem cell-natural killer cell interactions: evidence that activated NK cells are capable of killing MSCs, whereas MSCs can inhibit IL-2-induced NK-cell proliferation, Blood, vol.107, issue.4, pp.1484-149010, 2005.
DOI : 10.1182/blood-2005-07-2775

A. Nauta, G. Westerhuis, A. Kruisselbrink, E. Lurvink, R. Willemze et al., Donor-derived mesenchymal stem cells are immunogenic in an allogeneic host and stimulate donor graft rejection in a nonmyeloablative setting, Blood, vol.108, issue.6, pp.2114-2120, 2006.
DOI : 10.1182/blood-2005-11-011650

M. Crop, S. Korevaar, R. De-kuiper, J. Ij, N. Van-besouw et al., Human Mesenchymal Stem Cells Are Susceptible to Lysis by CD8<SUP>+</SUP> T Cells and NK Cells, Cell Transplantation, vol.20, issue.10, pp.1547-155910, 2011.
DOI : 10.3727/096368910X564076

R. Lim, P. Milton, S. Murphy, H. Dickinson, S. Chan et al., Human Mesenchymal Stem Cells Reduce Lung Injury in Immunocompromised Mice but Not in Immunocompetent Mice, Respiration, vol.85, issue.4, pp.332-341, 2013.
DOI : 10.1159/000343078

M. Breitbach, T. Bostani, W. Roell, Y. Xia, O. Dewald et al., Potential risks of bone marrow cell transplantation into infarcted hearts, Blood, vol.110, issue.4, pp.1362-136910, 2007.
DOI : 10.1182/blood-2006-12-063412

C. Bouffi, F. Djouad, M. Mathieu, D. Noel, and C. Jorgensen, Multipotent mesenchymal stromal cells and rheumatoid arthritis: risk or benefit?, Rheumatology, vol.48, issue.10, pp.1185-1189, 2009.
DOI : 10.1093/rheumatology/kep162

P. Mishra, P. Mishra, J. Glod, and D. Banerjee, Mesenchymal Stem Cells: Flip Side of the Coin, Cancer Research, vol.69, issue.4, pp.1255-1258, 2009.
DOI : 10.1158/0008-5472.CAN-08-3562

M. Miura, Y. Miura, H. Padilla-nash, A. Molinolo, B. Fu et al., Accumulated Chromosomal Instability in Murine Bone Marrow Mesenchymal Stem Cells Leads to Malignant Transformation, STEM CELLS, vol.6, issue.4, pp.1095-11032005, 2006.
DOI : 10.1634/stemcells.2005-0403

J. Tolar, A. Nauta, M. Osborn, P. Mortari, A. Mcelmurry et al., Sarcoma Derived from Cultured Mesenchymal Stem Cells, Stem Cells, vol.15, issue.2, pp.371-379, 2007.
DOI : 10.1634/stemcells.2005-0620

K. Tarte, J. Gaillard, J. Lataillade, L. Fouillard, M. Becker et al., Clinical-grade production of human mesenchymal stromal cells: occurrence of aneuploidy without transformation, Blood, vol.115, issue.8, pp.1549-155310, 2010.
DOI : 10.1182/blood-2009-05-219907

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

M. Bernardo, N. Zaffaroni, F. Novara, A. Cometa, M. Avanzini et al., Human Bone Marrow Derived Mesenchymal Stem Cells Do Not Undergo Transformation after Long-term In vitro Culture and Do Not Exhibit Telomere Maintenance Mechanisms, Cancer Research, vol.67, issue.19, pp.9142-9149100008, 1158.
DOI : 10.1158/0008-5472.CAN-06-4690

F. Djouad, C. Bony, F. Apparailly, P. Louis-plence, C. Jorgensen et al., Earlier Onset of Syngeneic Tumors in the Presence of Mesenchymal Stem Cells, Transplantation, vol.82, issue.8, 2006.
DOI : 10.1097/01.tp.0000236098.13804.0b

M. Lalu, L. Mcintyre, C. Pugliese, D. Fergusson, B. Winston et al., Canadian Critical Care Trials G (2012) Safety of cell therapy with mesenchymal stromal cells (SafeCell): a systematic review and meta-analysis of clinical trials, PLoS One, vol.7, issue.10

L. Da-silva-meirelles, P. Chagastelles, and N. Nardi, Mesenchymal stem cells reside in virtually all post-natal organs and tissues, Journal of Cell Science, vol.119, issue.11, pp.2204-2213, 2006.
DOI : 10.1242/jcs.02932

M. Najar, G. Raicevic, H. Boufker, F. Kazan, H. et al., Mesenchymal stromal cells use PGE2 to modulate activation and proliferation of lymphocyte subsets: Combined comparison of adipose tissue, Wharton???s Jelly and bone marrow sources, Cellular Immunology, vol.264, issue.2, pp.171-179, 2010.
DOI : 10.1016/j.cellimm.2010.06.006

J. Montesinos, E. Flores-figueroa, S. Castillo-medina, P. Flores-guzman, E. Hernandez-estevez et al., Human mesenchymal stromal cells from adult and neonatal sources: comparative analysis of their morphology, immunophenotype, differentiation patterns and neural protein expression, Cytotherapy, vol.11, issue.2, pp.163-17610, 1080.
DOI : 10.1080/14653240802582075

P. Luz-crawford, M. Torres, D. Noel, A. Fernandez, K. Toupet et al., The immunosuppressive signature of menstrual blood mesenchymal stem cells entails opposite effects on experimental arthritis and graft versus host diseases, STEM CELLS, vol.32, issue.2, 2015.
DOI : 10.1002/stem.2244

N. Kaltz, A. Funari, S. Hippauf, B. Delorme, D. Noel et al., In Vivo Osteoprogenitor Potency of Human Stromal Cells from Different Tissues Does Not Correlate with Expression of POU5F1 or Its Pseudogenes, Stem Cells, vol.24, issue.9, pp.2419-24242008, 2008.
DOI : 10.1634/stemcells.2008-0304

B. Sousa, R. Parreira, E. Fonseca, M. Amaya, F. Tonelli et al., Human adult stem cells from diverse origins: An overview from multiparametric immunophenotyping to clinical applications, Cytometry Part A, vol.183, issue.Suppl 2, pp.43-77, 2014.
DOI : 10.1002/cyto.a.22402

D. Ugarte, D. Morizono, K. Elbarbary, A. Alfonso, Z. Zuk et al., Comparison of Multi-Lineage Cells from Human Adipose Tissue and Bone Marrow, Cells Tissues Organs, vol.174, issue.3, pp.101-10971150, 2003.
DOI : 10.1159/000071150

S. Roche, B. Delorme, R. Oostendorp, R. Barbet, D. Caton et al., Comparative proteomic analysis of human mesenchymal and embryonic stem cells: Towards the definition of a mesenchymal stem cell proteomic signature, PROTEOMICS, vol.281, issue.2, pp.223-232, 2009.
DOI : 10.1002/pmic.200800035

URL : https://hal.archives-ouvertes.fr/hal-00357170

K. Sivasubramaniyan, D. Lehnen, R. Ghazanfari, M. Sobiesiak, A. Harichandan et al., Phenotypic and functional heterogeneity of human bone marrow- and amnion-derived MSC subsets, Annals of the New York Academy of Sciences, vol.15, issue.Suppl. 1, pp.94-106, 2012.
DOI : 10.1111/j.1749-6632.2012.06551.x

F. Djouad, C. Bony, T. Haupl, G. Uze, N. Lahlou et al., Transcriptional profiles discriminate bone marrow-derived and synovium-derived mesenchymal stem cells, Arthritis Research & Therapy, vol.7, issue.6, pp.1304-131510, 2005.
DOI : 10.1186/ar1827

URL : https://hal.archives-ouvertes.fr/inserm-00089978

D. Phinney and L. Sensebe, Mesenchymal stromal cells: misconceptions and evolving concepts, Cytotherapy, vol.15, issue.2, pp.140-145, 2013.
DOI : 10.1016/j.jcyt.2012.11.005

T. Deuse, M. Stubbendorff, K. Tang-quan, N. Phillips, M. Kay et al., Immunogenicity and Immunomodulatory Properties of Umbilical Cord Lining Mesenchymal Stem Cells, Cell Transplantation, vol.20, issue.5, pp.655-66710, 2011.
DOI : 10.3727/096368910X536473

H. Wegmeyer, A. Broske, M. Leddin, K. Kuentzer, A. Nisslbeck et al., Mesenchymal Stromal Cell Characteristics Vary Depending on Their Origin, Stem Cells and Development, vol.22, issue.19, pp.2606-2618, 2013.
DOI : 10.1089/scd.2013.0016

D. Noel, D. Caton, S. Roche, C. Bony, S. Lehmann et al., Cell specific differences between human adipose-derived and mesenchymal???stromal cells despite similar differentiation potentials, Experimental Cell Research, vol.314, issue.7, pp.1575-1584, 2008.
DOI : 10.1016/j.yexcr.2007.12.022

URL : https://hal.archives-ouvertes.fr/hal-00275173

B. Puissant, C. Barreau, P. Bourin, C. Clavel, J. Corre et al., Immunomodulatory effect of human adipose tissue-derived adult stem cells: comparison with bone marrow mesenchymal stem cells, British Journal of Haematology, vol.76, issue.2, pp.118-129, 2005.
DOI : 10.1091/mbc.E02-02-0105

M. Strioga, S. Viswanathan, A. Darinskas, O. Slaby, and J. Michalek, Same or Not the Same? Comparison of Adipose Tissue-Derived Versus Bone Marrow-Derived Mesenchymal Stem and Stromal Cells, Stem Cells and Development, vol.21, issue.14, pp.2724-2752, 2012.
DOI : 10.1089/scd.2011.0722

M. Gupta and A. Ajay, Fat on sale: role of adipose-derived stem cells as anti-fibrosis agent in regenerative medicine, Stem Cell Research & Therapy, vol.5, issue.1, pp.233-243, 2015.
DOI : 10.1186/s13287-015-0246-3

M. Pikula, N. Marek-trzonkowska, A. Wardowska, A. Renkielska, and P. Trzonkowski, Adipose tissue-derived stem cells in clinical applications, Expert Opinion on Biological Therapy, vol.16, issue.10, pp.1357-1370, 2013.
DOI : 10.3727/096368911X566217

K. Lin, Y. Matsubara, Y. Masuda, K. Togashi, T. Ohno et al., Characterization of adipose tissue-derived cells isolated with the Celution??? system, Cytotherapy, vol.10, issue.4, pp.417-42610, 1080.
DOI : 10.1080/14653240801982979

J. Gimble, F. Guilak, and B. Bunnell, Clinical and preclinical translation of cell-based therapies using adipose tissue-derived cells, Stem Cell Research & Therapy, vol.1, issue.2, pp.19-29, 2010.
DOI : 10.1186/scrt19

A. Bailey, S. Kapur, and A. Katz, Characterization of Adipose-Derived Stem Cells: An Update, Current Stem Cell Research & Therapy, vol.5, issue.2, pp.95-102, 2010.
DOI : 10.2174/157488810791268555

W. Ong, C. Tan, K. Chan, G. Goesantoso, X. Chan et al., Identification of Specific Cell-Surface Markers of Adipose-Derived Stem Cells from Subcutaneous and Visceral Fat Depots, Stem Cell Reports, vol.2, issue.2, pp.171-179, 2014.
DOI : 10.1016/j.stemcr.2014.01.002

C. Bony, M. Cren, S. Domergue, K. Toupet, C. Jorgensen et al., Adipose Mesenchymal Stem Cells Isolated after Manual or Water-jet-Assisted Liposuction Display Similar Properties, Frontiers in Immunology, vol.31, issue.3, 2015.
DOI : 10.1007/s00266-006-0186-5

I. Bochev, G. Elmadjian, D. Kyurkchiev, L. Tzvetanov, I. Altankova et al., Mesenchymal stem cells from human bone marrow or adipose tissue differently modulate mitogen-stimulated B-cell immunoglobulin production in vitro, Cell Biology International, vol.32, issue.4, pp.384-393, 2008.
DOI : 10.1016/j.cellbi.2007.12.007

E. Ivanova-todorova, I. Bochev, M. Mourdjeva, R. Dimitrov, D. Bukarev et al., Adipose tissue-derived mesenchymal stem cells are more potent suppressors of dendritic cells differentiation compared to bone marrow-derived mesenchymal stem cells, Immunology Letters, vol.126, issue.1-2, 2009.
DOI : 10.1016/j.imlet.2009.07.010

J. Rehman, D. Traktuev, J. Li, S. Merfeld-clauss, C. Temm-grove et al., Secretion of Angiogenic and Antiapoptotic Factors by Human Adipose Stromal Cells, Circulation, vol.109, issue.10, pp.1292-1298, 2004.
DOI : 10.1161/01.CIR.0000121425.42966.F1

W. Kim, B. Park, H. Kim, J. Park, K. Kim et al., Evidence supporting antioxidant action of adipose-derived stem cells: Protection of human dermal fibroblasts from oxidative stress, Journal of Dermatological Science, vol.49, issue.2, pp.133-142, 2008.
DOI : 10.1016/j.jdermsci.2007.08.004

W. Kim, B. Park, and J. Sung, The wound-healing and antioxidant effects of adipose-derived stem cells, Expert Opinion on Biological Therapy, vol.43, issue.7, pp.879-88710, 2009.
DOI : 10.1111/j.1346-8138.2007.00341.x

C. Chang, Unmet needs in the treatment of autoimmunity: From aspirin to stem cells, Autoimmunity Reviews, vol.13, issue.4-5, pp.331-346, 2014.
DOI : 10.1016/j.autrev.2014.01.052

Y. Nie, C. Lau, A. Lie, G. Chan, and M. Mok, Defective phenotype of mesenchymal stem cells in patients with systemic lupus erythematosus, Lupus, vol.19, issue.7, pp.850-85910, 2010.
DOI : 10.1177/0961203310361482

M. Kastrinaki, P. Sidiropoulos, S. Roche, J. Ringe, S. Lehmann et al., Functional, molecular and proteomic characterisation of bone marrow mesenchymal stem cells in rheumatoid arthritis, Annals of the Rheumatic Diseases, vol.67, issue.6, pp.741-749076174, 2007.
DOI : 10.1136/ard.2007.076174

P. Cipriani, D. Benedetto, P. Ruscitti, P. Campese, A. Liakouli et al., Impaired Endothelium-Mesenchymal Stem Cells Cross-talk in Systemic Sclerosis: a Link Between Vascular and Fibrotic Features, Arthritis Research & Therapy, vol.42, issue.5, pp.442-452, 2014.
DOI : 10.1186/s13075-014-0442-z

P. Cipriani, A. Marrelli, P. Benedetto, V. Liakouli, F. Carubbi et al., Scleroderma Mesenchymal Stem Cells display a different phenotype from healthy controls; implications for regenerative medicine, Angiogenesis, vol.88, issue.9, pp.595-607, 2013.
DOI : 10.1007/s10456-013-9338-9

M. Orciani, S. Svegliati, S. Gorbi, T. Spadoni, R. Lazzarini et al., Alterations of ROS pathways in scleroderma begin at stem cell level, J Biol Regul Homeost Agents, vol.27, issue.1, pp.211-224, 2013.

E. Zappia, S. Casazza, E. Pedemonte, F. Benvenuto, I. Bonanni et al., Mesenchymal stem cells ameliorate experimental autoimmune encephalomyelitis inducing T-cell anergy, Blood, vol.106, issue.5, pp.1755-176110, 2005.
DOI : 10.1182/blood-2005-04-1496

A. Augello, R. Tasso, S. Negrini, R. Cancedda, and G. Pennesi, Cell therapy using allogeneic bone marrow mesenchymal stem cells prevents tissue damage in collagen-induced arthritis, Arthritis & Rheumatism, vol.26, issue.4, pp.1175-1186, 2007.
DOI : 10.1002/art.22511

C. Bouffi, C. Bony, C. Jorgensen, and D. Noel, Skin fibroblasts are potent suppressors of inflammation in experimental arthritis, Annals of the Rheumatic Diseases, vol.70, issue.9, pp.1671-1676, 2011.
DOI : 10.1136/ard.2010.143297

K. Zhou, H. Zhang, J. O. Feng, X. Yao, G. Hou et al., Transplantation of Human Bone Marrow Mesenchymal Stem Cell Ameliorates the Autoimmune Pathogenesis in MRL/lpr Mice, Cellular and Molecular Immunology, vol.5, issue.6, pp.417-424, 2008.
DOI : 10.1038/cmi.2008.52

O. Ringden, M. Uzunel, I. Rasmusson, M. Remberger, B. Sundberg et al., Mesenchymal Stem Cells for Treatment of Therapy-Resistant Graft-versus-Host Disease, Transplantation, vol.81, issue.10, pp.1390-1397, 2006.
DOI : 10.1097/01.tp.0000214462.63943.14

R. Lee, M. Seo, R. Reger, J. Spees, A. Pulin et al., Multipotent stromal cells from human marrow home to and promote repair of pancreatic islets and renal glomeruli in diabetic NOD/scid mice, Proceedings of the National Academy of Sciences, vol.103, issue.46, pp.17438-17443, 2006.
DOI : 10.1073/pnas.0608249103

D. Polchert, J. Sobinsky, G. Douglas, M. Kidd, A. Moadsiri et al., IFN-gamma activation of mesenchymal stem cells for treatment and prevention of graft versus host disease, 2008.

A. Borchers, N. Leibushor, S. Naguwa, G. Cheema, Y. Shoenfeld et al., Lupus nephritis: A critical review, Autoimmunity Reviews, vol.12, issue.2, pp.174-194, 2012.
DOI : 10.1016/j.autrev.2012.08.018

R. Cervera, A. Doria, Z. Amoura, M. Khamashta, M. Schneider et al., Patterns of systemic lupus erythematosus expression in Europe, Autoimmunity Reviews, vol.13, issue.6, pp.621-629, 2014.
DOI : 10.1016/j.autrev.2013.11.007

P. Riboldi, M. Gerosa, C. Luzzana, and L. Catelli, Cardiac Involvement in Systemic Autoimmune Diseases, Clinical Reviews in Allergy & Immunology, vol.23, issue.3, pp.247-261247, 2002.
DOI : 10.1385/CRIAI:23:3:247

S. Sitia, F. Atzeni, P. Sarzi-puttini, D. Bello, V. Tomasoni et al., Cardiovascular involvement in systemic autoimmune diseases, Autoimmunity Reviews, vol.8, issue.4, pp.281-286, 2009.
DOI : 10.1016/j.autrev.2008.08.004

G. Wu, H. Liu, R. Leng, X. Li, X. Li et al., Subclinical atherosclerosis in patients with systemic lupus erythematosus: A systemic review and meta-analysis, Autoimmunity Reviews, vol.15, issue.1, pp.22-37, 2016.
DOI : 10.1016/j.autrev.2015.10.002

J. Qian, Y. Wang, C. Huang, X. Yang, J. Zhao et al., Survival and prognostic factors of systemic lupus erythematosus-associated pulmonary arterial hypertension: A PRISMA-compliant systematic review and meta-analysis, Autoimmunity Reviews, vol.15, issue.3, pp.250-257, 2016.
DOI : 10.1016/j.autrev.2015.11.012

N. Sarbu, F. Alobeidi, P. Toledano, G. Espinosa, I. Giles et al., Brain abnormalities in newly diagnosed neuropsychiatric lupus: Systematic MRI approach and correlation with clinical and laboratory data in a large multicenter cohort, Autoimmunity Reviews, vol.14, issue.2, pp.153-159, 2015.
DOI : 10.1016/j.autrev.2014.11.001

D. Roccatello, S. Sciascia, S. Baldovino, D. Rossi, M. Alpa et al., A 4-year observation in lupus nephritis patients treated with an intensified B-lymphocyte depletion without immunosuppressive maintenance treatment???Clinical response compared to literature and immunological re-assessment, Autoimmunity Reviews, vol.14, issue.12, pp.1123-1130, 2015.
DOI : 10.1016/j.autrev.2015.07.017

H. Jeltsch-david and S. Muller, Neuropsychiatric systemic lupus erythematosus and cognitive dysfunction: The MRL-lpr mouse strain as a model, Autoimmunity Reviews, vol.13, issue.9, pp.963-973, 2014.
DOI : 10.1016/j.autrev.2014.08.015

M. Gatto, M. Zen, A. Ghirardello, S. Bettio, N. Bassi et al., Emerging and critical issues in the pathogenesis of lupus, Autoimmunity Reviews, vol.12, issue.4, pp.523-536, 2013.
DOI : 10.1016/j.autrev.2012.09.003

P. Sawla, A. Hossain, B. Hahn, and R. Singh, Regulatory T cells in systemic lupus erythematosus (SLE); Role of peptide tolerance, Autoimmunity Reviews, vol.11, issue.9, pp.611-614, 2012.
DOI : 10.1016/j.autrev.2011.09.008

I. Ruiz-arruza, C. Barbosa, A. Ugarte, and G. Ruiz-irastorza, Comparison of high versus low???medium prednisone doses for the treatment of systemic lupus erythematosus patients with high activity at diagnosis, Autoimmunity Reviews, vol.14, issue.10, pp.875-879, 2015.
DOI : 10.1016/j.autrev.2015.05.011

J. Hannah, A. Casian, D. Cruz, and D. , Tacrolimus use in lupus nephritis: A systematic review and meta-analysis, Autoimmunity Reviews, vol.15, issue.1, 2016.
DOI : 10.1016/j.autrev.2015.09.006

M. Frieri and H. Stampfl, Systemic lupus erythematosus and atherosclerosis: Review of the literature, Autoimmunity Reviews, vol.15, issue.1, pp.16-21, 2016.
DOI : 10.1016/j.autrev.2015.08.007

Y. Cohen and A. Nagler, Treatment of refractory autoimmune diseases with ablative immunotherapy, Autoimmunity Reviews, vol.3, issue.2, pp.21-2910, 2004.
DOI : 10.1016/S1568-9972(03)00083-1

S. Sciascia, L. Ceberio, C. Garcia-fernandez, D. Roccatello, Y. Karim et al., Systemic lupus erythematosus and infections: Clinical importance of conventional and upcoming biomarkers, Autoimmunity Reviews, vol.12, issue.2, pp.157-163, 2012.
DOI : 10.1016/j.autrev.2012.03.009

R. Davies, S. Sangle, N. Jordan, L. Aslam, M. Lewis et al., Rituximab in the treatment of resistant lupus nephritis: therapy failure in rapidly progressive crescentic lupus nephritis, Lupus, vol.64, issue.6, pp.574-58210, 2013.
DOI : 10.1097/01.ASN.0000108969.21691.5D

A. Kamal and M. Khamashta, The efficacy of novel B cell biologics as the future of SLE treatment: A review, Autoimmunity Reviews, vol.13, issue.11, 2014.
DOI : 10.1016/j.autrev.2014.08.020

J. Guerrero, A. Schwarting, J. Merrill, W. Chatham, W. Stohl et al., A phase III, randomized, placebo-controlled study of belimumab, a monoclonal antibody that inhibits B lymphocyte stimulator, in patients with systemic lupus erythematosus, Arthritis Rheum, vol.63, issue.12, pp.3918-3930, 2011.

J. Liao, C. Chang, H. Wu, and Q. Lu, Cell-based therapies for systemic lupus erythematosus, Autoimmunity Reviews, vol.14, issue.1, pp.43-48, 2015.
DOI : 10.1016/j.autrev.2014.10.001

H. Papadaki, D. Boumpas, F. Gibson, D. Jayne, J. Axford et al., Increased apoptosis of bone marrow CD34+ cells and impaired function of bone marrow stromal cells in patients with systemic lupus erythematosus, British Journal of Haematology, vol.312, issue.1, pp.167-174, 2001.
DOI : 10.1016/S0140-6736(95)91273-8

L. Sun, K. Akiyama, H. Zhang, T. Yamaza, Y. Hou et al., Mesenchymal Stem Cell Transplantation Reverses Multiorgan Dysfunction in Systemic Lupus Erythematosus Mice and Humans, Stem Cells, vol.27, issue.6, pp.1421-1432, 2009.
DOI : 10.1002/stem.68

L. Sun, H. Zhang, X. Feng, Y. Hou, L. Lu et al., Abnormality of bone marrow-derived mesenchymal stem cells in patients with systemic lupus erythematosus, Lupus, vol.18, issue.2, pp.121-128, 2007.
DOI : 10.1046/j.1365-2141.2001.03076.x

Y. Tang, X. Ma, H. Zhang, Z. Gu, Y. Hou et al., Gene Expression Profile Reveals Abnormalities of Multiple Signaling Pathways in Mesenchymal Stem Cell Derived from Patients with Systemic Lupus Erythematosus, Clinical and Developmental Immunology, vol.5, issue.4, 2012.
DOI : 10.1677/JOE-07-0532

Z. Gu, X. Cao, J. Jiang, L. Li, Z. Da et al., Upregulation of p16INK4A promotes cellular senescence of bone marrow-derived mesenchymal stem cells from systemic lupus erythematosus patients, Cellular Signalling, vol.24, issue.12, pp.2307-2314, 2012.
DOI : 10.1016/j.cellsig.2012.07.012

Z. Gu, W. Tan, G. Feng, Y. Meng, B. Shen et al., Wnt/??-catenin signaling mediates the senescence of bone marrow-mesenchymal stem cells from systemic lupus erythematosus patients through the p53/p21 pathway, Molecular and Cellular Biochemistry, vol.317, issue.1-2, pp.27-37, 2014.
DOI : 10.1007/s11010-013-1866-5

X. Li, L. Liu, D. Meng, D. Wang, J. Zhang et al., Enhanced Apoptosis and Senescence of Bone-Marrow-Derived Mesenchymal Stem Cells in Patients with Systemic Lupus Erythematosus, Stem Cells and Development, vol.21, issue.13, pp.2387-2394, 2012.
DOI : 10.1089/scd.2011.0447

F. Gu, I. Molano, P. Ruiz, L. Sun, and G. Gilkeson, Differential effect of allogeneic versus syngeneic mesenchymal stem cell transplantation in MRL/lpr and (NZB/NZW)F1 mice, Clinical Immunology, vol.145, issue.2, pp.142-152, 2012.
DOI : 10.1016/j.clim.2012.08.012

E. Collins, F. Gu, M. Qi, I. Molano, P. Ruiz et al., Differential Efficacy of Human Mesenchymal Stem Cells Based on Source of Origin, The Journal of Immunology, vol.193, issue.9, pp.4381-4390, 2014.
DOI : 10.4049/jimmunol.1401636

K. Zhou, H. Zhang, J. O. Feng, X. Yao, G. Hou et al., Transplantation of Human Bone Marrow Mesenchymal Stem Cell Ameliorates the Autoimmune Pathogenesis in MRL/lpr Mice, Cellular and Molecular Immunology, vol.5, issue.6, pp.417-424, 2008.
DOI : 10.1038/cmi.2008.52

X. Ma, C. N. Gu, Z. Huang, J. Wang, D. Liang et al., Allogenic Mesenchymal Stem Cell Transplantation Ameliorates Nephritis in Lupus Mice Via Inhibition of B-Cell Activation, Cell Transplantation, vol.22, issue.12, pp.2279-229010, 2013.
DOI : 10.3727/096368912X658692

M. Youd, C. Blickarz, L. Woodworth, T. Touzjian, A. Edling et al., Allogeneic mesenchymal stem cells do not protect NZBxNZW F1 mice from developing lupus disease, Clin Exp Immunol, vol.161, issue.1, pp.176-186, 2010.

F. Schena, C. Gambini, A. Gregorio, M. Mosconi, D. Reverberi et al., Interferon-??-dependent inhibition of B cell activation by bone marrow-derived mesenchymal stem cells in a murine model of systemic lupus erythematosus, Arthritis & Rheumatism, vol.289, issue.9, pp.2776-2786, 2010.
DOI : 10.1002/art.27560

M. Park, S. Kwok, S. Lee, E. Kim, S. Park et al., Adipose tissuederived mesenchymal stem cells induce expansion of interleukin-10-producing regulatory B cells and ameliorate autoimmunity in a murine model of systemic lupus erythematosus, Cell Transplant, vol.243727, issue.11, pp.2367-237710, 2015.

E. Choi, I. Shin, J. Song, T. Yun, Y. J. Choi et al., Transplantation of Adipose Tissue-Derived Mesenchymal Stem Cells Prevents the Development of Lupus Dermatitis, Stem Cells and Development, vol.24, issue.17, pp.2041-2051, 2015.
DOI : 10.1089/scd.2015.0021

E. Choi, H. Lee, S. Shin, J. Park, T. Yun et al., Comparative Efficacies of Long-Term Serial Transplantation of Syngeneic, Allogeneic, Xenogeneic, or CTLA4Ig-Overproducing Xenogeneic Adipose Tissue-Derived Mesenchymal Stem Cells on Murine Systemic Lupus Erythematosus, Cell Transplantation, vol.25, issue.6, p.10, 2015.
DOI : 10.3727/096368915X689442

E. Choi, I. Shin, S. Park, J. Park, J. Kim et al., Reversal of serologic, immunologic, and histologic dysfunction in mice with systemic lupus erythematosus by long-term serial adipose tissue-derived mesenchymal stem cell transplantation, Arthritis & Rheumatism, vol.199, issue.1, pp.243-253, 2012.
DOI : 10.1002/art.33313

Z. Gu, K. Akiyama, X. Ma, H. Zhang, X. Feng et al., Transplantation of umbilical cord mesenchymal stem cells alleviates lupus nephritis in MRL/lpr mice, Lupus, vol.26, issue.13, pp.1502-1514100961203310373782, 1177.
DOI : 10.1152/ajpcell.00401.2005

J. Chang, S. Hung, H. Wu, W. Wu, A. Yang et al., Therapeutic Effects of Umbilical Cord Blood-Derived Mesenchymal Stem Cell Transplantation in Experimental Lupus Nephritis, Cell Transplantation, vol.20, issue.2, pp.245-25710, 2011.
DOI : 10.3727/096368910X520056

F. Carrion, N. E. Ruiz, C. Diaz, F. Inostroza, C. Rojo et al., Autologous mesenchymal stem cell treatment increased T regulatory cells with no effect on disease activity in two systemic lupus erythematosus patients, Lupus, vol.29, issue.3, pp.317-322, 2010.
DOI : 10.1182/blood-2005-04-1496

J. Liang, H. Zhang, B. Hua, H. Wang, L. Lu et al., Allogenic mesenchymal stem cells transplantation in refractory systemic lupus erythematosus: a pilot clinical study, Annals of the Rheumatic Diseases, vol.69, issue.8, pp.1423-1429123463, 2009.
DOI : 10.1136/ard.2009.123463

L. Sun, D. Wang, J. Liang, H. Zhang, X. Feng et al., Umbilical cord mesenchymal stem cell transplantation in severe and refractory systemic lupus erythematosus, Arthritis & Rheumatism, vol.37, issue.Suppl 1, pp.2467-2475, 2010.
DOI : 10.1002/art.27548

X. Li, D. Wang, J. Liang, H. Zhang, and L. Sun, Mesenchymal SCT ameliorates refractory cytopenia in patients with systemic lupus erythematosus, Bone Marrow Transplantation, vol.48, issue.4, pp.544-550, 2013.
DOI : 10.1002/art.24499

J. Liang, F. Gu, H. Wang, B. Hua, Y. Hou et al., Mesenchymal stem cell transplantation for diffuse alveolar hemorrhage in SLE, Nature Reviews Rheumatology, vol.8, issue.8, pp.486-489, 2010.
DOI : 10.1038/nrrheum.2010.80

D. Shi, D. Wang, X. Li, H. Zhang, C. N. Lu et al., Allogeneic transplantation of umbilical cord-derived mesenchymal stem cells for diffuse alveolar hemorrhage in systemic lupus erythematosus, Clinical Rheumatology, vol.137, issue.5, pp.841-84610, 2012.
DOI : 10.1007/s10067-012-1943-2

D. Wang, H. Zhang, J. Liang, X. Li, X. Feng et al., Allogeneic mesenchymal stem cell transplantation in severe and refractory systemic lupus erythematosus: 4 years experience, Cell Transplantation, vol.223727, issue.12, pp.2267-227710, 2013.
DOI : 10.3727/096368912X658719

F. Gu, D. Wang, H. Zhang, X. Feng, G. Gilkeson et al., Allogeneic mesenchymal stem cell transplantation for lupus nephritis patients refractory to conventional therapy, Clinical Rheumatology, vol.27, issue.12, pp.1611-161910, 2014.
DOI : 10.1007/s10067-014-2754-4

D. Wang, J. Li, Y. Zhang, M. Zhang, J. Chen et al., Umbilical cord mesenchymal stem cell transplantation in active and refractory systemic lupus erythematosus: a multicenter clinical study, Arthritis Research & Therapy, vol.16, issue.2, pp.79-89, 2014.
DOI : 10.1007/s00441-011-1249-8

V. Modena, G. Bianchi, and D. Roccatello, Cost-effectiveness of biologic treatment for rheumatoid arthritis in clinical practice: An achievable target?, Autoimmunity Reviews, vol.12, issue.8, pp.835-838, 2013.
DOI : 10.1016/j.autrev.2012.11.009

O. Epis, L. Giacomelli, S. Deidda, and E. Bruschi, Tight control applied to the biological therapy of rheumatoid arthritis, Autoimmunity Reviews, vol.12, issue.8, 2013.
DOI : 10.1016/j.autrev.2012.11.010

T. Dimitroulas, S. Nikas, P. Trontzas, and G. Kitas, Biologic therapies and systemic bone loss in rheumatoid arthritis, Autoimmunity Reviews, vol.12, issue.10, pp.958-966, 2013.
DOI : 10.1016/j.autrev.2013.03.015

M. Cutolo and S. Nadler, Advances in CTLA-4-Ig-mediated modulation of inflammatory cell and immune response activation in rheumatoid arthritis, Autoimmunity Reviews, vol.12, issue.7, pp.758-767, 2013.
DOI : 10.1016/j.autrev.2013.01.001

B. Chen, J. Hu, L. Liao, Z. Sun, Q. Han et al., mesenchymal stem cells aggravate collagen-induced arthritis by up-regulating interleukin-6, Clinical & Experimental Immunology, vol.113, issue.3, pp.292-302, 2010.
DOI : 10.1111/j.1365-2249.2009.04069.x

F. Djouad, V. Fritz, F. Apparailly, P. Louis-plence, C. Bony et al., Reversal of the immunosuppressive properties of mesenchymal stem cells by tumor necrosis factor ?? in collagen-induced arthritis, Arthritis & Rheumatism, vol.101, issue.5, pp.1595-1603, 2005.
DOI : 10.1002/art.21012

Y. Liu, R. Mu, S. Wang, L. Long, X. Liu et al., Therapeutic potential of human umbilical cord mesenchymal stem cells in the treatment of rheumatoid arthritis, Arthritis Research & Therapy, vol.12, issue.6, pp.210-220, 2010.
DOI : 10.1186/ar3187

M. Gonzalez, E. Gonzalez-rey, L. Rico, D. Buscher, and M. Delgado, Treatment of experimental arthritis by inducing immune tolerance with human adipose-derived mesenchymal stem cells, Arthritis & Rheumatism, vol.14, issue.4, pp.1006-1019, 2009.
DOI : 10.1002/art.24405

L. Wang, L. Wang, X. Cong, G. Liu, J. Zhou et al., Human Umbilical Cord Mesenchymal Stem Cell Therapy for Patients with Active Rheumatoid Arthritis: Safety and Efficacy, Stem Cells and Development, vol.22, issue.24, pp.3192-3202, 2013.
DOI : 10.1089/scd.2013.0023

L. Wang, Y. Zhang, H. Li, J. Hong, X. Chen et al., Clinical Observation of Employment of Umbilical Cord Derived Mesenchymal Stem Cell for Juvenile Idiopathic Arthritis Therapy, Stem Cells International, vol.91, issue.8, pp.9165267-9165277, 2016.
DOI : 10.1002/art.27548

S. Khalili, D. Faustman, Y. Liu, Y. Sumita, D. Blank et al., Treatment for salivary gland hypofunction at both initial and advanced stages of Sj??gren-like disease: a comparative study of bone marrow therapy versus spleen cell therapy with a 1-year monitoring period, Cytotherapy, vol.16, issue.3, pp.412-423, 2014.
DOI : 10.1016/j.jcyt.2013.10.006

J. Xu, D. Wang, D. Liu, Z. Fan, H. Zhang et al., Allogeneic mesenchymal stem cell treatment alleviates experimental and clinical Sjogren syndrome, Blood, vol.120, issue.15, pp.3142-315110, 2012.
DOI : 10.1182/blood-2011-11-391144

J. Voswinkel, S. Francois, J. Simon, M. Benderitter, N. Gorin et al., Use of Mesenchymal Stem Cells (MSC) in Chronic Inflammatory Fistulizing and Fibrotic Diseases: a Comprehensive Review, Clinical Reviews in Allergy & Immunology, vol.18, issue.3, pp.180-19210, 2013.
DOI : 10.1007/s12016-012-8347-6

Y. Cho, W. Lee, K. Park, M. Kim, H. Yoo et al., Autologous Adipose Tissue-Derived Stem Cells for the Treatment of Crohn's Fistula: A Phase I Clinical Study, Cell Transplantation, vol.22, issue.2, pp.279-28510, 2013.
DOI : 10.3727/096368912X656045

M. Duijvestein, A. Vos, H. Roelofs, M. Wildenberg, B. Wendrich et al., Autologous bone marrow-derived mesenchymal stromal cell treatment for refractory luminal Crohn's disease: results of a phase I study, Gut, vol.59, issue.12, pp.1662-1669, 2010.
DOI : 10.1136/gut.2010.215152

F. De-la-portilla, F. Alba, D. Garcia-olmo, J. Herrerias, F. Gonzalez et al., Expanded allogeneic adipose-derived stem cells (eASCs) for the treatment of complex perianal fistula in Crohn???s disease: results from a multicenter phase I/IIa clinical trial, International Journal of Colorectal Disease, vol.69, issue.3, pp.313-32310, 2013.
DOI : 10.1007/s00384-012-1581-9

M. Gregorini, R. Maccario, M. Avanzini, V. Corradetti, A. Moretta et al., Antineutrophil Cytoplasmic Antibody-Associated Renal Vasculitis Treated With Autologous Mesenchymal Stromal Cells: Evaluation of the Contribution of Immune-Mediated Mechanisms, Mayo Clinic Proceedings, vol.88, issue.10, pp.1174-1179, 2013.
DOI : 10.1016/j.mayocp.2013.06.021

F. Davatchi, B. Nikbin, H. Shams, S. Abdollahi, B. Mohyeddin et al., Mesenchymal stem cell therapy unable to rescue the vision from advanced Behcet's disease retinal vasculitis: report of three patients, International Journal of Rheumatic Diseases, vol.106, issue.9817, pp.139-14710, 2013.
DOI : 10.1111/1756-185X.12068

S. Lablanche, S. Borot, A. Wojtusciszyn, F. Bayle, R. Tetaz et al., Five-Year Metabolic, Functional, and Safety Results of Patients With Type 1 Diabetes Transplanted With Allogenic Islets Within the Swiss-French GRAGIL Network, Diabetes Care, vol.38, issue.9, pp.1714-172210, 2015.
DOI : 10.2337/dc15-0094

I. Boumaza, S. Srinivasan, W. Witt, C. Feghali-bostwick, Y. Dai et al., Autologous bone marrow-derived rat mesenchymal stem cells promote PDX-1 and insulin expression in the islets, alter T cell cytokine pattern and preserve regulatory T cells in the periphery and induce sustained normoglycemia, Journal of Autoimmunity, vol.32, issue.1, pp.33-42, 2009.
DOI : 10.1016/j.jaut.2008.10.004

Y. Xin, X. Jiang, Y. Wang, X. Su, M. Sun et al., Insulin-Producing Cells Differentiated from Human Bone Marrow Mesenchymal Stem Cells In Vitro Ameliorate Streptozotocin-Induced Diabetic Hyperglycemia, PLOS ONE, vol.23, issue.10, 2016.
DOI : 10.1371/journal.pone.0145838.g006

E. Gerdoni, B. Gallo, S. Casazza, S. Musio, I. Bonanni et al., Mesenchymal stem cells effectively modulate pathogenic immune response in experimental autoimmune encephalomyelitis, Annals of Neurology, vol.38, issue.3, pp.219-227, 2007.
DOI : 10.1002/ana.21076

I. Kassis, N. Grigoriadis, B. Gowda-kurkalli, R. Mizrachi-kol, T. Ben-hur et al., Neuroprotection and Immunomodulation With Mesenchymal Stem Cells in Chronic Experimental Autoimmune Encephalomyelitis, Archives of Neurology, vol.65, issue.6, pp.753-761, 2008.
DOI : 10.1001/archneur.65.6.753

D. Wang, S. Li, J. Fu, L. Bai, and L. Guo, Resveratrol augments therapeutic efficiency of mouse bone marrow mesenchymal stem cell-based therapy in experimental autoimmune encephalomyelitis, International Journal of Developmental Neuroscience, vol.49, pp.60-66, 2016.
DOI : 10.1016/j.ijdevneu.2016.01.005

P. Connick, M. Kolappan, C. Crawley, D. Webber, R. Patani et al., Autologous mesenchymal stem cells for the treatment of secondary progressive multiple sclerosis: an open-label phase 2a proof-of-concept study, The Lancet Neurology, vol.11, issue.2, pp.150-15610, 2012.
DOI : 10.1016/S1474-4422(11)70305-2

F. Lublin, J. Bowen, J. Huddlestone, M. Kremenchutzky, A. Carpenter et al., Human placenta-derived cells (PDA-001) for the treatment of adults with multiple sclerosis: A randomized, placebo-controlled, multiple-dose study, Multiple Sclerosis and Related Disorders, vol.3, issue.6, pp.696-704, 2014.
DOI : 10.1016/j.msard.2014.08.002

S. Llufriu, M. Sepulveda, Y. Blanco, P. Marin, B. Moreno et al., Randomized placebocontrolled phase II trial of autologous mesenchymal stem cells in multiple sclerosis, PLoS One, vol.9, issue.12, 2014.

J. Li, D. Zhang, T. Geng, L. Chen, H. Huang et al., The Potential of Human Umbilical Cord-Derived Mesenchymal Stem Cells as a Novel Cellular Therapy for Multiple Sclerosis, Cell Transplantation, vol.23, issue.1, pp.113-12210, 2014.
DOI : 10.3727/096368914X685005

L. Blanc, K. Frassoni, F. Ball, L. Locatelli, F. Roelofs et al., Mesenchymal stem cells for treatment of steroid-resistant, severe, acute graftversus-host disease: a phase II study, Developmental Committee of the European Group for B, Marrow T Lancet, vol.371, issue.962408, pp.1579-158610, 2008.

R. Yanez, M. Lamana, J. Garcia-castro, I. Colmenero, M. Ramirez et al., Adipose Tissue-Derived Mesenchymal Stem Cells Have In Vivo Immunosuppressive Properties Applicable for the Control of the Graft-Versus-Host Disease, Stem Cells, vol.101, issue.11, pp.2582-259110, 2006.
DOI : 10.1634/stemcells.2006-0228

B. Fang, Y. Song, L. Liao, Y. Zhang, and R. Zhao, Favorable Response to Human Adipose Tissue-Derived Mesenchymal Stem Cells in Steroid-Refractory Acute Graft-Versus-Host Disease, Transplantation Proceedings, vol.39, issue.10, pp.3358-3362, 2007.
DOI : 10.1016/j.transproceed.2007.08.103

Q. Kong, B. Sun, G. Wang, D. Zhai, L. Mu et al., BM stromal cells ameliorate experimental autoimmune myasthenia gravis by altering the balance of Th cells through the secretion of IDO, European Journal of Immunology, vol.74, issue.3, pp.800-809, 2009.
DOI : 10.1002/eji.200838729

Q. Kong, B. Sun, S. Bai, D. Zhai, G. Wang et al., Administration of bone marrow stromal cells ameliorates experimental autoimmune myasthenia gravis by altering the balance of Th1/Th2/Th17/Treg cell subsets through the secretion of TGFbeta, J Neuroimmunol, vol.20712, issue.12, pp.83-91005, 2008.

J. Yu, C. Zheng, X. Ren, J. Li, M. Liu et al., Intravenous Administration of Bone Marrow Mesenchymal Stem Cells Benefits Experimental Autoimmune Myasthenia Gravis Mice Through an Immunomodulatory Action, Scandinavian Journal of Immunology, vol.43, issue.3, pp.242-249, 2010.
DOI : 10.1111/j.1365-3083.2010.02445.x

K. Tiev and J. Cabane, Digestive tract involvement in systemic sclerosis, Autoimmunity Reviews, vol.11, issue.1, pp.68-73, 2011.
DOI : 10.1016/j.autrev.2010.06.002

C. Chizzolini, N. Brembilla, E. Montanari, and M. Truchetet, Fibrosis and immune dysregulation in systemic sclerosis, Autoimmunity Reviews, vol.10, issue.5, pp.276-281, 2011.
DOI : 10.1016/j.autrev.2010.09.016

I. Almeida, R. Faria, P. Vita, and C. Vasconcelos, Systemic sclerosis refractory disease: From the skin to the heart, Autoimmunity Reviews, vol.10, issue.11, pp.693-701, 2011.
DOI : 10.1016/j.autrev.2011.04.025

F. Galluccio and M. Matucci-cerinic, Two faces of the same coin: Raynaud phenomenon and digital ulcers in systemic sclerosis, Autoimmunity Reviews, vol.10, issue.5, 2011.
DOI : 10.1016/j.autrev.2010.09.008

S. Guiducci, R. Giacomelli, and M. Cerinic, Vascular complications of scleroderma, Autoimmunity Reviews, vol.6, issue.8, pp.520-523, 2007.
DOI : 10.1016/j.autrev.2006.12.006

S. Lambova and U. Muller-ladner, Pulmonary arterial hypertension in systemic sclerosis, Autoimmunity Reviews, vol.9, issue.11, pp.761-770, 2010.
DOI : 10.1016/j.autrev.2010.06.006

I. Chora, S. Guiducci, M. Manetti, E. Romano, C. Mazzotta et al., Vascular biomarkers and correlation with peripheral vasculopathy in systemic sclerosis, Autoimmunity Reviews, vol.14, issue.4, pp.314-322, 2015.
DOI : 10.1016/j.autrev.2014.12.001

U. Nussinovitch and Y. Shoenfeld, Atherosclerosis and macrovascular involvement in systemic sclerosis: Myth or reality, Autoimmunity Reviews, vol.10, issue.5, pp.259-266, 2011.
DOI : 10.1016/j.autrev.2010.09.014

J. Baraut, L. Michel, F. Verrecchia, and D. Farge, Relationship between cytokine profiles and clinical outcomes in patients with systemic sclerosis, Autoimmunity Reviews, vol.10, issue.2, pp.65-73, 2010.
DOI : 10.1016/j.autrev.2010.08.003

S. Bosello, D. Luca, G. Tolusso, B. Lama, G. Angelucci et al., B cells in systemic sclerosis: A possible target for therapy, Autoimmunity Reviews, vol.10, issue.10, 2011.
DOI : 10.1016/j.autrev.2011.04.013

S. Mehra, J. Walker, K. Patterson, and M. Fritzler, Autoantibodies in systemic sclerosis, Autoimmunity Reviews, vol.12, issue.3, pp.340-354, 2013.
DOI : 10.1016/j.autrev.2012.05.011

T. Czompoly, D. Simon, L. Czirjak, and P. Nemeth, Anti-topoisomerase I autoantibodies in systemic sclerosis, Autoimmunity Reviews, vol.8, issue.8, pp.692-696, 2009.
DOI : 10.1016/j.autrev.2009.02.018

F. Perosa, M. Prete, D. Lernia, G. Ostuni, C. Favoino et al., Anticentromere protein A antibodies in systemic sclerosis: Significance and origin, Autoimmun Rev, vol.15, issue.1, 2016.

E. Hachulla and D. Launay, Diagnosis and Classification of Systemic Sclerosis, Clinical Reviews in Allergy & Immunology, vol.43, issue.2, pp.78-83, 2011.
DOI : 10.1007/s12016-010-8198-y

C. Almeida, I. Almeida, and C. Vasconcelos, Quality of life in systemic sclerosis, Autoimmunity Reviews, vol.14, issue.12, pp.1087-1096, 2015.
DOI : 10.1016/j.autrev.2015.07.012

C. Bruni, J. Raja, C. Denton, and M. Matucci-cerinic, The clinical relevance of sexual dysfunction in systemic sclerosis, Autoimmunity Reviews, vol.14, issue.12, 2015.
DOI : 10.1016/j.autrev.2015.07.016

A. Wells, V. Steen, and G. Valentini, Pulmonary complications: one of the most challenging complications of systemic sclerosis, Rheumatology, vol.48, issue.Supplement 3, p.48, 2009.
DOI : 10.1093/rheumatology/kep109

G. Bussone and L. Mouthon, Interstitial lung disease in systemic sclerosis, Autoimmunity Reviews, vol.10, issue.5, pp.248-255, 2011.
DOI : 10.1016/j.autrev.2010.09.012

L. Mouthon, A. Berezne, G. Bussone, L. Noel, P. Villiger et al., Scleroderma Renal Crisis: A Rare but Severe Complication of Systemic Sclerosis, Clinical Reviews in Allergy & Immunology, vol.66, issue.7, pp.84-91, 2011.
DOI : 10.1007/s12016-009-8191-5

P. Carreira, Pulmonary hypertension in autoimmune rheumatic diseases, Autoimmunity Reviews, vol.3, issue.4, pp.313-320, 2004.
DOI : 10.1016/j.autrev.2003.11.004

G. Murdaca, M. Contatore, R. Gulli, P. Mandich, and F. Puppo, Genetic factors and systemic sclerosis, Autoimmunity Reviews, vol.15, issue.5, 2016.
DOI : 10.1016/j.autrev.2016.01.016

P. Dieude, C. Boileau, and Y. Allanore, Immunogenetics of systemic sclerosis, Autoimmunity Reviews, vol.10, issue.5, pp.282-290, 2011.
DOI : 10.1016/j.autrev.2010.09.017

I. Marie, J. Gehanno, M. Bubenheim, A. Duval-modeste, P. Joly et al., Prospective study to evaluate the association between systemic sclerosis and occupational exposure and review of the literature, Autoimmunity Reviews, vol.13, issue.2, pp.151-156, 2014.
DOI : 10.1016/j.autrev.2013.10.002

L. Bossini-castillo, E. Lopez-isac, and J. Martin, Immunogenetics of systemic sclerosis: Defining heritability, functional variants and shared-autoimmunity pathways, Journal of Autoimmunity, vol.64, 2015.
DOI : 10.1016/j.jaut.2015.07.005

J. Wei, S. Bhattacharyya, W. Tourtellotte, and J. Varga, Fibrosis in systemic sclerosis: Emerging concepts and implications for targeted therapy, Autoimmunity Reviews, vol.10, issue.5, 2011.
DOI : 10.1016/j.autrev.2010.09.015

P. Cipriani, A. Marrelli, V. Liakouli, D. Benedetto, P. Giacomelli et al., Cellular players in angiogenesis during the course of systemic sclerosis, Autoimmunity Reviews, vol.10, issue.10, pp.641-646, 2011.
DOI : 10.1016/j.autrev.2011.04.016

V. Liakouli, P. Cipriani, A. Marrelli, S. Alvaro, P. Ruscitti et al., Angiogenic cytokines and growth factors in systemic sclerosis, Autoimmunity Reviews, vol.10, issue.10, pp.590-594, 2011.
DOI : 10.1016/j.autrev.2011.04.019

A. Servettaz, P. Guilpain, C. Goulvestre, C. Chereau, C. Hercend et al., Radical oxygen species production induced by advanced oxidation protein products predicts clinical evolution and response to treatment in systemic sclerosis, Annals of the Rheumatic Diseases, vol.66, issue.9, pp.1202-1209067504, 2006.
DOI : 10.1136/ard.2006.067504

D. Launay, L. Savale, A. Berezne, L. Pavec, J. Hachulla et al., Working Group on Heart/Lung transplantation in systemic sclerosis of the French Network on Pulmonary H (2014) Lung and heart-lung transplantation for systemic sclerosis patients. A monocentric experience of 13 patients, review of the literature and position paper of a multidisciplinary Working Group, Presse Med, vol.4310, issue.2, pp.345-363

V. Sobanski, D. Launay, E. Hachulla, and M. Humbert, Current Approaches to the Treatment of Systemic-Sclerosis-Associated Pulmonary Arterial Hypertension (SSc-PAH), Current Rheumatology Reports, vol.171, issue.Suppl 1, pp.10-10, 2016.
DOI : 10.1007/s11926-015-0560-x

M. Elhai, C. Meune, J. Avouac, A. Kahan, and Y. Allanore, Trends in mortality in patients with systemic sclerosis over 40 years: a systematic review and meta-analysis of cohort studies, Rheumatology, vol.51, issue.6, pp.1017-1026, 2012.
DOI : 10.1093/rheumatology/ker269

M. Vonk, Z. Marjanovic, F. Van-den-hoogen, S. Zohar, A. Schattenberg et al., Long-term follow-up results after autologous haematopoietic stem cell transplantation for severe systemic sclerosis, Annals of the Rheumatic Diseases, vol.67, issue.1, pp.98-104071464, 2007.
DOI : 10.1136/ard.2007.071464

J. Van-laar, D. Farge, J. Sont, K. Naraghi, Z. Marjanovic et al., Autologous Hematopoietic Stem Cell Transplantation vs Intravenous Pulse Cyclophosphamide in Diffuse Cutaneous Systemic Sclerosis, JAMA, vol.311, issue.24, pp.2490-2498, 2014.
DOI : 10.1001/jama.2014.6368

D. Farge, J. Marolleau, S. Zohar, Z. Marjanovic, J. Cabane et al., Autologous bone marrow transplantation in the treatment of refractory systemic sclerosis: early results from a French multicentre phase I-II study, British Journal of Haematology, vol.93, issue.3, pp.726-739, 2002.
DOI : 10.1002/sim.920

J. Van-laar, K. Naraghi, and A. Tyndall, Haematopoietic stem cell transplantation for poor-prognosis systemic sclerosis, Rheumatology, vol.54, issue.12, pp.2126-2133, 2015.
DOI : 10.1093/rheumatology/kev117

A. Tyndall, Successes and failures of stem cell transplantation in autoimmune diseases. Hematology / the Education Program of the, Hematology Education Program, vol.2011, pp.280-284, 2011.

A. Tyndall, Mesenchymal stem cell treatments in rheumatology???a glass half full?, Nature Reviews Rheumatology, vol.27, issue.2, pp.117-124, 2014.
DOI : 10.1007/s10067-011-1816-0

J. Larghero, D. Farge, A. Braccini, S. Lecourt, A. Scherberich et al., Phenotypical and functional characteristics of in vitro expanded bone marrow mesenchymal stem cells from patients with systemic sclerosis, Annals of the Rheumatic Diseases, vol.67, issue.4, pp.443-449071233, 2007.
DOI : 10.1136/ard.2007.071233

C. Bocelli-tyndall, L. Bracci, G. Spagnoli, A. Braccini, M. Bouchenaki et al., Bone marrow mesenchymal stromal cells (BM-MSCs) from healthy donors and auto-immune disease patients reduce the proliferation of autologous- and allogeneic-stimulated lymphocytes in vitro, Rheumatology, vol.46, issue.3, pp.403-408, 2007.
DOI : 10.1093/rheumatology/kel267

URL : https://hal.archives-ouvertes.fr/inserm-00479753

D. Papa, N. Quirici, N. Soligo, D. Scavullo, C. Cortiana et al., Bone marrow endothelial progenitors are defective in systemic sclerosis, Arthritis & Rheumatism, vol.28, issue.8, pp.2605-2615, 2006.
DOI : 10.1002/art.22035

P. Cipriani, S. Guiducci, I. Miniati, M. Cinelli, S. Urbani et al., Impairment of endothelial cell differentiation from bone marrow???derived mesenchymal stem cells: New insight into the pathogenesis of systemic sclerosis, Arthritis & Rheumatism, vol.40, issue.6, pp.1994-2004, 2007.
DOI : 10.1002/art.22698

P. Cipriani, D. Benedetto, P. Liakouli, V. , D. Papa et al., ) with a functional phenotype: implications for cellular-based therapy, Clinical & Experimental Immunology, vol.47, issue.Suppl. 2, pp.195-206, 2013.
DOI : 10.1111/cei.12111

S. Guiducci, M. Manetti, E. Romano, B. Mazzanti, C. Ceccarelli et al., Bone marrow-derived mesenchymal stem cells from early diffuse systemic sclerosis exhibit a paracrine machinery and stimulate angiogenesis in vitro, Annals of the Rheumatic Diseases, vol.70, issue.11, pp.2011-2021, 2011.
DOI : 10.1136/ard.2011.150607

V. Vanneaux, D. Farge-bancel, S. Lecourt, J. Baraut, A. Cras et al., Expression of transforming growth factor beta receptor II in mesenchymal stem cells from systemic sclerosis patients, BMJ Open, vol.3, issue.1, pp.10-1136, 2013.

N. Scuderi, S. Ceccarelli, M. Onesti, P. Fioramonti, C. Guidi et al., Human Adipose-Derived Stromal Cells for Cell-Based Therapies in the Treatment of Systemic Sclerosis, Cell Transplantation, vol.22, issue.5, pp.779-79510, 2013.
DOI : 10.3727/096368912X639017

F. Batteux, N. Kavian, and A. Servettaz, New insights on chemically induced animal models of systemic sclerosis, Current Opinion in Rheumatology, vol.23, issue.6, pp.511-518, 2011.
DOI : 10.1097/BOR.0b013e32834b1606

A. Inamdar and A. Inamdar, Mesenchymal stem cell therapy in lung disorders: Pathogenesis of lung diseases and mechanism of action of mesenchymal stem cell, Experimental Lung Research, vol.91, issue.8, pp.315-327, 2013.
DOI : 10.1002/art.30071

M. Rojas, J. Xu, C. Woods, A. Mora, W. Spears et al., Bone Marrow???Derived Mesenchymal Stem Cells in Repair of the Injured Lung, American Journal of Respiratory Cell and Molecular Biology, vol.33, issue.2, pp.145-1522004, 2005.
DOI : 10.1165/rcmb.2004-0330OC

M. Kumamoto, T. Nishiwaki, N. Matsuo, H. Kimura, and K. Matsushima, Minimally cultured bone marrow mesenchymal stem cells ameliorate fibrotic lung injury, European Respiratory Journal, vol.34, issue.3, pp.740-748, 2009.
DOI : 10.1183/09031936.00128508

A. Cargnoni, L. Gibelli, A. Tosini, P. Signoroni, C. Nassuato et al., Transplantation of Allogeneic and Xenogeneic Placenta-Derived Cells Reduces Bleomycin-Induced Lung Fibrosis, Cell Transplantation, vol.18, issue.4, pp.405-42210096368909788809857, 2009.
DOI : 10.3727/096368909788809857

L. Ortiz, M. Dutreil, C. Fattman, A. Pandey, G. Torres et al., Interleukin 1 receptor antagonist mediates the antiinflammatory and antifibrotic effect of mesenchymal stem cells during lung injury, Proceedings of the National Academy of Sciences, vol.104, issue.26, pp.11002-11007, 2007.
DOI : 10.1073/pnas.0704421104

R. Lee, A. Pulin, M. Seo, D. Kota, J. Ylostalo et al., Intravenous hMSCs Improve Myocardial Infarction in Mice because Cells Embolized in Lung Are Activated to Secrete the Anti-inflammatory Protein TSG-6, Cell Stem Cell, vol.5, issue.1, pp.54-63, 2009.
DOI : 10.1016/j.stem.2009.05.003

S. Lee, A. Jang, Y. Kim, J. Cha, T. Kim et al., Modulation of cytokine and nitric oxide by mesenchymal stem cell transfer in lung injury/fibrosis, Respiratory Research, vol.276, issue.Pt 1, pp.16-26, 2010.
DOI : 10.1126/science.276.5309.71

M. Conese, A. Carbone, S. Castellani, D. Gioia, and S. , Paracrine Effects and Heterogeneity of Marrow-Derived Stem/Progenitor Cells: Relevance for the Treatment of Respiratory Diseases, Cells Tissues Organs, vol.197, issue.6, pp.445-473, 2013.
DOI : 10.1159/000348831

A. Servettaz, C. Goulvestre, N. Kavian, C. Nicco, P. Guilpain et al., Selective Oxidation of DNA Topoisomerase 1 Induces Systemic Sclerosis in the Mouse, The Journal of Immunology, vol.182, issue.9, pp.5855-5864, 2009.
DOI : 10.4049/jimmunol.0803705

URL : https://hal.archives-ouvertes.fr/hal-00517814

T. Hua-huy, N. Le-dong, S. Duong-quy, Y. Bei, S. Riviere et al., Increased exhaled nitric oxide precedes lung fibrosis in two murine models of systemic sclerosis, Journal of Breath Research, vol.9, issue.3, pp.36007-36017, 2015.
DOI : 10.1088/1752-7155/9/3/036007

N. Kavian, W. Marut, A. Servettaz, C. Nicco, C. Chereau et al., Reactive oxygen species-mediated killing of activated fibroblasts by arsenic trioxide ameliorates fibrosis in a murine model of systemic sclerosis, Arthritis & Rheumatism, vol.34, issue.10, pp.3430-3440, 2012.
DOI : 10.1002/art.34534

N. Kavian, A. Servettaz, W. Marut, C. Nicco, C. Chereau et al., Sunitinib inhibits the phosphorylation of platelet-derived growth factor receptor ?? in the skin of mice with scleroderma-like features and prevents the development of the disease, Arthritis & Rheumatism, vol.21, issue.6, pp.1990-2000, 2012.
DOI : 10.1002/art.34354

N. Kavian, A. Servettaz, C. Mongaret, A. Wang, C. Nicco et al., Targeting ADAM-17/notch signaling abrogates the development of systemic sclerosis in a murine model, Arthritis & Rheumatism, vol.13, issue.11, pp.3477-3487, 2010.
DOI : 10.1002/art.27626

URL : https://hal.archives-ouvertes.fr/hal-00554848

W. Marut, V. Jamier, N. Kavian, A. Servettaz, P. Winyard et al., The natural organosulfur compound dipropyltetrasulfide prevents HOCl-induced systemic sclerosis in the mouse, Arthritis Research & Therapy, vol.15, issue.5, pp.10-4351, 1186.
DOI : 10.4049/jimmunol.1103538

W. Marut, N. Kavian, A. Servettaz, T. Hua-huy, C. Nicco et al., Amelioration of Systemic Fibrosis in Mice by Angiotensin II Receptor Blockade, Arthritis & Rheumatism, vol.20, issue.5, pp.1367-1377, 2013.
DOI : 10.1002/art.37873

W. Marut, N. Kavian, A. Servettaz, C. Nicco, L. Ba et al., The Organotelluride Catalyst (PHTE)2NQ Prevents HOCl-Induced Systemic Sclerosis in Mouse, Journal of Investigative Dermatology, vol.132, issue.4, pp.1125-1132455, 2011.
DOI : 10.1038/jid.2011.455

A. Servettaz, N. Kavian, C. Nicco, V. Deveaux, C. Chereau et al., Targeting the Cannabinoid Pathway Limits the Development of Fibrosis and Autoimmunity in a Mouse Model of Systemic Sclerosis, The American Journal of Pathology, vol.177, issue.1, 2010.
DOI : 10.2353/ajpath.2010.090763

G. Bagnato, A. Bitto, N. Irrera, G. Pizzino, D. Sangari et al., Propylthiouracil prevents cutaneous and pulmonary fibrosis in the reactive oxygen species murine model of systemic sclerosis, Arthritis Research & Therapy, vol.15, issue.5, pp.120-130, 2013.
DOI : 10.1111/j.1540-8159.2009.02310.x

G. Bagnato, A. Bitto, G. Pizzino, N. Irrera, D. Sangari et al., Simvastatin attenuates the development of pulmonary and cutaneous fibrosis in a murine model of systemic sclerosis, Rheumatology, vol.52, issue.8, pp.1377-1386, 2013.
DOI : 10.1093/rheumatology/ket144

N. Naderi, E. Combellack, M. Griffin, T. Sedaghati, M. Javed et al., The regenerative role of adipose-derived stem cells (ADSC) in plastic and reconstructive surgery, International Wound Journal, vol.14, issue.9, 2016.
DOI : 10.1111/iwj.12569

J. Lataillade, C. Doucet, E. Bey, H. Carsin, C. Huet et al., New approach to radiation burn treatment by dosimetry-guided surgery combined with autologous mesenchymal stem cell therapy, Regenerative Medicine, vol.2, issue.5, pp.785-794, 2007.
DOI : 10.2217/17460751.2.5.785

E. Bey, M. Prat, P. Duhamel, M. Benderitter, M. Brachet et al., Emerging therapy for improving wound repair of severe radiation burns using local bone marrow-derived stem cell administrations, Wound Repair and Regeneration, vol.18, issue.1, pp.50-58, 2010.
DOI : 10.1111/j.1524-475X.2009.00562.x

T. Leclerc, C. Thepenier, P. Jault, E. Bey, J. Peltzer et al., Cell therapy of burns, Cell Proliferation, vol.35, issue.Suppl. A, 2011.
DOI : 10.1111/j.1365-2184.2010.00727.x

C. Linard, E. Busson, V. Holler, C. Strup-perrot, J. Lacave-lapalun et al., Repeated Autologous Bone Marrow-Derived Mesenchymal Stem Cell Injections Improve Radiation-Induced Proctitis in Pigs, STEM CELLS Translational Medicine, vol.130, issue.11, pp.916-927, 2013.
DOI : 10.5966/sctm.2013-0030

C. Linard, F. Tissedre, E. Busson, V. Holler, T. Leclerc et al., Therapeutic Potential of Gingival Fibroblasts for Cutaneous Radiation Syndrome: Comparison to Bone Marrow-Mesenchymal Stem Cell Grafts, Stem Cells and Development, vol.24, issue.10, pp.1182-1193, 2015.
DOI : 10.1089/scd.2014.0486

M. Christopeit, M. Schendel, J. Foll, L. Muller, G. Keysser et al., Marked improvement of severe progressive systemic sclerosis after transplantation of mesenchymal stem cells from an allogeneic haploidentical-related donor mediated by ligation of CD137L, Leukemia, vol.79, issue.5, pp.1062-1064, 2008.
DOI : 10.1038/sj.leu.2404996

G. Keyszer, M. Christopeit, S. Fick, M. Schendel, B. Taute et al., Treatment of severe progressive systemic sclerosis with transplantation of mesenchymal stromal cells from allogeneic related donors: Report of five cases, Arthritis & Rheumatism, vol.62, issue.5, pp.2540-2542, 2011.
DOI : 10.1002/art.30431

S. Guiducci, F. Porta, R. Saccardi, S. Guidi, L. Ibba-manneschi et al., Autologous Mesenchymal Stem Cells Foster Revascularization of Ischemic Limbs in Systemic Sclerosis, Annals of Internal Medicine, vol.153, issue.10, pp.650-654, 2010.
DOI : 10.7326/0003-4819-153-10-201011160-00007

B. Granel, A. Daumas, E. Jouve, J. Harle, P. Nguyen et al., Safety, tolerability and potential efficacy of injection of autologous adipose-derived stromal vascular fraction in the fingers of patients with systemic sclerosis: an open-label phase I trial, Annals of the Rheumatic Diseases, vol.74, issue.12, pp.2175-218210, 1136.
DOI : 10.1136/annrheumdis-2014-205681