N. Polanco, Spontaneous Remission of Nephrotic Syndrome in Idiopathic Membranous Nephropathy, Journal of the American Society of Nephrology, vol.21, issue.4, pp.697-704, 2010.
DOI : 10.1681/ASN.2009080861

N. Polanco, Spontaneous remission of nephrotic syndrome in membranous nephropathy with chronic renal impairment, Nephrology Dialysis Transplantation, vol.27, issue.1, pp.231-234, 2012.
DOI : 10.1093/ndt/gfr285

R. J. Glassock, Diagnosis and natural course of membranous nephropathy, Seminars in Nephrology, vol.23, issue.4, pp.324-332, 2003.
DOI : 10.1016/S0270-9295(03)00049-4

D. Cattran, Management of Membranous Nephropathy: When and What for Treatment, Journal of the American Society of Nephrology, vol.16, issue.5, pp.1188-1194, 2005.
DOI : 10.1681/ASN.2005010028

R. J. Glassock, The treatment of idiopathic membranous nephropathy: A dilemma or a conundrum?, American Journal of Kidney Diseases, vol.44, issue.3, pp.562-566, 2004.
DOI : 10.1016/S0272-6386(04)00868-6

M. Waldman, H. A. Austin, and . Iii, Controversies in the treatment of idiopathic membranous nephropathy, Nature Reviews Nephrology, vol.19, issue.8, pp.469-479, 2009.
DOI : 10.1038/nrneph.2009.101

D. C. Cattran, H. N. Reich, S. J. Kim, and S. Troyanov, Have We Changed the Outcome in Membranous Nephropathy? A Propensity Study on the Role of Immunosuppressive Therapy, Clinical Journal of the American Society of Nephrology, vol.6, issue.7, pp.1591-1598, 2011.
DOI : 10.2215/CJN.11001210

W. Heymann, D. B. Hackel, S. Harwood, S. G. Wilson, and J. L. Hunter, Production of Nephrotic Syndrome in Rats by Freund's Adjuvants and Rat Kidney Suspensions., Experimental Biology and Medicine, vol.100, issue.4, pp.660-664, 1959.
DOI : 10.3181/00379727-100-24736

B. J. Van-damme, G. J. Fleuren, W. W. Bakker, R. L. Vernier, and P. J. Hoedemaeker, Experimental glomerulonephritis in the rat induced by antibodies directed against tubular antigens. V. Fixed glomerular antigens in the pathogenesis of heterologous immune complex glomerulonephritis, Lab. Invest, vol.38, pp.502-510, 1978.

W. G. Couser, D. R. Steinmuller, M. M. Stilmant, D. J. Salant, and L. M. Lowenstein, Experimental glomerulonephritis in the isolated perfused rat kidney., Journal of Clinical Investigation, vol.62, issue.6, pp.1275-1287, 1978.
DOI : 10.1172/JCI109248

W. A. Border, H. J. Ward, E. S. Kamil, and A. H. Cohen, Induction of Membranous Nephropathy in Rabbits by Administration of an Exogenous Cationic Antigen, Journal of Clinical Investigation, vol.69, issue.2, pp.451-461, 1982.
DOI : 10.1172/JCI110469

S. G. Adler, H. Wang, H. J. Ward, A. H. Cohen, and W. A. Border, Electrical Charge. ITS ROLE IN THE PATHOGENESIS AND PREVENTION OF EXPERIMENTAL MEMBRANOUS NEPHROPATHY IN THE RABBIT, Journal of Clinical Investigation, vol.71, issue.3, pp.487-499, 1983.
DOI : 10.1172/JCI110793

D. Kerjaschki and M. G. Farquhar, The pathogenic antigen of Heymann nephritis is a membrane glycoprotein of the renal proximal tubule brush border., Proc. Natl Acad. Sci. USA 79, pp.5557-5561, 1982.
DOI : 10.1073/pnas.79.18.5557

D. Kerjaschki and M. G. Farquhar, Immunocytochemical localization of the Heymann nephritis antigen (GP330) in glomerular epithelial cells of normal Lewis rats, Journal of Experimental Medicine, vol.157, issue.2, pp.667-686, 1983.
DOI : 10.1084/jem.157.2.667

H. Debiec, Early-Childhood Membranous Nephropathy Due to Cationic Bovine Serum Albumin, New England Journal of Medicine, vol.364, issue.22, pp.2101-2110, 2011.
DOI : 10.1056/NEJMoa1013792

H. Debiec, Antenatal Membranous Glomerulonephritis Due to Anti???Neutral Endopeptidase Antibodies, New England Journal of Medicine, vol.346, issue.26, pp.2053-2060, 2002.
DOI : 10.1056/NEJMoa012895

H. Debiec, Role of truncating mutations in MME gene in fetomaternal alloimmunisation and antenatal glomerulopathies, The Lancet, vol.364, issue.9441, pp.1252-1259, 2004.
DOI : 10.1016/S0140-6736(04)17142-0

L. H. Beck and . Jr, Receptor as Target Antigen in Idiopathic Membranous Nephropathy, New England Journal of Medicine, vol.361, issue.1, pp.11-21, 2009.
DOI : 10.1056/NEJMoa0810457

URL : https://hal.archives-ouvertes.fr/hal-00417718

H. C. Stanescu, R1 Alleles in Idiopathic Membranous Nephropathy, New England Journal of Medicine, vol.364, issue.7, pp.616-626, 2011.
DOI : 10.1056/NEJMoa1009742

M. Prunotto, Autoimmunity in Membranous Nephropathy Targets Aldose Reductase and SOD2, Journal of the American Society of Nephrology, vol.21, issue.3, pp.507-519, 2010.
DOI : 10.1681/ASN.2008121259

M. Bruschi, Direct characterization of target podocyte antigens and auto-antibodies in human membranous glomerulonephritis: Alfa-enolase and borderline antigens, Journal of Proteomics, vol.74, issue.10, pp.2008-2017, 2011.
DOI : 10.1016/j.jprot.2011.05.021

W. G. Couser and D. J. Salant, In situ immune complex formation and glomerular injury, Kidney International, vol.17, issue.1, pp.1-13, 1980.
DOI : 10.1038/ki.1980.1

F. J. Dixon, J. D. Feldman, and J. J. Vazquez, EXPERIMENTAL GLOMERULONEPHRITIS: THE PATHOGENESIS OF A LABORATORY MODEL RESEMBLING THE SPECTRUM OF HUMAN GLOMERULONEPHRITIS, Journal of Experimental Medicine, vol.113, issue.5
DOI : 10.1084/jem.113.5.899

C. B. Wilson and F. J. Dixon, Quantitation of acute and chronic serum sickness in the rabbit, J. Exp. Med, vol.134, pp.7-18, 1971.

A. Saito, Mapping rat megalin: the second cluster of ligand binding repeats contains a 46-amino acid pathogenic epitope involved in the formation of immune deposits in Heymann nephritis., Proc. Natl Acad. Sci. USA 93, pp.8601-8605, 1996.
DOI : 10.1073/pnas.93.16.8601

H. Yamazaki, All four putative ligand?binding domains in megalin contain pathogenic epitopes capable of inducing passive Heymann nephritis, J. Am. Soc. Nephrol, vol.9, pp.1638-1644, 1998.

L. Allegri, Polyvalent antigen?antibody interactions are required for the formation of electron?dense immune deposits in passive Heymann's nephritis, Am. J. Pathol, vol.125, pp.1-6, 1986.

R. Raychowdhury, G. Zheng, D. Brown, and R. T. Mccluskey, Induction of Heymann nephritis with a gp330/megalin fusion protein, Am. J. Pathol, vol.148, pp.1613-1623, 1996.

A. V. Oleinikov, B. J. Feliz, and S. P. Makker, A small N?terminal 60?kD fragment of gp600 (megalin), the major autoantigen of active Heymann nephritis, can induce a full?blown disease, J. Am. Soc. Nephrol, vol.11, pp.57-64, 2000.

A. Tramontano and S. P. Makker, Conformation and Glycosylation of a Megalin Fragment Correlate with Nephritogenicity in Heymann Nephritis, The Journal of Immunology, vol.172, issue.4
DOI : 10.4049/jimmunol.172.4.2367

A. Tramontano, T. Knight, D. Vizzuso, and S. P. Makker, Nested N-Terminal Megalin Fragments Induce High-Titer Autoantibody and Attenuated Heymann Nephritis, Journal of the American Society of Nephrology, vol.17, issue.7, pp.1979-1985, 2006.
DOI : 10.1681/ASN.2005101144

P. Shah, A. Tramontano, and S. P. Makker, Intramolecular Epitope Spreading in Heymann Nephritis, Journal of the American Society of Nephrology, vol.18, issue.12, pp.3060-3066, 2007.
DOI : 10.1681/ASN.2007030342

P. Ronco, A monoclonal antibody to brush border and passive Heymann nephritis, Clin. Exp. Immunol, vol.55, pp.319-332, 1984.

K. J. Assmann, M. M. Tangelder, W. P. Lange, T. M. Tadema, and R. A. Koene, Membranous glomerulonephritis in the mouse, Kidney International, vol.24, issue.3, pp.303-312, 1983.
DOI : 10.1038/ki.1983.159

K. J. Assmann, Comparison of antigenic targets involved in antibody?mediated membranous glomerulonephritis in the mouse and rat, Am. J. Pathol, vol.121, pp.112-122, 1985.

K. J. Assmann, Involvement of an antigen distinct from the Heymann antigen in membranous glomerulonephritis in the mouse, Lab. Invest, vol.60, pp.138-146, 1989.

T. N. Meyer, A new mouse model of immune-mediated podocyte injury, Kidney International, vol.72, issue.7, pp.841-852, 2007.
DOI : 10.1038/sj.ki.5002450

C. Meyer?schwesinger, Nephrotic Syndrome and Subepithelial Deposits in a Mouse Model of Immune-Mediated Anti-Podocyte Glomerulonephritis, The Journal of Immunology, vol.187, issue.6, pp.3218-3229, 2011.
DOI : 10.4049/jimmunol.1003451

A. V. Cybulsky, R. J. Quigg, and D. J. Salant, Experimental membranous nephropathy redux, AJP: Renal Physiology, vol.289, issue.4, pp.660-671, 2005.
DOI : 10.1152/ajprenal.00437.2004

M. Nangaku, S. J. Shankland, and W. G. Couser, Cellular Response to Injury in Membranous Nephropathy, Journal of the American Society of Nephrology, vol.16, issue.5, pp.1195-1204, 2005.
DOI : 10.1681/ASN.2004121098

P. N. Cunningham and R. J. Quigg, Contrasting Roles of Complement Activation and Its Regulation in Membranous Nephropathy, Journal of the American Society of Nephrology, vol.16, issue.5, pp.1214-1222, 2005.
DOI : 10.1681/ASN.2005010096

A. Koyama, H. Inage, M. Kobayashi, M. Narita, and S. Tojo, Effect of chemical cationization of antigen on glomerular localization of immune complexes in active models of serum sickness nephritis in rabbits, Immunology, vol.58, pp.529-534, 1986.

P. S. Bass, A. F. Drake, Y. Wang, J. H. Thomas, and D. R. Davies, Cationization of bovine serum albumin alters its conformation as well as its charge, Lab. Invest, vol.62, pp.185-188, 1990.

N. G. Wright, N. A. Mohammed, P. D. Eckersall, and A. S. Nash, Experimental immune complex glomerulonephritis in dogs receiving cationized bovine serum albumin, Res. Vet. Sci, vol.38, pp.322-328, 1985.

A. Koyama, Role of antigenic charge and antibody avidity on the glomerular immune complex localization in serum sickness of mice, Clin. Exp. Immunol, vol.64, pp.606-614, 1986.

J. S. Chen, Mouse model of membranous nephropathy induced by cationic bovine serum albumin: antigen dose-response relations and strain differences, Nephrology Dialysis Transplantation, vol.19, issue.11, pp.2721-2728, 2004.
DOI : 10.1093/ndt/gfh419

C. C. Wu, Kinetics of adaptive immunity to cationic bovine serum albumin-induced membranous nephropathy, Kidney International, vol.72, issue.7, pp.831-840, 2007.
DOI : 10.1038/sj.ki.5002426

R. E. Urizar, J. Cerda, and A. Reilly, Glomerulitis induced by cationized bovine serum albumin in the rat, Pediatric Nephrology, vol.134, issue.Suppl, pp.149-155, 1989.
DOI : 10.1007/BF00852897

T. M. Schmiedeke, Histones have high affinity for the glomerular basement membrane. Relevance for immune complex formation in lupus nephritis, Journal of Experimental Medicine, vol.169, issue.6, pp.1879-1894, 1989.
DOI : 10.1084/jem.169.6.1879

R. M. Termaat, Anti-DNA antibodies can bind to the glomerulus via two distinct mechanisms, Kidney International, vol.42, issue.6, pp.1363-1371, 1992.
DOI : 10.1038/ki.1992.428

S. Matsuo, Experimental glomerulonephritis induced in rats by a lectin and its antibodies, Kidney International, vol.36, issue.6, pp.1011-1021, 1989.
DOI : 10.1038/ki.1989.295

S. S. Iskandar, J. M. Zhang, and E. Rodriguez, Nephropathy induced in a nephritis?resistant inbred mouse strain with the use of a cationized antigen, Am. J. Pathol, vol.123, pp.67-72, 1986.

P. Ronco and H. Debiec, Molecular Pathomechanisms of Membranous Nephropathy: From Heymann Nephritis to Alloimmunization, Journal of the American Society of Nephrology, vol.16, issue.5
DOI : 10.1681/ASN.2004121080

R. Stahl, E. Hoxha, and K. Fechner, PLA2R Autoantibodies and Recurrent Membranous Nephropathy after Transplantation, New England Journal of Medicine, vol.363, issue.5, pp.496-498, 2010.
DOI : 10.1056/NEJMc1003066

H. Debiec, Autoantibodies Specific for the Phospholipase A2 Receptor in Recurrent and???De Novo???Membranous Nephropathy, American Journal of Transplantation, vol.79, issue.suppl, pp.2144-2152, 2011.
DOI : 10.1111/j.1600-6143.2011.03643.x

URL : https://hal.archives-ouvertes.fr/inserm-00919176

L. East and C. M. Isacke, The mannose receptor family, Biochimica et Biophysica Acta (BBA) - General Subjects, vol.1572, issue.2-3, pp.364-386, 2002.
DOI : 10.1016/S0304-4165(02)00319-7

A. P. West, . Jr, A. B. Herr, and P. J. Bjorkman, The Chicken Yolk Sac IgY Receptor, a Functional Equivalent of the Mammalian MHC-Related Fc Receptor, Is a Phospholipase A2 Receptor Homolog, Immunity, vol.20, issue.5, pp.601-610, 2004.
DOI : 10.1016/S1074-7613(04)00113-X

O. Llorca, Extended and bent conformations of the mannose receptor family, Cellular and Molecular Life Sciences, vol.65, issue.9, pp.1302-1310, 2008.
DOI : 10.1007/s00018-007-7497-9

V. Pedchenko, Molecular Architecture of the Goodpasture Autoantigen in Anti-GBM Nephritis, New England Journal of Medicine, vol.363, issue.4, pp.343-354, 2010.
DOI : 10.1056/NEJMoa0910500

V. Pedchenko, R. Vanacore, and B. Hudson, Goodpasture??s disease: molecular architecture of the autoantigen provides clues to etiology and pathogenesis, Current Opinion in Nephrology and Hypertension, vol.20, issue.3, pp.290-296, 2011.
DOI : 10.1097/MNH.0b013e328344ff20

W. M. Bagchus, P. J. Hoedemaeker, J. T. Vos, and W. W. Bakker, Thymocytes Reacting with Heterologous Antibodies against GP 330 in Autologous Immune Complex Glomerulopathy (AICG) in the Rat.The Relation between Susceptibility for AICG and Anti-GP 330-Binding Thymocytes, Immunobiology, vol.179, issue.4-5, pp.432-444, 1989.
DOI : 10.1016/S0171-2985(89)80047-6

R. W. Vaughan, A. G. Demaine, and K. I. Welsh, A DQA1 allele is strongly associated with idiopathic membranous nephropathy, Tissue Antigens, vol.45, issue.Suppl. 16, pp.261-269, 1989.
DOI : 10.1111/j.1399-0039.1989.tb01741.x

P. A. Dyer, C. D. Short, E. A. Clarke, and N. P. Mallick, HLA antigen and gene polymorphisms and haplotypes established by family studies in membranous nephropathy, Nephrol. Dial. Transplant, vol.7, pp.42-47, 1992.

F. C. Berthoux, Immunogenetics and immunopathology of human primary membranous glomerulonephritis: HLA?A, B, DR antigens; functional activity of splenic macrophage Fc?receptors and peripheral blood T?lymphocyte subpopulations, Clin. Nephrol, vol.22, pp.15-20, 1984.

S. H. Sacks, C. Warner, R. D. Campbell, and I. Dunham, Molecular mapping of the HLA class II region in HLA?DR3 associated idiopathic membranous nephropathy, Kidney Int, pp.13-19, 1993.

Y. H. Liu, Association of phospholipase A2 receptor 1 polymorphisms with idiopathic membranous nephropathy in Chinese patients in Taiwan, Journal of Biomedical Science, vol.17, issue.1, pp.81-88, 2010.
DOI : 10.1186/1423-0127-17-81

S. Kim, Single Nucleotide Polymorphisms in the Phospholipase A2 Receptor Gene Are Associated with Genetic Susceptibility to Idiopathic Membranous Nephropathy, Nephron Clinical Practice, vol.117, issue.3, pp.253-258, 2011.
DOI : 10.1159/000320194

C. Bantis, Tumor Necrosis Factor-?? Gene G-308A Polymorphism Is a Risk Factor for the Development of Membranous Glomerulonephritis, American Journal of Nephrology, vol.26, issue.1, pp.12-15, 2006.
DOI : 10.1159/000090706

D. Thibaudin, TNFA2 and d2 alleles of the tumor necrosis factor alpha gene polymorphism are associated with onset/occurrence of idiopathic membranous nephropathy, Kidney International, vol.71, issue.5, pp.431-437, 2007.
DOI : 10.1038/sj.ki.5002054

E. Honkanen, E. Von-willebrand, A. M. Teppo, T. Törnroth, and C. Grönhagen?riska, Adhesion molecules and urinary tumor necrosis factor-?? in idiopathic membranous glomerulonephritis, Kidney International, vol.53, issue.4, pp.909-917, 1998.
DOI : 10.1111/j.1523-1755.1998.00833.x

T. J. Neale, Tumor necrosis factor?alpha is expressed by glomerular visceral epithelial cells in human membranous nephropathy, Am. J. Pathol, vol.146, pp.1444-1454, 1995.

S. Tabibzadeh, TNF-?? induces dyscohesion of epithelial cells. Association with disassembly of actin filaments, Endocrine, vol.11, issue.8, pp.549-556, 1995.
DOI : 10.1007/BF02953018

T. B. Huber and T. Benzing, The slit diaphragm: a signaling platform to regulate podocyte function, Current Opinion in Nephrology and Hypertension, vol.14, issue.3, pp.211-216, 2005.
DOI : 10.1097/01.mnh.0000165885.85803.a8

W. Y. Lo, Association between genetic polymorphisms of the NPHS1 gene and membranous glomerulonephritis in the Taiwanese population, Clinica Chimica Acta, vol.411, issue.9-10, pp.714-718, 2010.
DOI : 10.1016/j.cca.2010.02.004

C. H. Chen, Impact of plasminogen activator inhibitor-1 gene polymorphisms on primary membranous nephropathy, Nephrology Dialysis Transplantation, vol.23, issue.10, pp.3166-3173, 2008.
DOI : 10.1093/ndt/gfn258

S. Y. Chen, Association of STAT4 polymorphisms with susceptibility to primary membranous glomerulonephritis and renal failure, Clinica Chimica Acta, vol.412, issue.21-22, pp.1899-1904, 2011.
DOI : 10.1016/j.cca.2011.06.020

S. Y. Chen, C-572G polymorphism on idiopathic membranous nephropathy risk in a han chinese population, Renal Failure, vol.36, issue.10, pp.1172-1176, 2010.
DOI : 10.1046/j.1523-1755.2001.0590051983.x

W. Qin, Anti-Phospholipase A2 Receptor Antibody in Membranous Nephropathy, Journal of the American Society of Nephrology, vol.22, issue.6, pp.1137-1143, 2011.
DOI : 10.1681/ASN.2010090967

M. Knehtl, A Case of Phospholipase A2 Receptor???Positive Membranous Nephropathy Preceding Sarcoid-Associated Granulomatous Tubulointerstitial Nephritis, American Journal of Kidney Diseases, vol.57, issue.1, pp.140-143, 2011.
DOI : 10.1053/j.ajkd.2010.09.015

E. Hoxha, An immunofluorescence test for phospholipase-A2-receptor antibodies and its clinical usefulness in patients with membranous glomerulonephritis, Nephrology Dialysis Transplantation, vol.26, issue.8, pp.2526-2532, 2011.
DOI : 10.1093/ndt/gfr247

J. M. Hofstra, L. H. Beck, . Jr, D. M. Beck, J. F. Wetzels et al., Anti-Phospholipase A2 Receptor Antibodies Correlate with Clinical Status in Idiopathic Membranous Nephropathy, Clinical Journal of the American Society of Nephrology, vol.6, issue.6, pp.1286-1291, 2011.
DOI : 10.2215/CJN.07210810

L. H. Beck, &. Jr, and D. J. Salant, Membranous nephropathy: recent travels and new roads ahead, Kidney International, vol.77, issue.9, pp.765-770, 2010.
DOI : 10.1038/ki.2010.34

URL : http://doi.org/10.1038/ki.2010.34

L. H. Beck and . Jr, Rituximab-Induced Depletion of Anti-PLA2R Autoantibodies Predicts Response in Membranous Nephropathy, Journal of the American Society of Nephrology, vol.22, issue.8, pp.1543-1550, 2011.
DOI : 10.1681/ASN.2010111125

H. Debiec and P. Ronco, R Glomerular Deposits in Membranous Nephropathy, New England Journal of Medicine, vol.364, issue.7, pp.689-690, 2011.
DOI : 10.1056/NEJMc1011678

M. Zhang, Identification of the target self-antigens in reperfusion injury, The Journal of Experimental Medicine, vol.281, issue.1, pp.141-152, 2006.
DOI : 10.1593/neo.05376

O. B. Herrera, A Novel Pathway of Alloantigen Presentation by Dendritic Cells, The Journal of Immunology, vol.173, issue.8
DOI : 10.4049/jimmunol.173.8.4828

B. Terrier, Alpha-enolase: A target of antibodies in infectious and autoimmune diseases, Autoimmunity Reviews, vol.6, issue.3, pp.176-182, 2007.
DOI : 10.1016/j.autrev.2006.10.004

A. Servettaz, Identification of target antigens of antiendothelial cell antibodies in healthy individuals: A proteomic approach, PROTEOMICS, vol.1, issue.5, pp.1000-1008, 2008.
DOI : 10.1002/pmic.200700794

S. C. Jordan, B. Buckingham, R. Sakai, and D. Olson, Studies of Immune-Complex Glomerulonephritis Mediated by Human Thyroglobulin, New England Journal of Medicine, vol.304, issue.20, pp.1212-1215, 1981.
DOI : 10.1056/NEJM198105143042006

Y. Takekoshi, Free Small and IgG-Associated Large Hepatitis B e antigen in the Serum and Glomerular Capillary Walls of Two Patients with Membranous Glomerulonephritis, New England Journal of Medicine, vol.300, issue.15, pp.814-819, 1979.
DOI : 10.1056/NEJM197904123001502

W. H. Hörl and D. Kerjaschki, Membranous glomerulonephritis (MGN), J. Nephrol, vol.13, pp.291-316, 2000.

R. Kain, Molecular mimicry in pauci-immune focal necrotizing glomerulonephritis, Nature Medicine, vol.102, issue.10, pp.1088-1096, 2008.
DOI : 10.1038/nm.1874

K. Lundberg and P. J. Venables, Epitope spreading in animal models: array of hope in rheumatoid arthritis and multiple sclerosis, Arthritis Research & Therapy, vol.10, issue.6, p.122, 2008.
DOI : 10.1186/ar2544

L. Chen, A Nephritogenic Peptide Induces Intermolecular Epitope Spreading on Collagen IV in Experimental Autoimmune Glomerulonephritis, Journal of the American Society of Nephrology, vol.17, issue.11, pp.3076-3081, 2006.
DOI : 10.1681/ASN.2006070688

P. G. Tipping, A. R. Kitching, and . Glomerulonephritis, Glomerulonephritis, Th1 and Th2: what's new?, Clinical and Experimental Immunology, vol.31, issue.2, pp.207-215, 2005.
DOI : 10.1136/ard.2004.024737

A. Kuroki, M. Iyoda, T. Shibata, and T. Sugisaki, Th2 cytokines increase and stimulate B cells to produce IgG4 in idiopathic membranous nephropathy, Kidney International, vol.68, issue.1, pp.302-310, 2005.
DOI : 10.1111/j.1523-1755.2005.00415.x

J. S. Van-der-zee, P. Van-swieten, and R. C. Aalberse, Inhibition of complement activation by IgG4 antibodies, Clin. Exp. Immunol, vol.64, pp.415-422, 1986.

R. C. Aalberse and J. Schuurman, IgG4 breaking the rules, Immunology, vol.105, issue.1, pp.9-19, 2002.
DOI : 10.1038/349243a0

K. Lhotta, R. Würzner, and P. König, Glomerular deposition of mannose-binding lectin in human glomerulonephritis, Nephrology Dialysis Transplantation, vol.14, issue.4, pp.881-886, 1999.
DOI : 10.1093/ndt/14.4.881

S. T. Spicer, Induction of Passive Heymann Nephritis in Complement Component 6-Deficient PVG Rats, The Journal of Immunology, vol.179, issue.1, pp.172-178, 2007.
DOI : 10.4049/jimmunol.179.1.172

P. L. Leenaerts, B. M. Hall, B. J. Van-damme, M. R. Daha, and Y. Vanrenterghem, Active Heymann nephritis in complement component C6 deficient rats, Kidney International, vol.47, issue.6, pp.1604-1614, 1995.
DOI : 10.1038/ki.1995.224

D. Kerjaschki, Pathogenic antibodies inhibit the binding of apolipoproteins to megalin/gp330 in passive Heymann nephritis., Journal of Clinical Investigation, vol.100, issue.9, pp.2303-2309, 1997.
DOI : 10.1172/JCI119768

K. Hanasaki and H. Arita, Phospholipase A2 receptor: a regulator of biological functions of secretory phospholipase A2, Prostaglandins & Other Lipid Mediators, vol.68, issue.69, pp.68-69, 2002.
DOI : 10.1016/S0090-6980(02)00022-9

A. V. Cybulsky, T. Takano, J. Papillon, and A. J. Mctavish, Complement-induced phospholipase A2 activation in experimental membranous nephropathy1 See Editorial by Shankland, p. 1204., Kidney International, vol.57, issue.3, pp.1052-1062, 2000.
DOI : 10.1046/j.1523-1755.2000.00932.x

E. Zvaritch, G. Lambeau, and M. Lazdunski, Endocytic properties of the M?type 180?kDa receptor for secretory phospholipases A 2, J. Biol. Chem, vol.271, pp.250-257, 1996.

A. Augert, The M-type receptor PLA2R regulates senescence through the p53 pathway, EMBO reports, vol.283, issue.3, pp.271-277, 2009.
DOI : 10.1016/j.cell.2007.12.032

URL : https://hal.archives-ouvertes.fr/hal-00417702

B. Sis, Accelerated expression of senescence associated cell cycle inhibitor p16INK4A in kidneys with glomerular disease, Kidney International, vol.71, issue.3, pp.218-226, 2007.
DOI : 10.1038/sj.ki.5002039

A. L. Berg, P. Nilsson?ehle, and M. Arnadottir, Beneficial effects of ACTH on the serum lipoprotein profile and glomerular function in patients with membranous nephropathy, Kidney International, vol.56, issue.4, pp.1534-1543, 1999.
DOI : 10.1046/j.1523-1755.1999.00675.x

A. S. Bomback, Treatment of nephrotic syndrome with adrenocorticotropic hormone (ACTH) gel, Drug Design, Development and Therapy, vol.5, pp.147-153, 2011.
DOI : 10.2147/DDDT.S17521

K. G. Mountjoy, L. S. Robbins, M. T. Mortrud, and R. D. Cone, The cloning of a family of genes that encode the melanocortin receptors, Science, vol.257, issue.5074, pp.1248-1251, 1992.
DOI : 10.1126/science.1325670

A. Lindskog, Melanocortin 1 Receptor Agonists Reduce Proteinuria, Journal of the American Society of Nephrology, vol.21, issue.8, pp.1290-1298, 2010.
DOI : 10.1681/ASN.2009101025

J. L. Nortier, Neonatal disease in neutral endopeptidase alloimmunization: lessons for immunological monitoring, Pediatric Nephrology, vol.106, issue.10, pp.1399-1405, 2006.
DOI : 10.1007/s00467-006-0203-6

C. Atkinson, F. Qiao, H. Song, G. S. Gilkeson, and S. Tomlinson, Low-Dose Targeted Complement Inhibition Protects against Renal Disease and Other Manifestations of Autoimmune Disease in MRL/lpr Mice, The Journal of Immunology, vol.180, issue.2, pp.1231-1238, 2008.
DOI : 10.4049/jimmunol.180.2.1231

Y. Huang, F. Qiao, C. Atkinson, V. M. Holers, and S. Tomlinson, A Novel Targeted Inhibitor of the Alternative Pathway of Complement and Its Therapeutic Application in Ischemia/Reperfusion Injury, The Journal of Immunology, vol.181, issue.11, pp.8068-8076, 2008.
DOI : 10.4049/jimmunol.181.11.8068