A. Rahman and D. A. Isenberg, Systemic lupus erythematosus, N Engl J Med, vol.358, pp.929-968, 2008.

F. Tamirou, L. Arnaud, R. Talarico, C. A. Scirè, T. Alexander et al., Systemic lupus erythematosus: state of the art on clinical practice guidelines, RMD Open, vol.4, issue.2, 2018.

N. Danchenko, J. A. Satia, and M. S. Anthony, Epidemiology of systemic lupus erythematosus: a comparison of worldwide disease burden, Lupus, vol.15, pp.308-318, 2006.

M. J. Lewis and A. S. Jawad, The effect of ethnicity and genetic ancestry on the epidemiology, clinical features and outcome of systemic lupus erythematous, Rheumatology, vol.56, pp.67-77, 2017.

, Lupus érythémateux systémique : Protocole national de diagnostic et de soins, HAS, 2010.

H. I. Brunner, D. D. Gladman, D. Ibanez, M. D. Urowitz, and E. D. Silverman, Difference in disease features between childhood-onset and adult-onset systemic lupus erythematosus, Arthritis Rheum, vol.58, pp.556-62, 2008.

B. Dema and N. Charles, Advances in mechanisms of systemic lupus erythematosus, Discov Med, vol.17, pp.247-55, 2014.

, Campus dermatologie, 2010.

P. Lehmann, E. Hölzle, P. Kind, G. Goerz, and G. Plewig, Experimental reproduction of skin lesions in lupus erythematosus by UVA and UVB radiation, J Am Acad Dermatol, vol.22, pp.181-187, 1990.

A. Kuhn and S. Beissert, Photosensitivity in lupus erythematosus, Autoimmunity, vol.38, pp.519-529, 2005.

J. Sánchez-guerrero, A. G. Uribe, L. Jiménez-santana, M. Mestanza-peralta, P. Lara-reyes et al., A trial of contraceptive methods in women with systemic lupus erythematosus, N Engl J Med. 15 déc, vol.353, issue.24, pp.2539-2588, 2005.

A. T. Borchers, S. M. Naguwa, C. L. Keen, and M. E. Gershwin, The implications of autoimmunity and pregnancy, J Autoimmun. mai, vol.34, issue.3, pp.287-299, 2010.

A. Mathian, L. Arnaud, and Z. Amoura, Physiopathologie du lupus systémique : le point en, 2014.

S. R. Johnson, O. N. Goek, D. Singh-grewal, S. C. Vlad, B. M. Feldman et al., Classification criteria in rheumatic diseases: a review of methodologic properties, Arthritis Rheum, vol.57, pp.1119-1152, 2007.

A. S. Cohen, W. E. Reynolds, E. C. Franklin, J. P. Kulka, M. W. Ropes et al., Preliminary criteria for the classication of systemic lupus erythematosus, Bull Rheum Dis, vol.21, pp.643-651, 1971.

E. M. Tan, A. S. Cohen, J. F. Fries, A. T. Masi, M. Shane et al., The 1982 revised criteria for the classification of systemic lupus erythematosus, Arthritis Rheum, vol.25, pp.1271-1278, 1982.

M. C. Hochberg, Updating the American College of Rheumatology revised criteria for the classification of systemic lupus erythematosus, Arthritis Rheum, vol.40, p.1725, 1997.

P. Piette, Z. Amoura, E. Hachulla, J. Véronique-le-guern, and . Sibilla, , 2013.

M. Aringer, K. Costenbader, and R. Brinks, OP0020 Validation of new systemic lupus erythematosus classification criteria, Ann Rheum Dis, vol.77, issue.2, p.60, 2018.

M. Aringer and K. Costenbader, David Daikh et al ; European League Against Rheumatism/American College of Rheumatology Classification Criteria for Systemic Lupus Erythematosus, 2019.

S. Giannouli, M. Voulgarelis, P. D. Ziakas, and A. G. Tzioufas, Anaemia in systemic lupus erythematosus: from pathophysiology to clinical assessment, Ann Rheum Dis, vol.65, pp.144-152, 2006.

F. Fusio, E. M. Bohen, C. M. Yuan, and P. G. Welch, Review of thrombotic thrombocytopenic purpura in the setting of systemic lupus erythematosus, Semin Arthritis Rheum, 1998.

O. Meyer and . Lupus, Encyclopédie Médico-Chirurgicale, pp.14-244, 2004.

P. N. Malleson, M. Sailer, and M. J. Mackinnon, Silverman ED Usefulness of antinuclear antibody testing to screen for rheumatic diseases, Arch Dis Child, vol.77, pp.299-304, 1997.

A. Perdriger and E. , Dumontet Maladies auto-immunes non sépcifiques d'organe, septembre, 2014.

O. Meyer, Actualités sur les anti-SSA/Ro et anti-SSB/La, Ann Med Interne, vol.153, issue.8, pp.520-529, 2002.

E. M. Tan and H. G. Kunkel, Characteristics of a soluble nuclear antigen precipitating with sera of patients with systemic lupus erythematosus, J. Immunol, vol.96, issue.3, pp.1297-304, 1966.

M. O. Lupus-Érythémateux-systémique and . Emc--rhumatologie--orthopédie, , vol.2, pp.1-32, 2005.

M. Reinchlin and W. J. Van-venrooij, Autoantibodies to the URNP : relationship to clinical diagnosis and nephritis, Clin Exp Immunol, vol.83, pp.286-290, 1991.

W. Lloyd and P. H. Schur, Immune complexes, complement, and anti-DNA in exacerbations of systemic lupus erythematosus (SLE), Medicine, vol.60, issue.3, pp.208-225, 1981.

P. H. Schur, Complement and lupus erythematosus, Arthritis and rheumatism, vol.25, issue.7, pp.793-801, 1982.

E. N. Harris, G. R. Hughes, and A. E. Gharavi, Antiphospholipid antibodies: an elderly statesman Dons new garments, J Rheumatol Suppl, vol.14, issue.13, pp.208-221, 1987.

Z. Amoura, S. Koutouzov, H. Chabre, P. Cacoub, I. Amoura et al., Presence of antinucleosome autoantibodies in a restricted set of connective tissue diseases: antinucleosome antibodies of the IgG3 subclass are markers of renal pathogenicity in systemic lupus erythematosus, Arthritis and rheumatism, vol.43, issue.1, pp.76-84, 2000.

G. Moroni, A. Radice, G. Giammarresi, S. Quaglini, B. Gallelli et al., Are laboratory tests useful for monitoring the activity of lupus nephritis? A 6-year prospective study in a cohort of 228 patients with lupus nephritis. Annals of the rheumatic diseases, vol.68, pp.234-241, 2009.

J. A. Gómez-puerta, R. W. Burlingame, and R. Cervera, Anti-chromatin (anti-nucleosome) antibodies: diagnostic and clinical value, Autoimmun. Rev, vol.7, issue.8, pp.606-611, 2008.

A. Verlemyr and L. Truedsson, Lillemor Skattum ; Analysis of Anti-C1q Autoantibodies by Western Blot, Lennart Truedsson, 2018.

G. Mihaylova, . Stoyanov, . Vasilev, and . Vasil, Tzaritza Ioanna -ISUL" Ivanova, Desislava; Radanova, Maria; Autoantibodies against components of the complement complex C1 in patients with lupus nephritis, 2020.

C. Johanet, C. Andre, J. Sibilia, A. Baquey, F. Oksman et al., Signification clinique des anticorps antiribosomes, Rev Med Interne, vol.21, pp.510-516, 2000.

C. Bombardier, D. D. Gladman, and M. B. Urowitz, Derivation of the SLEDAI. A disease activity index for lupus patients. The Committee on Prognosis Studies in SLE, Arthritis Rheum, vol.35, issue.6, p.40, 1992.

C. Yee, L. Teh, and C. Gordon, Assessment of disease activity and quality of life in systemic lupus erythematosus New aspects

D. Gladman, E. Ginzler, C. Goldsmith, P. Fortin, M. Liang et al., The development and initial validation of the Systemic Lupus International Collaborating Clinics/American College of Rheumatology damage index for systemic lupus erythematosus, Arthritis Rheum, vol.39, pp.363-372, 1996.

A. Karras, Atteinte rénale du lupus érythémateux disséminé Presse Médicale, vol.41, pp.260-266, 2012.

D. Yap, C. Tang, M. Ma, M. F. Lam, and T. M. Chan, Survival analysis and causes of mortality in patients with lupus nephritis, Août, vol.27, issue.8, pp.3248-54, 2012.

E. Johnson, C. Gordon, F. D. Hobbs, and P. A. Bacon, Undiagnosed systemic lupus erythematosus in the community, Lancet, vol.347, pp.367-376, 1996.

C. C. Mok, Towards new avenues in the management of lupus glomerulonephritis, Nat Rev Rheumatol, vol.12, pp.221-255, 2016.

J. S. Cameron and . Lupus, J Am Soc Nephrol, vol.10, issue.2, pp.413-424, 1999.

M. Mannik, C. E. Merrill, L. D. Stamps, and M. H. Wener, Multiple auto antibodies form the glomerular immune deposits in patients with systemic lupus erythematosus, J Rheumatol, vol.30, pp.1495-504, 2003.

A. Hedberg and E. S. Mortensen, Rekvig OP Chromatin as a target antigen in human and murine lupus nephritis, Arthritis Res Ther, vol.13, p.214, 2011.

P. Niaudet, Signes cliniques et biologiques des néphropathies glomérulaires, EMC Pédiatrie, vol.2, pp.12-30, 2005.

A. Karras, La néphropathie lupique : le point en, Rev Med Interne, vol.36, issue.2, pp.98-106, 2014.

J. S. Cameron and . Lupus, J Am Soc Nephrol, vol.10, pp.413-437, 1999.

S. Bajaj, L. Albert, and D. D. Gladman, Serial renal biopsy in systemic lupus erythematosus, J Rheumatol, vol.27, p.2822, 2000.

H. Vanquaethem,

E. Renaudineau, Y. L. Renaudineau, A. Meur, P. Chauveau, and . Youinou, Intérêt des nouveaux examens sérologiques pour la néphropathie lupique. Immunoanalyse et biologie spécialisée, vol.23, pp.137-142, 2008.

J. Weening, D. 'agativd, and M. M. Schwartz, The Classification of Glomerulonephritis in Systemic Lupus Erythematosus Revisited, J. Am. Soc. Nephrol, vol.15, pp.241-250, 2004.

I. M. Bajema, Revision of the International Society of Nephrology/Renal Pathology Society classification for lupus nephritis: clarification of definitions, and modified National Institutes of Health activity and chronicity indices, Kidney International, vol.93, pp.789-796, 2018.

H. Hôpital and . Lyon, , 2009.

M. M. Schwartz, S. M. Korbet, and E. J. Lewis, The prognosis and pathogenesis of severe lupus glomerulonephritis. Collaborative Study Group Nephrol Dial Transplant, vol.23, pp.1298-306, 2008.

L. Bono, J. S. Cameron, and J. A. Hicks, The very long-term prognosis and complications of lupus nephritis and its treatment, QJM Mon. J. Assoc. Physicians, vol.92, pp.211-218, 1999.

, Kidney int, vol.65, pp.521-551, 2004.

A. Karras and F. Martinez, Revue du Rhumatisme, vol.72, pp.162-167, 2005.

H. A. Austin, L. R. Muenz, K. M. Joyce, T. T. Antonovych, and J. E. Balow, Diffuse proliferative lupus nephritis: identification of specific pathologic features affecting renal outcome, Kidney Int, vol.25, issue.4, pp.689-695, 1984.

Z. Xuejing, T. Jiazhen, L. Jun, X. Xiangqing, Y. Shuguang et al., Urinary TWEAK Level as a Marker of Lupus Nephritis Activity in 46 Cases, 2012.

L. M. Ortega, D. R. Schultz, and O. Lenz, Review: Lupus nephritis: pathologic features, epidemiology and a guide to therapeutic decisions, Lupus, vol.19, p.557, 2010.

A. J. Howie, N. Turhan, and D. Adu, Powerful morphometric indicator of prognosis in lupus nephritis, Q J Med, vol.96, pp.411-420, 2003.

R. S. Flanc, M. A. Roberts, G. F. Strippoli, S. J. Chadban, P. G. Kerr et al., Treatment for lupus nephritis, Cochrane Database Syst Rev.CD002922, 2004.

R. P. Sloan, M. M. Schwartz, and S. M. Korbet, Long-term outcome in systemic lupus erythematosus membranous glomerulonephritis. Lupus Nephritis Collaborative Study Group, J. Am. Soc. Nephrol, vol.7, pp.299-305, 1996.

M. El-hachmi, M. Jadoul, C. Lefebvre, G. Depresseux, and F. A. Houssiau, Relapses of lupus nephritis : incidence, risk factors, serology and impact on outcome, vol.12, pp.692-698, 2003.

G. K. Bertsias, M. Tektonidou, and Z. Amoura, Joint European League Against Rheumatism and European Renal Association-European Dialysis and Transplant Association (EULAR/ERA-EDTA) recommendations for the management of adult and paediatric lupus nephritis, Ann Rheum Dis, vol.71, pp.1771-1782, 2012.

, Kidney Disease: Improving Global Outcomes (KDIGO) Glomerulonephritis Work Group. KDIGO clinical practice guideline for glomerulonephritis, Kidney Int Suppl, vol.2, pp.139-274, 2012.

E. Masson, La néphropathie lupique : le point en, 2014.

. Em-consulte,

P. Niaudet, Traitement de la néphropathie du lupus érythémateux disséminé chez l'enfant, Arch Pédiatr, vol.6, pp.1312-1333, 1999.

M. B. Condon, D. Ashby, R. J. Pepper, H. T. Cook, J. B. Levy et al., Prospective observational single-centre cohort study to evaluate the effectiveness oftreating lupus nephritis with rituximab and mycophenolate mofetil but no oral steroids, Ann Rheum Dis, vol.72, pp.1280-1286, 2013.

L. Beck, A. S. Bomback, and M. J. Choi, KDOQI US commentary on the 2012 KDIGO clinical practice guideline for glomerulonephritis, Am. J. Kidney Dis, vol.62, pp.403-441, 2013.

A. J. Sabucedo and G. Contreras, ESKD, transplantation, and dialysis in lupus nephritis, Semin Nephrol, vol.35, pp.500-508, 2015.

E. Daugas, D. Nochy, D. L. Huong, P. Duhaut, H. Beaufils et al., Antiphospholipid syndrome nephropathy in systemic lupus erythematosus, J AmSoc Nephrol, vol.13, pp.42-52, 2002.

G. G. Illei, E. Tackey, L. Lapteva, and P. E. Lipsky, Biomarkers in systemic lupus erythematosus. I. General overview of biomarkers and their applicability, Arthritis Rheum, vol.50, pp.1709-1720, 2004.

C. C. Liu, S. Manzi, and J. M. Ahearn, Biomarkers for systemic lupus erythematosus: A review and perspective, Curr Opin Rheumatol, vol.17, pp.543-549, 2005.

M. Lech and H. J. Anders, The pathogenesis of lupus nephritis, J Am Soc Nephrol, vol.24, pp.1357-66, 2013.

H. I. Brunner, M. R. Bennett, R. Mina, M. Suzuki, M. Petri et al., Association of non invasively measured renal protein biomarkers with histologic features of lupus nephritis, Arthritis Rheum, vol.64, pp.2687-2697, 2012.

S. M. Hewitt, J. Dear, and R. A. Star, Discovery of protein biomarkers for renal diseases, J Am Soc Nephrol, vol.15, pp.1677-1689, 2004.

C. C. Mok, Biomarkers for lupus nephritis: a critical appraisal, J. Biomed. Biotechnol, p.638413, 2010.

S. Soliman and C. Mohan, Lupus nephritis biomarkers, Clin Immunol, vol.185, pp.10-20, 2017.

C. C. Mok, Biomarkers for lupus nephritis: A critical appraisal, J Biomed Biotechnol, p.638413, 2010.

G. Contreras, V. Pardo, B. Leclercq, O. Lenz, and P. , Sequential therapies for proliferative lupus nephritis, N Engl J Med, vol.350, pp.971-980, 2004.

. Vasilv, . Vasilev, . Remi, M. Noe, S. Valentinj et al., Functional Characterization of Autoantibodies against Complement Component C3 in Patients with Lupus Nephritis Roumenina, 2015.

R. Satu-sinikka-pesickova, M. Rysava, L. Lenicek, E. Vitek, Z. Potlukova et al., Jakub Zavada, Marten Trendelenburg and Vladimir Tesar ;Prognostic value of anti-CRP antibodies in lupus nephritis in long-term follow-up, 2015.

M. Plawecki, E. Lheritier, G. Clavarino, N. Jourde-chiche, S. Ouili et al., Association between the Presence of Autoantibodies Targeting Ficolin-3 and Active Nephritis in Patients with Systemic Lupus Erythematosus, PloS One, vol.11, issue.9, p.160879, 2016.
URL : https://hal.archives-ouvertes.fr/hal-01371323

N. S. Merle, R. Noe, L. Halbwachs-mecarelli, V. Fremeaux-bacchi, and L. Roumenina, Complement system part II: role in immunity, vol.6, p.257, 2015.
URL : https://hal.archives-ouvertes.fr/hal-01216398

P. J. Lachmann, The amplification loop of the complement pathways, Adv. Immunol, vol.104, pp.115-149, 2009.

V. V. Vasilev, R. Noe, M. Dragon-durey, S. Chauvet, V. J. Lazarov et al., Functional characterization of autoantibodies against complement component C3 in patients with lupus nephritis, J Biol Chem, vol.290, issue.42, pp.25343-55, 2015.

J. L. Ngu and K. Blackett, Complement and immunoconglutinin changes in the nephrotic syndrome of adult Africans, J. Trop. Med. Hyg, vol.73, pp.250-254, 1970.

B. J. Potter, D. J. Brown, A. Watson, and D. P. Jewell, Complement inhibitors and immunoconglutinins in ulcerative colitis and Crohn's disease, Gut, vol.21, pp.1030-1034, 1980.

D. J. Birmingham, J. E. Bitter, E. G. Ndukwe, S. Dials, T. R. Gullo et al., Relationship of circulating anti-C3b and anti-C1q IgG to lupus nephritis and its flare, Clin J Am Soc Nephrol, vol.11, issue.1, pp.47-53, 2016.

J. V. Bertouch, P. J. Roberts-thompson, P. H. Feng, and J. Bradley, C-reactive protein and serological indices of disease activity in systemic lupus erythematosus, Ann Rheum Dis, vol.42, pp.655-663, 1983.

S. A. Bell, H. Faust, A. Schmid, and M. Meurer, Autoantibodies to C-reactive protein (CRP) and other acute-phase proteins in systemic autoimmune diseases, Clin Exp Immunol, vol.113, pp.327-359, 1998.

M. Minatani, S. Aotsuka, and T. Satoh, Autoantibodies against C-reactive protein (CRP) in sera of patients with systemic rheumatic diseases, Mod Rheumatol, vol.11, pp.127-158, 2001.

W. J. Jabs, B. A. Lögering, and P. Gerke, The kidney as a second site of human C-reactive protein formation in vivo, Eur. J. Immunol, vol.33, pp.152-161, 2003.

R. Satu-sinikka-pesickova, M. Rysava, L. Lenicek, E. Vitek, Z. Potlukova et al.,

, Prognostic value of anti-CRP antibodies in lupus nephritis in long-term follow-up, 2015.

Y. Yae, S. Inaba, H. Sato, K. Okochi, F. Tokunaga et al., Isolation and characterization of a thermolabile beta-2macroglycoprotein('thermolabile substance'or"Hakataantigen")detected by precipitating (auto)antibody in sera of patients with systemic lupus erythematosus, Biochim Biophys Acta, vol.1078, pp.369-376, 1991.

T. Andersen, L. Munthe-fog, P. Garred, and S. Jacobsen, Serum levels of ficolin-3 (Hakata antigen) in patients with systemic lupus erythematosus, J Rheumatol, vol.36, issue.4, pp.757-766, 2009.

E. Hein, L. A. Nielsen, C. T. Nielsen, L. Munthe-fog, M. Skjoedt et al., Ficolins and the lectin pathway of complement in patients with systemic lupus erythematosus, Mol Immunol, vol.63, issue.2, pp.209-223, 2015.

M. Plawecki, E. Lheritier, G. Clavarino, N. Ouili, S. Paul et al., Chantal Dumestre-Pérard ; Association between the Presence of Autoantibodies Targeting Ficolin-3and Active Nephritis in Patients with Systemic Lupus Erythematosus, 2016.

S. Colliard, N. Jourde-chiche, G. Clavarino, F. Sarrot-reynauld, E. Gout et al., Chantal Dumestre-Pérard, 2018.

M. Samson, F. Aubry, and M. Parmentier,

, Medecine/sciences, 1999.

O. Kulkarni and H. J. Anders, Chemokines in lupus nephritis, Front. Biosci, vol.13, pp.3312-3320, 2008.

G. Elia, Interferon-?-inducible chemokines in systemic lupus erythematosus, Clin. Ter, vol.166, issue.1, 2015.

J. W. Bauer, E. C. Baechler, M. Petri, F. M. Batliwalla, D. Crawford et al., Elevated serum levels of interferon-regulated chemokines are biomarkers for active human systemic lupus erythematosus, PLoS Med, vol.3, 2006.

L. M. Vila, M. J. Molina, A. M. Mayor, J. J. Cruz, E. Rios-olivares et al., Association of serum MIP-1 alpha, MIP-1 beta, and RANTES with clinical manifestations, disease activity, and damage accrual in systemic lupus erythematosus, Clin Rheumatol, vol.26, 2007.

Q. Fu, X. Chen, H. Cui, Y. Guo, J. Chen et al.,

, Association of elevated transcript levels of interferon-inducible chemokines with disease activity and organ damage in systemic lupus erythematosus patients, 2008.

A. Sang, Y. Zheng, and L. Morel, Contributions of B cells to lupus pathogenesis, Molecular Immunology, vol.62, issue.2, pp.329-338, 2014.

W. L. De-ning-he, K. Chen, X. Long, G. Zhang, and . Dong, Association of Serum CXCL13 with Intrarenal Ectopic Lymphoid Tissue Formation in Lupus Nephritis, 2016.

S. Bugatti, A. Manzo, and F. Benaglio, Serum levels of CXCL13 are associated with ultrasonographic synovitis and predict power Doppler persistence in early rheumatoid arthritis treated with non-biological disease-modifying anti-rheumatic drugs, Arthritis Res Ther, vol.14, p.34, 2012.

J. M. Kramer, E. Klimatcheva, and T. L. Rothstein, CXCL13 is elevated in Sjogren's syndrome in mice and humans and is implicated in disease pathogenesis, J Leukoc Biol, vol.94, pp.1079-89, 2013.

L. J. Vuga, J. R. Tedrow, and K. V. Pandit, C-X-C motif chemokine 13 (CXCL13) is a prognostic biomarker of idiopathic pulmonary fibrosis, Am J Respir Crit Care Med, vol.189, pp.966-74, 2014.

S. L. Gaffen, R. Jain, A. V. Garg, and D. J. Cua, The IL-23-IL-17 immune axis: from mechanisms to therapeutic testing, Nat Rev Immunol, vol.14, issue.9, pp.585-600, 2014.

C. K. Wong, C. Y. Ho, E. K. Li, and C. W. Lam, Elevation of proinflammatory cytokine (IL-18, IL-17, IL-12) and Th2 cytokine (IL-4) concentrations in patients with systemic lupus erythematosus, Lupus, vol.9, issue.8, pp.589-93, 2000.

Z. Zhang, V. C. Kyttaris, and G. C. Tsokos, The role of IL-23/IL-17 axis in lupus nephritis, J Immunol, vol.183, issue.5, pp.3160-3169, 2009.

D. Y. Chen, Y. M. Chen, M. C. Wen, T. Y. Hsieh, W. T. Hung et al., The potential role of Th17 cells and Th17-related cytokines in the pathogenesis of lupus nephritis, vol.21, pp.1385-96, 2012.

H. Dedong, Z. Feiyan, S. Jie, L. Xiaowei, and W. Shaoyang, Analysis of interleukin-17 and interleukin-23 for estimating disease activity and predicting the response to, 2019.

L. Gorelik, K. Gilbride, M. Dobles, S. L. Kalled, D. Zandman et al., Normal B cell homeostasis requires B cell activation factor production by radiation-resistant cells, J Exp Med, vol.198, pp.937-982, 2003.

J. V. Stein, M. Lopez-fraga, and F. A. Elustondo, APRIL modulates B and T cell immunity, J Clin Invest, vol.109, pp.1587-98, 2002.

J. Zhang, V. Roschke, and K. P. Baker, Cutting edge: a role for B lymphocyte stimulator in systemic lupus erythematosus, J Immunol, vol.166, pp.6-10, 2001.

M. Petri, W. Stohl, and W. Chatham, Association of plasma B lymphocyte stimulator levels and disease activity in systemic lupus erythematosus, Arthritis Rheum, vol.58, pp.2453-2462, 2008.

F. B. Vincent, M. Northcott, and A. Hoi, Association of serum B cell activating factor from the tumour necrosis factor family (BAFF) and a proliferation-inducing ligand (APRIL) with central nervous system and renal disease in systemic lupus erythematosus, Lupus, vol.22, pp.873-84, 2013.

I. Parodis, A. Zickert, B. Sundelin, M. Axelsson, and J. Gerhardsson, Elisabet Svenungsson, Vivianne Malmström, and Iva Gunnarsson ; Evaluation of B lymphocyte stimulator and a proliferation inducing ligand as candidate biomarkers in lupus nephritis based on clinical and histopathological outcome following induction therapy, 2015.

Z. Adhya, S. Borozdenkova, and M. Y. Karim, The role of cytokines as biomarkers in systemic lupus erythematosus and lupus nephritis, Nephrol. Dial. Transplant, vol.26, issue.10, pp.3273-3280, 2011.

A. Sabry, H. Sheashaa, A. El-husseini, K. El-dahshan, M. Abdel-rahim et al., Intercellular adhesion molecules in systemic lupus erythematosus patients with lupus nephritis, Clin. Rheumatol, vol.26, issue.11, pp.1819-1823, 2007.

P. E. Spronk, H. Bootsma, M. G. Huitema, P. C. Limburg, and C. G. Kallenberg, Levels of soluble VCAM-1, soluble ICAM-1, and soluble E-selectin during disease exacerbations in patients with systemic lupus erythematosus (SLE); a long term prospective study, Clin Exp Immunol, vol.97, pp.439-444, 1994.

N. Aljaberi, M. Bennett, and H. I. Brunner-&-prasad-devarajan, , 2019.

C. Piubelli, M. Galvani, and M. Hamdan, Proteome analysis of rat polymorphonuclear leukocytes: a two-dimensional electrophoresis/mass spectrometry approach, Electrophoresis, vol.23, pp.298-310, 2002.

M. A. Knepper, Porteomics and the kidney, J Am Soc Nephrol, vol.13, pp.1398-1408, 2002.

J. Solassol, N. Boulle, T. Maudelonde, and A. Mangé, Protéomique clinique : vers la détection précoce des cancers ?, 2005.

C. C. Mok, H. H. Ding, M. Kharboutli, and C. Mohan, Axl, Ferritin, IGFBP2 and TNFR2 as biomarkers in systemic lupus erythematosus, Arthritis Care Res, 2016.

G. Zizzo, B. A. Hilliard, M. Monestier, and P. L. Cohen, Efficient clearance of early apoptotic cells by human macrophages requires M2c polarization and MerTK induction, J Immunol, vol.189, pp.3508-3528, 2012.

J. Rothe, G. Gehr, H. Loetscher, and W. Lesslauer, Tumor necrosis factor receptors-structure and function, Immunol Res, vol.11, pp.81-90, 1992.

C. F. Ware, P. D. Crowe, T. L. Vanarsdale, J. L. Andrews, M. H. Grayson et al., Tumor necrosis factor (TNF) receptor expression in T lymphocytes. Differential regulation of the type I TNF receptor during activation of resting and effector T cells, J. Immunol, vol.147, issue.12, pp.4229-4238, 1991.

M. E. Munroe, E. S. Vista, J. M. Guthridge, L. F. Thompson, J. T. Merrill et al., Proinflammatory adaptive cytokine and shed tumor necrosis factor receptor levels are elevated preceding systemic lupus erythematosus disease flare, Arthritis Rheumatol, vol.66, pp.1888-99, 2014.

T. Wu, H. Ding, J. Han, C. Arriens, C. Wei et al.,

, Antibody-Array-Based Proteomic Screening of Serum Markers in Systemic Lupus Erythematosus: A Discovery Study, 2016.

P. Grellier, M. Sabbah, and B. Fouqueray, Characterization of insulin-like growth factor binding proteins and regulation of IGFBP3 in human mesangial cells, Kidney Int, vol.49, pp.1071-1079, 1996.

S. M. Feld and R. Hirschberg, Insulin-like growth factor-I and insulinlike growth factor-binding proteins in the nephrotic syndrome, Pediatr Nephrol, vol.10, pp.355-363, 1996.

R. P. Narayanan, B. Fu, and A. H. Heald, IGFBP2 is a biomarker for predicting longitudinal deterioration in renal function in type 2 diabetes, Endocr Connect, vol.1, pp.95-102, 2012.

L. Christopher-stine, M. Siedner, and J. Lin, Renal biopsy in lupus patients with low levels of proteinuria, J Rheumatol, vol.34, pp.332-337, 2007.

M. W. Carr, S. J. Roth, E. Luther, S. S. Rose, and T. A. Springer, Monocyte chemoattractant protein 1 acts as a Tlymphocyte chemoattractant, Proc Natl Acad Sci, vol.91, pp.3652-3658, 1994.

Y. H. Lee and G. G. Song, Urinary MCP-1 as a biomarker for lupus nephritis: a meta-analysis, Z Rheumatol, vol.76, pp.357-363, 2017.

M. Noris, S. Bernasconi, and F. Casiraghi, Monocyte chemoattractant protein-1 is excreted in excessive amounts in the urine of patients with lupus nephritis, Lab Invest, vol.73, pp.804-809, 1995.

B. H. Rovin, H. Song, D. J. Birmingham, L. A. Hebert, C. Yu et al., Urine chemokines as biomarkers of human systemic lupus erythematosus activity, Jam Soc Nephrol, vol.16, pp.467-473, 2005.

M. J. Kim and F. W. Tam, Urinary monocyte chemoattractant protein-1 in renal disease, Clin Chim Acta, vol.412, pp.2022-2052, 2011.

L. F. Neville, G. Mathiak, and O. Bagasra, The immunobiology of interferon-gamma inducible protein 10 kD (IP-10): a novel, pleiotropic member of the C-X-C chemokine superfamily, Cytokine Growth Factor Rev, vol.8, pp.207-219, 1997.

Y. Avihingsanon, B. Phumesinp, and . Tetal, Measurement of urinary chemokine and growth factor messenger RNAs: a noninvasive monitoring in lupus nephritis, Kidney Int, vol.69, pp.747-753, 2006.

P. Romagnani, E. Lazzeri, L. Lasagni, C. Mavilia, C. Beltrame et al., IP-10 and Mig production by glomerular cells in human proliferative glomerulonephritis and regulation by nitric oxide, J. Am. Soc. Nephrol, vol.13, issue.1, pp.53-64, 2002.

P. Enghard, J. Y. Humrich, B. Rudolph, S. Rosenberger, R. Biesen et al., CXCR3+CD4+ T cells are enriched in inflamed kidneys and urine and provide a new biomarker for acute nephritis flares in systemic lupus erythematosus patients, Arthritis Rheum, vol.60, issue.1, pp.199-206, 2009.

D. D. Taub and J. J. Oppenheim, Chemokines, inflammation and the immune system, Ther. Immunol, vol.1, issue.4, pp.229-246, 1994.

K. J. Moore, T. Wada, S. D. Barbee, and V. R. Kelley, Gene Transfer of RANTES Elicits Autoimmune Renal Injury in MRL-Fas(1pr) Mice (1pr), Kidney International, vol.53, pp.1631-1672, 1998.

S. Tian, J. Li, L. Wang, T. Liu, H. Liu et al., Urinary levels of RANTES and M-CSF are predictors of lupus nephritis flare, vol.56, pp.304-310, 2007.

R. W. Chan, F. M. Lai, E. K. Li, L. S. Tam, K. M. Chow et al., Messenger RNA expression of RANTES in the urinary sediment of patients with lupus nephritis, Nephrol. (Carlton), vol.11, issue.3, pp.219-225, 2006.

O. Dienz and M. Rincon, The effects of IL-6 on CD4 T cell responses, Clin Immunol, vol.130, pp.27-33, 2009.

M. Okada, M. Kitahara, S. Kishimoto, T. Matsuda, T. Hirano et al., IL-6/BSF-2 functions as a killer helper factor in the in vitro induction of cytotoxic T cells, J Immunol, vol.141, pp.1543-1549, 1988.

M. Linker-israeli, R. J. Deans, D. J. Wallace, J. Prehn, T. Ozeri-chen et al., Elevated levels of endogenous IL-6 in systemic lupus erythematosus. A putative role in pathogenesis, J. Immunol, vol.147, issue.1, pp.117-123, 1991.

R. Herrera-esparza, O. Barbosa-cisneros, R. Villalobos-hurtado, and E. Avalos-diaz, Renal expression of IL-6 and TNFalpha genes in lupus nephritis, vol.7, pp.154-162, 1998.

L. C. Burkly, J. S. Michaelson, K. Hahm, A. Jakubowski, and T. S. Zheng, TWEAKing tissue remodeling by a multifunctional cytokine: Role of TWEAK/Fn14 pathway in health and disease

S. R. Wiley, L. Cassiano, and T. Lofton, A novel TNF receptor family member binds TWEAK and is implicated in angiogenesis, Immunity, vol.15, pp.837-883, 2001.

H. X. Gao, S. R. Campbell, L. C. Burkly, A. Jakubowski, I. Jarchum et al., TNF like weak inducer of apoptosis (TWEAK) induces inflammatory and proliferative effects inhuman kidney cells, Cytokines, vol.46, pp.24-35, 2009.

N. Schwartz, T. Rubinstein, L. C. Burkly, and C. Putterman, Urinary TWEAK as a biomarker of lupus nephritis: a multicenter cohort study, Arthritis Res Ther, vol.11, p.143, 2009.

N. Schwartz, L. Su, C. Linda, M. Burkly, C. Mackay et al.,

, Urinary TWEAK and the activity of lupus nephritis, 2007.

, Atiporn Ingsathit, Nanticha Kamanamool, Vuddhidej Ophascharoensuk, Vasant Sumethakul, Yingyos Avihingsanon; Urine TWEAK level as a biomarker for early response to treatment in active lupus nephritis: a prospective multicentre study, 2018.

. Diez-jj and . Iglesias-p, The role of the novel adipocyte-derived hormone adiponectin in human disease, vol.148, pp.293-300, 2003.

B. H. Rovin, H. Song, L. A. Hebert, T. Nadasdy, G. Nadasdy et al., Plasma, urine, and renal expression of adiponectin in human systemic lupus erythematosus, Kidney Int, vol.68, issue.4, pp.1825-1833, 2005.

B. H. Rovin and H. Song, Chemokine induction by the adipocyte-derived cytokine adiponectin, Clin. Immunol, vol.120, issue.1, pp.99-105, 2006.

. Zoccali-c, . Mallmaci-f, and . Tripepi-g, Adiponectin, metabolic risk factors, and cardiovascular events among patients with endstage renal disease, JAmSoc Nephrol, vol.13, pp.134-141, 2002.

L. Maryam, H. Anousheh, B. Behrooz, A. Yousef, T. Maryam et al., Association between urinary adiponectin level and renal involvement in systemic lupus erythematous, 2016.

B. H. Rovin, H. Song, and L. A. Hebert, Plasma, urine, and renal expression of adiponectin in human systemic lupus erythematosus, Kidney Int, vol.68, issue.4, pp.1825-1858, 2005.

J. Chudek, M. Adamczak, and H. Karkozka, Plasma adiponectin concentration before and after successful kidney transplantation, Transplant Proc, vol.35, pp.2186-2189, 2003.

, Urinary angiostatin, CXCL4 and VCAM-1 as biomarkers of lupus nephritis, 2018.

T. Wu, C. Xie, and H. W. Wang, Elevated urinary VCAM-1, P-selectin, soluble TNF receptor-1, and CXC chemokine ligand 16 in multiple murine lupus strains and human lupus nephritis, J Immunol, vol.179, issue.10, pp.7166-7175, 2007.

Y. Molad, E. Miroshnik, J. Sulkes, S. Pitlik, and A. Weinberger, Monselise YUrinary soluble VCAM-1 in systemic lupus erythematosus: a clinical marker for monitoring disease activity and damage

, Clin Exp Rheumatol, vol.20, pp.403-409, 2002.

M. I. Abd-elkareem, A. Tamimy, H. M. Khamis, O. A. Abdellatif, S. S. Hussein et al., Increased urinary levels of the leukocyte adhesion molecules ICAM-1 and VCAM-1 in human lupus nephritis with advanced renal histological changes: preliminary findings, Clin Exp Nephrol, vol.14, pp.548-57, 2010.

P. Arrizabalaga, C. Solé, G. Iglesias, and . Escaramís, C Ascaso Renal expression of ICAM-1 and VCAM-1 in ANCA-associated glomerulonephritis--are there differences among serologic subgroups, 2006.

J. M. Lorenzen, H. Haller, and T. Thum, MicroRNAs as mediators and therapeutic targets in chronic kidney disease, Nat Rev Nephrol, vol.7, pp.286-294, 2011.

K. Chandrasekaran, D. S. Karolina, S. Sepramaniam, A. Armugam, E. M. Wintour et al., Role of microRNAs in kidney homeostasis and disease, Kidney Int, vol.81, pp.617-627, 2012.

V. Patel, L. Noureddine, F. A. Micrornas, V. Benes, J. A. Garson et al., The MIQE Guidelines: Minimum Information for Publication of Quantitative Real-Time PCR Experiments, Curr Opin Nephrol Hypertens, vol.21, pp.611-622, 2009.

P. Costa-reis, P. A. Russo, Z. Zhang, L. Colonna, K. Maurer et al., The Role of MicroRNAs and Human Epidermal Growth Factor Receptor 2 in Proliferative Lupus Nephritis, 2015.

J. Guan, G. Wang, L. S. Tam, B. C. Kwan, E. K. Li et al., Urinary sediment ICAM-1 level in lupus nephritis, Clin. Chim. Acta, vol.436, issue.11, pp.1190-1195, 2012.

G. Wang, L. S. Tam, E. K. Li, B. C. Kwan, K. M. Chow et al., Serum and urinary free microRNA level in patients with systemic lupus erythematosus, Lupus, vol.20, issue.5, pp.493-500, 2011.

E. Navarro-quiroz, L. Pacheco-lugo, H. Lorenzi, Y. Díaz-olmos, L. Almendrales et al., Highthroughput sequencing reveals circulating miRNAs as potential biomarkers of kidney damage in patients with systemic lupus erythematosus, PLoS ONE, vol.11, p.166202, 2016.

H. Kaga, A. Komatsuda, A. Omokawa, M. Ito, K. Teshima et al., Downregulated expression of miR-155, miR-17, and miR-181b, and upregulated expression of activation-induced cytidine deaminase and interferon-? in PBMCs from patients with SLE, Mod Rheumatol, vol.25, pp.865-70, 2015.

E. Oeyen, K. Van-mol, G. Baggerman, H. Willems, K. Boonen et al., Ultrafiltration and size exclusion chromatography combined with asymmetrical-flow field-flow fractionation for the isolation and characterisation of extracellular vesicles from urine, J Extracell Vesicles, vol.7, p.1490143, 2018.

Y. Yang, Y. Hong, E. Cho, G. B. Kim, and I. S. Kim, Extracellular vesicles as a platform for membraneassociated therapeutic protein delivery, J Extracell Vesicles, vol.7, p.1440131, 2018.

M. Krause, A. Rak-raszewska, F. Naillat, U. Searela, C. Schmidt et al., Exosomes as secondary inductive signals involved in kidney organogenesis, J Extracell Vesicles, vol.7, p.1422675, 2018.

B. W. Van-balkom, T. Pisitkun, M. C. Verhaar, and M. A. Knepper, Exosomes and the kidney: prospects for diagnosis and therapy of renal diseases, Kidney Int, vol.80, pp.1138-1183, 2011.

D. Y. Fang, H. W. King, J. Y. Li, and J. M. Gleadle, Exosomes and the kidney: blaming the messenger, Nephrology (Carlton), vol.18, pp.1-10, 2013.

E. Oeyen, K. Van-mol, G. Baggerman, H. Willems, K. Boonen et al., Ultrafiltration and size exclusion chromatography combined with asymmetrical-flow field-flow fractionation for the isolation and characterisation of extracellular vesicles from urine, J Extracell Vesicles, vol.7, p.1490143, 2018.

Y. Yang, Y. Hong, E. Cho, G. B. Kim, and I. S. Kim, Extracellular vesicles as a platform for membraneassociated therapeutic protein delivery, J Extracell Vesicles, vol.7, p.1440131, 2018.

C. Solé, J. Cortés-hernández, M. L. Felip, M. Vidal, and J. Ordi-ros,

, miR-29c in urinary exosomes as predictor of early renal fibrosis in lupus nephritis, 2015.

Y. Li, X. Xu, X. Tang, X. Bian, B. Shen et al., Zhiwen Chen and Keqin Zhang ; MicroRNA expression profile of urinary exosomes in Type IV lupus nephritis complicated by cellular crescent, 2018.

J. Reyes-thomas, I. Blanco, and C. Putterman, Urinary biomarkers in lupus nephritis, Clin Rev Allergy Immunol, vol.40, pp.138-50, 2011.

J. Oates, S. Varghese, and A. M. Bland, Prediction of urinary protein markers in lupus nephritis, Kidney Int, vol.68, issue.6, pp.2588-2592, 2005.

K. Mosley, F. W. Tam, R. J. Edwards, J. Crozier, C. D. Pusey et al., Urinary proteomic profiles distinguish between active and inactive lupus nephritis, Rheumatology, vol.45, issue.12, pp.1497-1504, 2006.

P. Somparn, N. Hirankarn, and A. Leelahavanichkul, Urinary proteomics revealed prostaglandin H(2)D-isomerase, not Zn-alpha2glycoprotein, as a biomarker for active lupus nephritis, J Proteomics, vol.75, issue.11, pp.3240-3247, 2012.

X. Zhang, M. Jin, and H. Wu, Biomarkers of lupus nephritis determined by serial urine proteomics, Kidney Int, vol.74, pp.799-807, 2008.

P. Lee, H. Peng, T. Gelbart, L. Wang, and E. Beutler, Regulation of hepcidin transcription by interleukin-1 and interleukin-6, Proceedings of the National Academy of Sciences of the United States of America, vol.102, pp.1906-1910, 2005.

A. Aggarwal, R. Gupta, and V. S. Negi, Urinary haptoglobin, alpha-1 anti-chymotrypsin and retinol binding protein identified by proteomics as potential biomarkers for lupus nephritis, Clin Exp Immunol, vol.188, issue.2, pp.254-262, 2017.

A. G. Norden, M. Lapsley, and R. J. Unwin, Urine retinol-binding protein 4: a functional biomarker of the proximal renal tubule, Adv Clin Chem, vol.63, pp.85-122, 2014.

M. Suzuki, G. F. Ross, and K. Wiers, Identification of a urinary proteomic signature for lupus nephritis in children, Pediatr Nephrol, vol.22, issue.12, pp.2047-2057, 2007.

S. Fleming and A. A. Gibson, Proteinase inhibitors in the kidney and its tumours, Histopathology, vol.10, pp.1303-1316, 1986.

Y. K. Lim, A. Jenner, and A. B. Ali, Haptoglobin reduces renal oxidative DNA and tissue damage during phenylhydrazine-induced hemolysis, Kidney Int, vol.58, pp.1033-1077, 2000.

T. Wu, Y. Du, and J. Han, Urinary angiostatin-a novel putative marker of renal pathology chronicity in lupus nephritis, Mol Cell Proteomics, vol.12, issue.5, pp.1170-1179, 2013.

M. S. O'reilly, L. Holmgren, Y. Shing, C. Chen, R. A. Rosenthal et al., Angiostatin: a novel angiogenesis inhibitor that mediates the suppression of metastases by a Lewis lung carcinoma, Cell, vol.79, pp.315-328, 1994.

M. S. O'reilly, L. Holmgren, Y. Shing, C. Chen, R. A. Rosenthal et al., Angiostatin: a circulating endothelial cell inhibitor that suppresses angiogenesis and tumor growth, Cold Spring Harb. Symp. Quant. Biol, vol.59, pp.471-482, 1994.

T. Rubinstein, M. Pitashny, and C. Putterman, The novel role of neutrophil gelatinase-B associated lipocalin (NGAL)/lipocalin-2 as a biomarker for lupus nephritis, Autoimmun. Rev, vol.7, issue.3, pp.229-234, 2008.

J. B. Cowland and N. Borregaard, Molecular characterization and pattern of tissue expression of the gene for neutrophil gelatinase-associated lipocalin from humans, Genomics, vol.45, issue.1, pp.17-23, 1997.

M. Bennett, C. L. Dent, and Q. Ma, Urine NGAL predicts severity of acute kidney injury after cardiac surgery: a prospective study, Clin J Am Soc Nephrol, vol.3, pp.665-73, 2008.

M. Haase, R. Bellomo, and P. Devarajan, Novel biomarkers early predict the severity of acute kidney injury after cardiac surgery in adults, Ann Thorac Surg, vol.88, pp.124-154, 2009.

T. Rubinstein, M. Pitashny, and C. Putterman, The novel role of neutrophil gelatinase-B associated lipocalin (NGAL)/Lipocalin-2 as a biomarker for lupus nephritis, Autoimmun Rev, vol.7, pp.229-234, 2008.

M. S. Shahawy, M. H. Hemida, A. Hafez, . Abdel-hafez, Z. Tarek et al., Urinary neutrophil gelatinase-associated lipocalin as a marker for disease activity in lupus nephritis, 2018.

T. Rubinstein, M. Pitashny, and B. Levine, Urinary neutrophil gelatinase-associated lipocalin as a novel biomarker for disease activity in lupus nephritis, Rheumatology (Oxford), vol.49, pp.960-71, 2010.

B. Satirapoj, C. Kitiyakara, and A. Leelahavanichkul, Yingyos Avihingsanon and Ouppatham Supasyndh ; Urine neutrophil gelatinase-associated lipocalin to predict renal response after induction therapy in active lupus nephritis, 2017.

Y. E. Housseini, J. Schmidtko, and P. Meier, Intérêt de la neutrophil gelatinase-associated lipocalin comme aide au diagnostic de l'insuffisance rénale aiguë, Rev Med Suisse, vol.9, pp.2101-2106, 2013.

T. Ichimura, C. C. Hung, S. A. Yang, J. L. Stevens, and J. V. Bonventre, Kidney injury molecule-1 (Kim-1): a tissue and urinary biomarker for nephrotoxicant-induced renal injury, Am J Physiol Renal Physiol, vol.286, pp.552-563, 2004.

M. M. Van-timmeren, M. C. Van-den-heuvel, and V. Bailly, Tubular kidney injury molecule-1 (KIM-1) in human renal disease, J Pathol, vol.212, pp.209-217, 2007.

Y. Nozaki, T. Kinoshita, T. Yano, . Shiga, . Hino et al., Estimation of kidney injury molecule-1 (Kim-1) in patients with lupus nephritis, 2014.

Y. Ding, L. Nie, Y. Pang, W. Wu, Y. Tan et al., Composite urinary biomarkers to predict pathological tubulointerstitial lesions in lupus nephritis, 2018.

W. K. Han, A. Alinani, and C. L. Wu, Human kidney injury molecule-1 is a tissue and urinary tumor marker of renal cell carcinoma, J Am Soc Nephrol, vol.16, pp.1126-1134, 2005.

E. W. Kuehn, K. M. Park, S. Somlo, and J. V. Bonventre, Kidney injury molecule-1 expression in murine polycystic kidney disease, Am J Physiol Renal Physiol, vol.283, pp.1326-1336, 2002.

W. K. Han, V. Bailly, R. Abichandani, and R. Thadhani, Kidney injury molecule-1 (Kim-1): a novel biomarker for human renal proximal tubule injury, Kidney Int, vol.62, pp.237-244, 2002.

U. Andersson, H. Erlandsson-harris, H. Yang, and K. J. Tracey, HMGB1 as a DNA-binding cytokine, J Leukoc Biol, vol.72, pp.1084-91, 2002.

Z. Xu, J. Lv, S. Fu, J. Chen, H. Yang et al., High mobility group protein B1 (HMGB1) increased in kidney tissues of patients with lupus nephritis, 2017.

A. Zickert, K. Palmblad, and B. Sundelin, Renal expression and serum levels of high mobility group box 1 protein in lupus nephritis, Arthritis Res Ther, 2012.

X. Qing, M. Pitashny, and D. B. Thomas, Pathogenic anti-DNA antibodies modulate gene expression in mesangial cells: involvement of HMGB1 in anti-DNA antibody-induced renal injury, Immunol Lett, vol.121, pp.61-73, 2008.

R. Neelakshi, I. Jog, I. Blanco, C. Lee, R. Putterman et al.,

, Urinary High Mobility Group Box-1 Associates Specifically with Lupus Nephritis Class V, 2016.

A. Bruchfeld, M. Wendt, J. Bratt, R. Abdul, S. Qureshi et al., Iva Gunnarsson; High-mobility group box-1 protein (HMGB1) is increased in antineutrophilic cytoplasmatic antibody (ANCA)-associated vasculitis with renal manifestations, 2010.

M. Kristiansen, J. H. Graversen, and C. Jacobsen, Identification of the haemoglobin scavenger receptor, Nature, vol.409, pp.198-201, 2001.

H. Van-gorp, P. L. Delputte, and H. J. Nauwynck, Scavenger receptor CD163, a Jack-of-all-trades and potential target for cell-directed therapy, MolImmunol, vol.47, pp.1650-1660, 2010.

A. Droste, C. Sorg, and P. Hogger, Shedding of CD163, a novel regulatory mechanism for a member of the scavenger receptor cysteine-richfamily, Biochem Biophys Res Commun, vol.256, pp.110-113, 1999.

H. J. Moller, N. A. Peterslund, and J. H. Graversen, Identification of the hemoglobin scavenger receptor/CD163 as a natural soluble protein in plasma, Blood, vol.99, pp.378-380, 2002.

Q. Samia, I. Khan, V. Khan, and . Gupta, CD11b Activity Modulates Pathogenesis of Lupus Nephritis, 2018.

T. Springer, G. Galfre, D. S. Secher, C. Milstein, T. Springer et al., Mac-1: a macrophage differentiation antigen identified by monoclonal antibody, Eur J Immunol, vol.23, issue.4, pp.301-307, 1979.

D. C. Anderson, L. J. Miller, F. C. Schmalstieg, R. Rothlein, T. 2. Springer et al., Contributions of the Mac-1 glycoprotein family to adherence-dependent granulocyte functions: structure-function assessments employing subunit-specific monoclonal antibodies, J Immunol, vol.137, issue.1, pp.15-27, 1986.

A. B. Kantor, A. M. Stall, S. Adams, L. A. Herzenberg, L. A. Herzenberg et al., Differential development of progenitor activity for three B-cell lineages, Proc Natl Acad Sci, vol.89, issue.8, pp.3320-3324, 1992.

H. I. Mcfarland, S. R. Nahill, J. W. Maciaszek, R. 2. Welsh, H. I. Mcfarland et al., CD11b (Mac-1): a marker for CD8+ cytotoxic T cell activation and memory in virus infection, J Immunol, vol.149, issue.4, pp.1326-1359, 1992.

D. I. Beller, T. A. Springer, R. 3. Schreiber, D. I. Beller, T. A. Springer et al., Anti-Mac-1 selectively inhibits the mouse and human type three complement receptor, J Exp Med, 1982.

S. C. Fagerholm, M. Varis, M. Stefanidakis, T. J. Hilden, C. 3. Gahmberg et al., alpha-Chain phosphorylation of the human leukocyte CD11b/CD18 (Mac-1) integrin is pivotal for integrin activation to bind ICAMs and leukocyte extravasation, Blood, 2006.

M. Phillipson, B. Heit, P. Colarusso, L. Liu, C. M. Ballantyne et al., Intraluminal crawling of neutrophils to emigration sites: a molecularly distinct process from adhesion in the recruitment cascade, J Exp Med, 2006.

J. J. Alexander, L. D. Chaves, A. Chang, A. Jacob, M. Ritchie et al., CD11b is protective in complement-mediated immune complex glomerulonephritis, Kidney Int, 2015.

J. B. Harley, M. E. Alarcon-riquelme, L. A. Criswell, J. Co, and R. P. Kimberly, International Consortium for Systemic Lupus Erythematosus Genetics, Genome-wide association scan in women with systemic lupus erythematosus identifies susceptibility variants in ITGAM, PXK, KIAA1542 and other loci, Nat Genet, 2008.

S. K. Nath, S. Han, X. Kim-howard, J. A. Kelly, P. Viswanathan et al., A nonsynonymous functional variant in integrin-alpha(M) (encoded by ITGAM) is associated with systemic lupus erythematosus, Nat Genet, 2008.