H. Noorchashm, A. Reed, S. Rostami, R. Mozaffari, G. Zekavat et al., B Cell-Mediated Antigen Presentation Is Required for the Pathogenesis of Acute Cardiac Allograft Rejection, The Journal of Immunology, vol.177, issue.11, pp.7715-7737, 2006.
DOI : 10.4049/jimmunol.177.11.7715

J. Du, Q. Li, X. Ji, G. Chen, X. Bai et al., Inhibition of T-Cell Expansion Caused by Inducible Costimulator/B7h Costimulation Blockade in Direct Allorecognition Pathway, Transplantation Proceedings, vol.43, issue.10, pp.3960-3963, 2011.
DOI : 10.1016/j.transproceed.2011.09.044

A. Burns and A. Chong, Alloantibodies Prevent the Induction of Transplantation Tolerance by Enhancing Alloreactive T Cell Priming, The Journal of Immunology, vol.186, issue.1, pp.214-235, 2011.
DOI : 10.4049/jimmunol.1001172

M. Sarwal, M. Chua, N. Kambham, S. Hsieh, T. Satterwhite et al., Molecular Heterogeneity in Acute Renal Allograft Rejection Identified by DNA Microarray Profiling, New England Journal of Medicine, vol.349, issue.2, pp.125-163, 2003.
DOI : 10.1056/NEJMoa035588

V. Zarkhin, N. Kambham, L. Li, S. Kwok, S. Hsieh et al., Characterization of intra-graft B cells during renal allograft rejection, Kidney International, vol.74, issue.5, pp.664-73249, 2008.
DOI : 10.1038/ki.2008.249

B. Hippen, A. Demattos, W. Cook, C. Kew, . Ii et al., Association of CD20+ Infiltrates with Poorer Clinical Outcomes in Acute Cellular Rejection of Renal Allografts, American Journal of Transplantation, vol.50, issue.9, pp.2248-52, 2005.
DOI : 10.1093/intimm/13.12.1583

J. Kwun and S. Knechtle, Overcoming chronic rejection-can it B? Transplantation, pp.955-61, 2009.
DOI : 10.1097/tp.0b013e3181b96646

K. Lamb, S. Lodhi, and H. Meier-kriesche, Long-Term Renal Allograft Survival in the United States: A Critical Reappraisal, American Journal of Transplantation, vol.3, issue.Suppl 2, pp.450-62, 2011.
DOI : 10.1111/j.1600-6143.2010.03283.x

H. Meier-kriesche, J. Schold, T. Srinivas, and B. Kaplan, Lack of Improvement in Renal Allograft Survival Despite a Marked Decrease in Acute Rejection Rates Over the Most Recent Era, American Journal of Transplantation, vol.70, issue.3, pp.378-83, 2004.
DOI : 10.1111/j.1600-6143.2004.00332.x

S. Mcdonald, G. Russ, S. Campbell, and S. Chadban, Kidney Transplant Rejection in Australia and New Zealand: Relationships Between Rejection and Graft Outcome, American Journal of Transplantation, vol.130, issue.6
DOI : 10.1097/01.tp.0000219703.39149.85

P. Libby and J. Pober, Chronic Rejection, Immunity, vol.14, issue.4, pp.387-97, 2001.
DOI : 10.1016/S1074-7613(01)00119-4

U. Steger, S. Ensminger, A. Bushell, and K. Wood, Investigation into the onset and progression of transplant arteriosclerosis in a mice aortic retransplantation model, Microsurgery, vol.223, issue.3, pp.182-188, 2008.
DOI : 10.1002/micr.20477

S. Nadig, J. Wieckiewicz, D. Wu, G. Warnecke, W. Zhang et al., In vivo prevention of transplant arteriosclerosis by ex vivo???expanded human regulatory T cells, Nature Medicine, vol.8, issue.7, pp.809-822, 2010.
DOI : 10.1038/nm.2154

J. Worthington, S. Martin, D. Husseini, P. Dyer, and R. Johnson, Posttransplantation production of donor HLA-specific antibodies as a predictor of renal transplant outcome, Transplantation, vol.75, 2003.

O. Thaunat, A. Field, J. Dai, L. Louedec, N. Patey et al., Lymphoid neogenesis in chronic rejection: Evidence for a local humoral alloimmune response, Proceedings of the National Academy of Sciences, vol.102, issue.41, pp.14723-14731, 2005.
DOI : 10.1073/pnas.0507223102

C. Sorrentino, A. Scarinci, T. Piccirilli, M. , D. Nicola et al., Endomyocardial infiltration by B and NK cells foreshadows the recurrence of cardiac allograft rejection, The Journal of Pathology, vol.63, issue.3, pp.400-410, 1980.
DOI : 10.1002/path.1980

C. Mauri, Regulation of immunity and autoimmunity by B cells, Current Opinion in Immunology, vol.22, issue.6, pp.761-768, 2010.
DOI : 10.1016/j.coi.2010.10.009

L. Texier, L. Thebault, P. Lavault, A. Usal, C. Merieau et al., Long-Term Allograft Tolerance Is Characterized by the Accumulation of B Cells Exhibiting an Inhibited Profile, American Journal of Transplantation, vol.15, issue.3, 2011.
DOI : 10.1111/j.1600-6143.2010.03336.x

Y. Li, T. Koshiba, A. Yoshizawa, Y. Yonekawa, K. Masuda et al., Analyses of Peripheral Blood Mononuclear Cells in Operational Tolerance After Pediatric Living Donor Liver Transplantation, American Journal of Transplantation, vol.168, issue.12, pp.2118-2143, 2004.
DOI : 10.1034/j.1600-0897.2002.01131.x

K. Newell, A. Asare, A. Kirk, T. Gisler, K. Bourcier et al., Identification of a B cell signature associated with renal transplant tolerance in humans, Journal of Clinical Investigation, vol.120, issue.6, pp.1836-1883, 2010.
DOI : 10.1172/JCI39933DS1

A. Pallier, S. Hillion, R. Danger, M. Giral, M. Racape et al., Patients with drug-free long-term graft function display increased numbers of peripheral B cells with a memory and inhibitory phenotype, Kidney International, vol.78, issue.5, pp.503-516, 2010.
DOI : 10.1038/ki.2010.162

H. Silva, M. Takenaka, P. Moraes-vieira, S. Monteiro, M. Hernandez et al., Preserving the B cell compartment favors operational tolerance in human renal transplantation, Mol Med, vol.18, pp.733-776, 2012.

P. Blair, L. Norena, F. Flores-borja, D. Rawlings, D. Isenberg et al., CD19+CD24hiCD38hi B Cells Exhibit Regulatory Capacity in Healthy Individuals but Are Functionally Impaired in Systemic Lupus Erythematosus Patients, Immunity, vol.32, issue.1, pp.129-169, 2010.
DOI : 10.1016/j.immuni.2009.11.009

L. Haynes, E. Jankowska-gan, A. Sheka, M. Keller, M. Hernandez-fuentes et al., Donor-specific indirect pathway analysis reveals a B-cellindependent signature which reflects outcomes in kidney transplant recipients

P. Matzinger and K. T. , Tissue-based class control: the other side of tolerance, Nature Reviews Immunology, vol.204, issue.3, pp.221-251, 2011.
DOI : 10.1038/nri2940

T. Lebien and T. Tedder, B lymphocytes: how they develop and function, Blood, vol.112, issue.5, pp.1570-80, 2008.
DOI : 10.1182/blood-2008-02-078071

S. Wolf, B. Dittel, F. Hardardottir, C. Janeway, and . Jr, Experimental Autoimmune Encephalomyelitis Induction in Genetically B Cell-deficient Mice, Journal of Experimental Medicine, vol.184, issue.6, 1996.
DOI : 10.1084/jem.184.6.2271

S. Fillatreau, C. Sweenie, M. Mcgeachy, D. Gray, and S. Anderton, B cells regulate autoimmunity by provision of IL-10, Nature Immunology, vol.3, issue.10, pp.944-50, 2002.
DOI : 10.1038/ni833

A. Mizoguchi, E. Mizoguchi, H. Takedatsu, R. Blumberg, and A. Bhan, Chronic Intestinal Inflammatory Condition Generates IL-10-Producing Regulatory B Cell Subset Characterized by CD1d Upregulation, Immunity, vol.16, issue.2, pp.219-249, 2002.
DOI : 10.1016/S1074-7613(02)00274-1

M. Gray, K. Miles, D. Salter, D. Gray, and J. Savill, Apoptotic cells protect mice from autoimmune inflammation by the induction of regulatory B cells, Proceedings of the National Academy of Sciences, vol.104, issue.35, pp.14080-14085, 2007.
DOI : 10.1073/pnas.0700326104

M. Matsumoto, Y. Fujii, A. Baba, M. Hikida, T. Kurosaki et al., The Calcium Sensors STIM1 and STIM2 Control B Cell Regulatory Function through Interleukin-10 Production, Immunity, vol.34, issue.5, pp.703-717, 2011.
DOI : 10.1016/j.immuni.2011.03.016

C. Mauri and A. Bosma, Immune Regulatory Function of B Cells, Annual Review of Immunology, vol.30, issue.1, pp.221-262, 2012.
DOI : 10.1146/annurev-immunol-020711-074934

T. Matsushita and T. Tedder, Identifying Regulatory B Cells (B10 Cells) that Produce IL-10 in Mice, Methods Mol Biol, vol.677, pp.99-111, 2011.
DOI : 10.1007/978-1-60761-869-0_7

J. Evans, K. Chavez-rueda, A. Eddaoudi, A. Meyer-bahlburg, D. Rawlings et al., Novel Suppressive Function of Transitional 2 B Cells in Experimental Arthritis, The Journal of Immunology, vol.178, issue.12, pp.7868-78, 2007.
DOI : 10.4049/jimmunol.178.12.7868

A. Mizoguchi, E. Mizoguchi, R. Smith, F. Preffer, and A. Bhan, Suppressive Role of B Cells in Chronic Colitis of ???T Cell Receptor ?? Mutant Mice, The Journal of Experimental Medicine, vol.153, issue.10, pp.1749-56, 1997.
DOI : 10.1016/0090-1229(75)90087-2

C. Mauri, D. Gray, N. Mushtaq, and M. Londei, Prevention of Arthritis by Interleukin 10???producing B Cells, The Journal of Experimental Medicine, vol.147, issue.4, pp.489-501, 2003.
DOI : 10.1016/S1074-7613(02)00274-1

R. Watanabe, M. Fujimoto, N. Ishiura, Y. Kuwano, H. Nakashima et al., CD19 Expression in B Cells Is Important for Suppression of Contact Hypersensitivity, The American Journal of Pathology, vol.171, issue.2, pp.560-70, 2007.
DOI : 10.2353/ajpath.2007.061279

K. Yanaba, J. Bouaziz, K. Haas, J. Poe, M. Fujimoto et al., A Regulatory B Cell Subset with a Unique CD1dhiCD5+ Phenotype Controls T Cell-Dependent Inflammatory Responses, Immunity, vol.28, issue.5, pp.639-50, 2008.
DOI : 10.1016/j.immuni.2008.03.017

Y. Shimomura, E. Mizoguchi, K. Sugimoto, R. Kibe, Y. Benno et al., Regulatory role of B-1 B cells in chronic colitis, International Immunology, vol.20, issue.6, pp.729-766, 2008.
DOI : 10.1093/intimm/dxn031

Q. Ding, M. Yeung, G. Camirand, Q. Zeng, H. Akiba et al., Regulatory B cells are identified by expression of TIM-1 and can be induced through TIM-1 ligation to promote tolerance in mice, Journal of Clinical Investigation, vol.121, issue.9, pp.3645-5610, 1172.
DOI : 10.1172/JCI46274DS1

L. Qian, C. Qian, Y. Chen, Y. Bai, Y. Bao et al., Regulatory dendritic cells program B cells to differentiate into CD19hiFc??IIbhi regulatory B cells through IFN-?? and CD40L, Blood, vol.120, issue.3, pp.581-91, 2012.
DOI : 10.1182/blood-2011-08-377242

F. Jensen, D. Muzzio, R. Soldati, S. Fest, and A. Zenclussen, Regulatory B10 cells restore pregnancy tolerance in a mouse model, Biol Reprod, vol.89, 2013.

Z. Liu, Y. Wu, J. Song, X. Liu, Z. Liu et al., B cells suppress food allergy-induced intestinal inflammation in mice, Allergy, vol.147, issue.10, pp.1241-1249, 2013.
DOI : 10.1111/all.12218

Y. Iwata, T. Matsushita, M. Horikawa, D. Dilillo, K. Yanaba et al., Characterization of a rare IL-10-competent B-cell subset in humans that parallels mouse regulatory B10 cells, Blood, vol.117, issue.2, pp.530-571, 2011.
DOI : 10.1182/blood-2010-07-294249

S. Lemoine, A. Morva, P. Youinou, and C. Jamin, Human T cells induce their own regulation through activation of B cells, Journal of Autoimmunity, vol.36, issue.3-4, 2011.
DOI : 10.1016/j.jaut.2011.01.005

URL : https://hal.archives-ouvertes.fr/hal-00771265

X. Li, H. Zhong, W. Bao, N. Boulad, J. Evangelista et al., Defective regulatory B-cell compartment in patients with immune thrombocytopenia, Blood, vol.120, issue.16, pp.3318-3343, 2012.
DOI : 10.1182/blood-2012-05-432575

B. Zha, L. Wang, X. Liu, J. Liu, Z. Chen et al., Decrease in proportion of CD19+ CD24(hi) CD27+ B cells and impairment of their suppressive function in Graves' disease, PLoS One, 2012.

I. Kalampokis, A. Yoshizaki, and T. Tedder, IL-10-producing regulatory B cells (B10 cells) in autoimmune disease, Arthritis Research & Therapy, vol.15, issue.Suppl 1, pp.10-1186, 2013.
DOI : 10.1002/eji.201041041

S. Lindner, K. Dahlke, K. Sontheimer, M. Hagn, C. Kaltenmeier et al., Interleukin 21-Induced Granzyme B-Expressing B Cells Infiltrate Tumors and Regulate T Cells, Cancer Research, vol.73, issue.8, pp.2468-79, 2013.
DOI : 10.1158/0008-5472.CAN-12-3450

W. Van-de-veen, B. Stanic, G. Yaman, M. Wawrzyniak, S. Sollner et al., IgG4 production is confined to human IL-10???producing regulatory B cells that suppress antigen-specific immune responses, Journal of Allergy and Clinical Immunology, vol.131, issue.4, pp.1204-1216, 2013.
DOI : 10.1016/j.jaci.2013.01.014

S. Deng, D. Moore, X. Huang, M. Lian, M. Mohiuddin et al., Cutting Edge: Transplant Tolerance Induced by Anti-CD45RB Requires B Lymphocytes, The Journal of Immunology, vol.178, issue.10, pp.6028-6060, 2007.
DOI : 10.4049/jimmunol.178.10.6028

V. Lampropoulou, K. Hoehlig, T. Roch, P. Neves, C. Gomez et al., TLR-Activated B Cells Suppress T Cell-Mediated Autoimmunity, The Journal of Immunology, vol.180, issue.7, pp.4763-73, 2008.
DOI : 10.4049/jimmunol.180.7.4763

URL : https://hal.archives-ouvertes.fr/pasteur-00324528

T. Barr, S. Brown, G. Ryan, J. Zhao, and D. Gray, TLR-mediated stimulation of APC: Distinct cytokine responses of B cells and dendritic cells, European Journal of Immunology, vol.176, issue.11, pp.3040-53, 2007.
DOI : 10.1002/eji.200636483

R. Brummel and P. Lenert, Activation of Marginal Zone B Cells from Lupus Mice with Type A(D) CpG-Oligodeoxynucleotides, The Journal of Immunology, vol.174, issue.4, pp.2429-2463, 2005.
DOI : 10.4049/jimmunol.174.4.2429

K. Yanaba, J. Bouaziz, T. Matsushita, T. Tsubata, and T. Tedder, The Development and Function of Regulatory B Cells Expressing IL-10 (B10 Cells) Requires Antigen Receptor Diversity and TLR Signals, The Journal of Immunology, vol.182, issue.12, pp.7459-72, 2009.
DOI : 10.4049/jimmunol.0900270

J. Poe, S. Smith, K. Haas, K. Yanaba, T. Tsubata et al., Amplified B Lymphocyte CD40 Signaling Drives Regulatory B10 Cell Expansion in Mice, PLoS ONE, vol.189, issue.7, 2011.
DOI : 10.1371/journal.pone.0022464.t001

A. Yoshizaki, T. Miyagaki, D. Dilillo, T. Matsushita, M. Horikawa et al., Regulatory B cells control T-cell autoimmunity through IL-21-dependent cognate interactions, Nature, vol.3, issue.7423, pp.264-272, 2012.
DOI : 10.1038/nature11501

M. Sabet-baktach, E. Eggenhofer, J. Rovira, P. Renner, M. Lantow et al., Double Deficiency for ROR??t and T-bet Drives Th2-Mediated Allograft Rejection in Mice, The Journal of Immunology, vol.191, issue.8, pp.4440-4446, 2013.
DOI : 10.4049/jimmunol.1301741

G. Sims, R. Ettinger, Y. Shirota, C. Yarboro, G. Illei et al., Identification and characterization of circulating human transitional B cells, Blood, vol.105, issue.11, pp.4390-4398, 2005.
DOI : 10.1182/blood-2004-11-4284

C. Jamin, A. Morva, S. Lemoine, C. Daridon, D. Mendoza et al., Regulatory B lymphocytes in humans: A potential role in autoimmunity, Arthritis & Rheumatism, vol.54, issue.7, pp.1900-1906, 2008.
DOI : 10.1002/art.23487

A. Kashipaz, M. Huggins, M. Lanyon, P. Robins, A. Powell et al., Assessment of Be1 and Be2 cells in systemic lupus erythematosus indicates elevated interleukin-10 producing CD5+ B cells, Lupus, vol.2, issue.5, pp.356-63, 2003.
DOI : 10.1191/0961203303lu338oa

F. Gantner, P. Hermann, K. Nakashima, S. Matsukawa, K. Sakai et al., CD40-dependent and -independent activation of human tonsil B???cells by CpG oligodeoxynucleotides, European Journal of Immunology, vol.33, issue.6, pp.1576-85, 2003.
DOI : 10.1002/eji.200323444

J. Furuzawa-carballeda, G. Lima, P. Simancas, D. Ramos-bello, M. Simancas et al., Peripheral Regulatory Cells Immunophenotyping in Kidney Transplant Recipients with Different Clinical Profiles: A Cross-Sectional Study, Journal of Transplantation, vol.175, issue.9, 2012.
DOI : 10.1177/0192623311430693

A. Nouël, I. Ségalen, C. Jamin, L. Doucet, S. Caillard et al., B cells display an abnormal distribution and an impaired suppressive function in patients with chronic antibody???mediated rejection, Kidney International, vol.85, issue.3, 2013.
DOI : 10.1038/ki.2013.457

J. Trapani and V. Sutton, Granzyme B: pro-apoptotic, antiviral and antitumor functions, Current Opinion in Immunology, vol.15, issue.5, pp.533-4310, 2003.
DOI : 10.1016/S0952-7915(03)00107-9

S. Lundy, Killer B lymphocytes: the evidence and the potential, Inflammation Research, vol.168, issue.7, pp.345-57, 2009.
DOI : 10.1007/s00011-009-0014-x

M. Mann, K. Maresz, L. Shriver, Y. Tan, and B. Dittel, B Cell Regulation of CD4+CD25+ T Regulatory Cells and IL-10 Via B7 is Essential for Recovery From Experimental Autoimmune Encephalomyelitis, The Journal of Immunology, vol.178, issue.6, pp.3447-56, 2007.
DOI : 10.4049/jimmunol.178.6.3447

A. Singh, W. Carson, E. Secor, . Jr, L. Guernsey et al., Regulatory Role of B Cells in a Murine Model of Allergic Airway Disease, The Journal of Immunology, vol.180, issue.11, pp.7318-7344, 2008.
DOI : 10.4049/jimmunol.180.11.7318

S. Lemoine, A. Morva, P. Youinou, and C. Jamin, Regulatory B Cells in Autoimmune Diseases, Annals of the New York Academy of Sciences, vol.118, issue.1, pp.260-267, 2009.
DOI : 10.1111/j.1749-6632.2009.04651.x

Y. Chen, Y. Park, E. Patel, and G. Silverman, IgM Antibodies to Apoptosis-Associated Determinants Recruit C1q and Enhance Dendritic Cell Phagocytosis of Apoptotic Cells, The Journal of Immunology, vol.182, issue.10, pp.6031-6074, 2009.
DOI : 10.4049/jimmunol.0804191

Y. Chen, S. Khanna, C. Goodyear, Y. Park, E. Raz et al., Regulation of Dendritic Cells and Macrophages by an Anti-Apoptotic Cell Natural Antibody that Suppresses TLR Responses and Inhibits Inflammatory Arthritis, The Journal of Immunology, vol.183, issue.2, pp.1346-59, 2009.
DOI : 10.4049/jimmunol.0900948

Y. Kaneko, F. Nimmerjahn, and J. Ravetch, Anti-Inflammatory Activity of Immunoglobulin G Resulting from Fc Sialylation, Science, vol.313, issue.5787, pp.670-673, 2006.
DOI : 10.1126/science.1129594

J. Seite, C. Goutsmedt, P. Youinou, J. Pers, and S. Hillion, Intravenous immunoglobulin induces a functional silencing program similar to anergy in human B cells, J Allergy Clin Immunol, vol.133, issue.1, 2013.
URL : https://hal.archives-ouvertes.fr/hal-00926676

C. Hess, A. Winkler, A. Lorenz, V. Holecska, V. Blanchard et al., T cell???independent B cell activation induces immunosuppressive sialylated IgG antibodies, Journal of Clinical Investigation, vol.123, issue.9, pp.3788-96, 2013.
DOI : 10.1172/JCI65938DS1

M. Willicombe, C. Roufosse, P. Brookes, J. Galliford, A. Mclean et al., Antibody-Mediated Rejection After Alemtuzumab Induction: Incidence, Risk Factors, and Predictors of Poor Outcome, Transplantation, vol.92, issue.2, pp.176-82, 2011.
DOI : 10.1097/TP.0b013e318222c9c6

M. Clatworthy, Targeting B Cells and Antibody in Transplantation, American Journal of Transplantation, vol.9, issue.suppl 2, pp.1359-67, 2011.
DOI : 10.1111/j.1600-6143.2011.03554.x

D. Ducloux, C. Courivaud, J. Bamoulid, B. Vivet, A. Chabroux et al., Prolonged CD4 T Cell Lymphopenia Increases Morbidity and Mortality after Renal Transplantation, Journal of the American Society of Nephrology, vol.21, issue.5, pp.868-75, 2010.
DOI : 10.1681/ASN.2009090976

URL : https://hal.archives-ouvertes.fr/inserm-00484264

A. Thibault-espitia, Y. Foucher, R. Danger, T. Migone, A. Pallier et al., BAFF and BAFF-R Levels Are Associated With Risk of Long-Term Kidney Graft Dysfunction and Development of Donor-Specific Antibodies, American Journal of Transplantation, vol.80, issue.9 Suppl, pp.2754-62, 2012.
DOI : 10.1111/j.1600-6143.2012.04194.x

G. Banham, D. Prezzi, S. Harford, C. Taylor, R. Hamer et al., Elevated Pretransplantation Soluble BAFF Is Associated With an Increased Risk of Acute Antibody-Mediated Rejection, Transplantation Journal, vol.96, issue.4, pp.413-433, 2013.
DOI : 10.1097/TP.0b013e318298dd65

J. Kwun, P. Bulut, E. Kim, W. Dar, B. Oh et al., The role of B cells in solid organ transplantation, Seminars in Immunology, vol.24, issue.2, pp.96-108, 2012.
DOI : 10.1016/j.smim.2011.08.022

T. Matsushita, K. Yanaba, J. Bouaziz, M. Fujimoto, and T. Tedder, Regulatory B cells inhibit EAE initiation in mice while other B cells promote disease progression, Journal of Clinical Investigation, vol.118, pp.3420-3450, 2008.
DOI : 10.1172/JCI36030

M. Clatworthy, C. Watson, G. Plotnek, V. Bardsley, A. Chaudhry et al., B-Cell???Depleting Induction Therapy and Acute Cellular Rejection, New England Journal of Medicine, vol.360, issue.25, pp.2683-2688, 2009.
DOI : 10.1056/NEJMc0808481

URL : http://www.ncbi.nlm.nih.gov/pmc/articles/PMC4143588

T. Kurosaki, Paradox of B cell???targeted therapies, Journal of Clinical Investigation, vol.118, pp.3260-3263, 2008.
DOI : 10.1172/JCI37099

N. Barnett, A. Dorling, N. Mamode, E. Hoevenaars, S. Yong et al., B cells in renal transplantation: pathological aspects and therapeutic interventions, Nephrology Dialysis Transplantation, vol.26, issue.3, pp.767-74198, 2011.
DOI : 10.1093/ndt/gfq716

A. Chong and R. Sciammas, Matchmaking the B-Cell Signature of Tolerance to Regulatory B Cells, American Journal of Transplantation, vol.2, issue.12, pp.2555-60, 2011.
DOI : 10.1111/j.1600-6143.2011.03773.x

T. Dorner, J. Kaufmann, W. Wegener, N. Teoh, D. Goldenberg et al., Correction: Initial clinical trial of epratuzumab (humanized anti-CD22 antibody) for immunotherapy of systemic lupus erythematosus, Arthritis Research & Therapy, vol.10, issue.5, pp.10-1186, 2006.
DOI : 10.1186/ar2499

C. Notley, M. Brown, G. Wright, and M. Ehrenstein, Natural IgM Is Required for Suppression of Inflammatory Arthritis by Apoptotic Cells, The Journal of Immunology, vol.186, issue.8, pp.4967-72, 2011.
DOI : 10.4049/jimmunol.1003021