Skip to main content
Top
Published in: Clinical Reviews in Allergy & Immunology 2/2017

01-10-2017

Dysregulated Lymphoid Cell Populations in Mouse Models of Systemic Lupus Erythematosus

Authors: Aurélie De Groof, Patrice Hémon, Olivier Mignen, Jacques-Olivier Pers, Edward K. Wakeland, Yves Renaudineau, Bernard R. Lauwerys

Published in: Clinical Reviews in Allergy & Immunology | Issue 2/2017

Login to get access

Abstract

Biases in the distribution and phenotype of T, B, and antigen-presenting cell populations are strongly connected to mechanisms of disease development in mouse models of systemic lupus erythematosus (SLE). Here, we describe longitudinal changes in lymphoid and antigen-presenting cell subsets in bone marrow, blood and spleen from two lupus-prone strains (MRL/lpr and B6.Sle1.Sle2.Sle3 tri-congenic mice), and how they integrate in our present understanding of the pathogenesis of the disease. In particular, we focus on (autoreactive) T cell activation patterns in lupus-prone mice. Break of T cell tolerance to chromatin constituents (histone peptides) is key to the development of the disease and is related to T cell intrinsic defects, contributed by genetic susceptibility factors and by extrinsic amplificatory mechanisms, in particular over-stimulation by antigen-presenting cells. We also describe shifts in B cell sub-populations, going from skewed immature B cell populations as an indication of disturbed central and peripheral tolerance checkpoints, to enriched long-lived plasma cells, which are key to persistent autoantibody production in the disease. B cell activation mechanisms in SLE are both T cell-dependent (break of tolerance and production of specific autoantibodies) and -independent (polyclonal B cell activation, production of autoantibodies by long-lived plasma cells). By providing a comprehensive evaluation of B and T cell surface markers in two major mouse models of SLE and a description of their changes before and after disease onset, this review illustrates how the study of lymphoid cell phenotype delivers key information regarding pathogenic pathways and supplies tools to assess the beneficial effects of novel therapeutic interventions.
Literature
3.
go back to reference Limaye N, Belobrajdic KA, Wandstrat AE, Bonhomme F, Edwards SV, Wakeland EK (2008) Prevalence and evolutionary origins of autoimmune susceptibility alleles in natural mouse populations. Genes Immun 9(1):61–68. doi:10.1038/sj.gene.6364446 PubMedCrossRef Limaye N, Belobrajdic KA, Wandstrat AE, Bonhomme F, Edwards SV, Wakeland EK (2008) Prevalence and evolutionary origins of autoimmune susceptibility alleles in natural mouse populations. Genes Immun 9(1):61–68. doi:10.​1038/​sj.​gene.​6364446 PubMedCrossRef
4.
go back to reference Konsta OD, Le Dantec C, Charras A, Brooks WH, Arleevskaya MI, Bordron A, Renaudineau Y (2015) An in silico approach reveals associations between genetic and epigenetic factors within regulatory elements in B cells from primary Sjogren’s syndrome patients. Front Immunol 6:437. doi:10.3389/fimmu.2015.00437 PubMedPubMedCentralCrossRef Konsta OD, Le Dantec C, Charras A, Brooks WH, Arleevskaya MI, Bordron A, Renaudineau Y (2015) An in silico approach reveals associations between genetic and epigenetic factors within regulatory elements in B cells from primary Sjogren’s syndrome patients. Front Immunol 6:437. doi:10.​3389/​fimmu.​2015.​00437 PubMedPubMedCentralCrossRef
7.
go back to reference Charras A, Konsta OD, Le Dantec C, Bagacean C, Kapsogeorgou EK, Tzioufas AG, Pers JO, Bordron A, Renaudineau Y (2017) Cell-specific epigenome-wide DNA methylation profile in long-term cultured minor salivary gland epithelial cells from patients with Sjogren’s syndrome. Ann Rheum Dis 76(3):625–628. doi:10.1136/annrheumdis-2016-210167 PubMedCrossRef Charras A, Konsta OD, Le Dantec C, Bagacean C, Kapsogeorgou EK, Tzioufas AG, Pers JO, Bordron A, Renaudineau Y (2017) Cell-specific epigenome-wide DNA methylation profile in long-term cultured minor salivary gland epithelial cells from patients with Sjogren’s syndrome. Ann Rheum Dis 76(3):625–628. doi:10.​1136/​annrheumdis-2016-210167 PubMedCrossRef
8.
go back to reference Morel L, Rudofsky UH, Longmate JA, Schiffenbauer J, Wakeland EK (1994) Polygenic control of susceptibility to murine systemic lupus erythematosus. Immunity 1(3):219–229PubMedCrossRef Morel L, Rudofsky UH, Longmate JA, Schiffenbauer J, Wakeland EK (1994) Polygenic control of susceptibility to murine systemic lupus erythematosus. Immunity 1(3):219–229PubMedCrossRef
9.
go back to reference Morel L, Yu Y, Blenman KR, Caldwell RA, Wakeland EK (1996) Production of congenic mouse strains carrying genomic intervals containing SLE-susceptibility genes derived from the SLE-prone NZM2410 strain. Mammalian genome : official journal of the International Mammalian Genome Society 7(5):335–339CrossRef Morel L, Yu Y, Blenman KR, Caldwell RA, Wakeland EK (1996) Production of congenic mouse strains carrying genomic intervals containing SLE-susceptibility genes derived from the SLE-prone NZM2410 strain. Mammalian genome : official journal of the International Mammalian Genome Society 7(5):335–339CrossRef
10.
go back to reference Morel L, Croker BP, Blenman KR, Mohan C, Huang G, Gilkeson G, Wakeland EK (2000) Genetic reconstitution of systemic lupus erythematosus immunopathology with polycongenic murine strains. Proc Natl Acad Sci U S A 97(12):6670–6675PubMedPubMedCentralCrossRef Morel L, Croker BP, Blenman KR, Mohan C, Huang G, Gilkeson G, Wakeland EK (2000) Genetic reconstitution of systemic lupus erythematosus immunopathology with polycongenic murine strains. Proc Natl Acad Sci U S A 97(12):6670–6675PubMedPubMedCentralCrossRef
11.
go back to reference Khamashta M, Merrill JT, Werth VP, Furie R, Kalunian K, Illei GG, Drappa J, Wang L, Greth W, investigators CDs (2016) Sifalimumab, an anti-interferon-alpha monoclonal antibody, in moderate to severe systemic lupus erythematosus: a randomised, double-blind, placebo-controlled study. Ann Rheum Dis 75(11):1909–1916. doi:10.1136/annrheumdis-2015-208562 PubMedPubMedCentralCrossRef Khamashta M, Merrill JT, Werth VP, Furie R, Kalunian K, Illei GG, Drappa J, Wang L, Greth W, investigators CDs (2016) Sifalimumab, an anti-interferon-alpha monoclonal antibody, in moderate to severe systemic lupus erythematosus: a randomised, double-blind, placebo-controlled study. Ann Rheum Dis 75(11):1909–1916. doi:10.​1136/​annrheumdis-2015-208562 PubMedPubMedCentralCrossRef
13.
go back to reference Lauwerys BR, Hachulla E, Spertini F, Lazaro E, Jorgensen C, Mariette X, Haelterman E, Grouard-Vogel G, Fanget B, Dhellin O, Vandepapeliere P, Houssiau FA (2013) Down-regulation of interferon signature in systemic lupus erythematosus patients by active immunization with interferon alpha-kinoid. Arthritis & Rheumatism 65(2):447–456. doi:10.1002/art.37785 CrossRef Lauwerys BR, Hachulla E, Spertini F, Lazaro E, Jorgensen C, Mariette X, Haelterman E, Grouard-Vogel G, Fanget B, Dhellin O, Vandepapeliere P, Houssiau FA (2013) Down-regulation of interferon signature in systemic lupus erythematosus patients by active immunization with interferon alpha-kinoid. Arthritis & Rheumatism 65(2):447–456. doi:10.​1002/​art.​37785 CrossRef
14.
go back to reference Ducreux J, Houssiau FA, Vandepapeliere P, Jorgensen C, Lazaro E, Spertini F, Colaone F, Roucairol C, Laborie M, Croughs T, Grouard-Vogel G, Lauwerys BR (2016) Interferon alpha kinoid induces neutralizing anti-interferon alpha antibodies that decrease the expression of interferon-induced and B cell activation associated transcripts: analysis of extended follow-up data from the interferon alpha kinoid phase I/II study. Rheumatology (Oxford) 55(10):1901–1905. doi:10.1093/rheumatology/kew262 CrossRef Ducreux J, Houssiau FA, Vandepapeliere P, Jorgensen C, Lazaro E, Spertini F, Colaone F, Roucairol C, Laborie M, Croughs T, Grouard-Vogel G, Lauwerys BR (2016) Interferon alpha kinoid induces neutralizing anti-interferon alpha antibodies that decrease the expression of interferon-induced and B cell activation associated transcripts: analysis of extended follow-up data from the interferon alpha kinoid phase I/II study. Rheumatology (Oxford) 55(10):1901–1905. doi:10.​1093/​rheumatology/​kew262 CrossRef
15.
go back to reference Liu Z, Bethunaickan R, Huang W, Lodhi U, Solano I, Madaio MP, Davidson A (2011) Interferon-alpha accelerates murine systemic lupus erythematosus in a T cell-dependent manner. Arthritis & Rheumatism 63(1):219–229. doi:10.1002/art.30087 CrossRef Liu Z, Bethunaickan R, Huang W, Lodhi U, Solano I, Madaio MP, Davidson A (2011) Interferon-alpha accelerates murine systemic lupus erythematosus in a T cell-dependent manner. Arthritis & Rheumatism 63(1):219–229. doi:10.​1002/​art.​30087 CrossRef
16.
go back to reference Mathian A, Weinberg A, Gallegos M, Banchereau J, Koutouzov S (2005) IFN-alpha induces early lethal lupus in preautoimmune (New Zealand black x New Zealand White) F1 but not in BALB/c mice. J Immunol 174(5):2499–2506PubMedCrossRef Mathian A, Weinberg A, Gallegos M, Banchereau J, Koutouzov S (2005) IFN-alpha induces early lethal lupus in preautoimmune (New Zealand black x New Zealand White) F1 but not in BALB/c mice. J Immunol 174(5):2499–2506PubMedCrossRef
17.
19.
go back to reference Ichii O, Konno A, Sasaki N, Endoh D, Hashimoto Y, Kon Y (2008) Autoimmune glomerulonephritis induced in congenic mouse strain carrying telomeric region of chromosome 1 derived from MRL/MpJ. Histol Histopathol 23(4):411–422PubMed Ichii O, Konno A, Sasaki N, Endoh D, Hashimoto Y, Kon Y (2008) Autoimmune glomerulonephritis induced in congenic mouse strain carrying telomeric region of chromosome 1 derived from MRL/MpJ. Histol Histopathol 23(4):411–422PubMed
20.
go back to reference Santiago-Raber ML, Haraldsson MK, Theofilopoulos AN, Kono DH (2007) Characterization of reciprocal Lmb1-4 interval MRL-Faslpr and C57BL/6-Faslpr congenic mice reveals significant effects from Lmb3. J Immunol 178(12):8195–8202PubMedCrossRef Santiago-Raber ML, Haraldsson MK, Theofilopoulos AN, Kono DH (2007) Characterization of reciprocal Lmb1-4 interval MRL-Faslpr and C57BL/6-Faslpr congenic mice reveals significant effects from Lmb3. J Immunol 178(12):8195–8202PubMedCrossRef
21.
go back to reference Kong PL, Morel L, Croker BP, Craft J (2004) The centromeric region of chromosome 7 from MRL mice (Lmb3) is an epistatic modifier of Fas for autoimmune disease expression. J Immunol 172(5):2785–2794PubMedCrossRef Kong PL, Morel L, Croker BP, Craft J (2004) The centromeric region of chromosome 7 from MRL mice (Lmb3) is an epistatic modifier of Fas for autoimmune disease expression. J Immunol 172(5):2785–2794PubMedCrossRef
22.
go back to reference Yamada A, Miyazaki T, Lu LM, Ono M, Ito MR, Terada M, Mori S, Hata K, Nozaki Y, Nakatsuru S, Nakamura Y, Onji M, Nose M (2003) Genetic basis of tissue specificity of vasculitis in MRL/lpr mice. Arthritis & Rheumatism 48(5):1445–1451. doi:10.1002/art.10952 CrossRef Yamada A, Miyazaki T, Lu LM, Ono M, Ito MR, Terada M, Mori S, Hata K, Nozaki Y, Nakatsuru S, Nakamura Y, Onji M, Nose M (2003) Genetic basis of tissue specificity of vasculitis in MRL/lpr mice. Arthritis & Rheumatism 48(5):1445–1451. doi:10.​1002/​art.​10952 CrossRef
23.
go back to reference Kamogawa J, Terada M, Mizuki S, Nishihara M, Yamamoto H, Mori S, Abe Y, Morimoto K, Nakatsuru S, Nakamura Y, Nose M (2002) Arthritis in MRL/lpr mice is under the control of multiple gene loci with an allelic combination derived from the original inbred strains. Arthritis & Rheumatism 46(4):1067–1074CrossRef Kamogawa J, Terada M, Mizuki S, Nishihara M, Yamamoto H, Mori S, Abe Y, Morimoto K, Nakatsuru S, Nakamura Y, Nose M (2002) Arthritis in MRL/lpr mice is under the control of multiple gene loci with an allelic combination derived from the original inbred strains. Arthritis & Rheumatism 46(4):1067–1074CrossRef
25.
27.
28.
go back to reference Seret G, Canas F, Pougnet-Di Costanzo L, Hanrotel-Saliou C, Jousse-Joulin S, Le Meur Y, Saraux A, Valeri A, Putterman C, Youinou P, Rojas-Villarraga A, Anaya JM, Renaudineau Y (2015) Anti-alpha-actinin antibodies are part of the anti-cell membrane antibody spectrum that characterize patients with lupus nephritis. J Autoimmun 61:54–61. doi:10.1016/j.jaut.2015.05.009 PubMedCrossRef Seret G, Canas F, Pougnet-Di Costanzo L, Hanrotel-Saliou C, Jousse-Joulin S, Le Meur Y, Saraux A, Valeri A, Putterman C, Youinou P, Rojas-Villarraga A, Anaya JM, Renaudineau Y (2015) Anti-alpha-actinin antibodies are part of the anti-cell membrane antibody spectrum that characterize patients with lupus nephritis. J Autoimmun 61:54–61. doi:10.​1016/​j.​jaut.​2015.​05.​009 PubMedCrossRef
29.
go back to reference Renaudineau Y, Croquefer S, Jousse S, Renaudineau E, Devauchelle V, Gueguen P, Hanrotel C, Gilburd B, Saraux A, Shoenfeld Y, Putterman C, Youinou P (2006) Association of alpha-actinin-binding anti-double-stranded DNA antibodies with lupus nephritis. Arthritis & Rheumatism 54(8):2523–2532. doi:10.1002/art.22015 CrossRef Renaudineau Y, Croquefer S, Jousse S, Renaudineau E, Devauchelle V, Gueguen P, Hanrotel C, Gilburd B, Saraux A, Shoenfeld Y, Putterman C, Youinou P (2006) Association of alpha-actinin-binding anti-double-stranded DNA antibodies with lupus nephritis. Arthritis & Rheumatism 54(8):2523–2532. doi:10.​1002/​art.​22015 CrossRef
30.
go back to reference Xie C, Zhou XJ, Liu X, Mohan C (2003) Enhanced susceptibility to end-organ disease in the lupus-facilitating NZW mouse strain. Arthritis & Rheumatism 48(4):1080–1092. doi:10.1002/art.10887 CrossRef Xie C, Zhou XJ, Liu X, Mohan C (2003) Enhanced susceptibility to end-organ disease in the lupus-facilitating NZW mouse strain. Arthritis & Rheumatism 48(4):1080–1092. doi:10.​1002/​art.​10887 CrossRef
32.
go back to reference Mohan C, Adams S, Stanik V, Datta SK (1993) Nucleosome: a major immunogen for pathogenic autoantibody-inducing T cells of lupus. J Exp Med 177(5):1367–1381PubMedCrossRef Mohan C, Adams S, Stanik V, Datta SK (1993) Nucleosome: a major immunogen for pathogenic autoantibody-inducing T cells of lupus. J Exp Med 177(5):1367–1381PubMedCrossRef
34.
go back to reference Wofsy D, Seaman WE (1985) Successful treatment of autoimmunity in NZB/NZW F1 mice with monoclonal antibody to L3T4. J Exp Med 161(2):378–391PubMedCrossRef Wofsy D, Seaman WE (1985) Successful treatment of autoimmunity in NZB/NZW F1 mice with monoclonal antibody to L3T4. J Exp Med 161(2):378–391PubMedCrossRef
35.
go back to reference Wofsy D, Seaman WE (1987) Reversal of advanced murine lupus in NZB/NZW F1 mice by treatment with monoclonal antibody to L3T4. J Immunol 138(10):3247–3253PubMed Wofsy D, Seaman WE (1987) Reversal of advanced murine lupus in NZB/NZW F1 mice by treatment with monoclonal antibody to L3T4. J Immunol 138(10):3247–3253PubMed
36.
go back to reference Carteron NL, Wofsy D, Schimenti C, Ermak TH (1990) F(ab’)2 anti-CD4 and intact anti-CD4 monoclonal antibodies inhibit the accumulation of CD4+ T cells, CD8+ T cells, and B cells in the kidneys of lupus-prone NZB/NZW mice. Clin Immunol Immunopathol 56(3):373–383PubMedCrossRef Carteron NL, Wofsy D, Schimenti C, Ermak TH (1990) F(ab’)2 anti-CD4 and intact anti-CD4 monoclonal antibodies inhibit the accumulation of CD4+ T cells, CD8+ T cells, and B cells in the kidneys of lupus-prone NZB/NZW mice. Clin Immunol Immunopathol 56(3):373–383PubMedCrossRef
37.
go back to reference Connolly K, Roubinian JR, Wofsy D (1992) Development of murine lupus in CD4-depleted NZB/NZW mice. Sustained inhibition of residual CD4+ T cells is required to suppress autoimmunity. J Immunol 149(9):3083–3088PubMed Connolly K, Roubinian JR, Wofsy D (1992) Development of murine lupus in CD4-depleted NZB/NZW mice. Sustained inhibition of residual CD4+ T cells is required to suppress autoimmunity. J Immunol 149(9):3083–3088PubMed
38.
go back to reference Santoro TJ, Portanova JP, Kotzin BL (1988) The contribution of L3T4+ T cells to lymphoproliferation and autoantibody production in MRL-lpr/lpr mice. J Exp Med 167(5):1713–1718PubMedCrossRef Santoro TJ, Portanova JP, Kotzin BL (1988) The contribution of L3T4+ T cells to lymphoproliferation and autoantibody production in MRL-lpr/lpr mice. J Exp Med 167(5):1713–1718PubMedCrossRef
39.
go back to reference Merino R, Iwamoto M, Fossati L, Izui S (1993) Polyclonal B cell activation arises from different mechanisms in lupus-prone (NZB x NZW)F1 and MRL/MpJ-lpr/lpr mice. J Immunol 151(11):6509–6516PubMed Merino R, Iwamoto M, Fossati L, Izui S (1993) Polyclonal B cell activation arises from different mechanisms in lupus-prone (NZB x NZW)F1 and MRL/MpJ-lpr/lpr mice. J Immunol 151(11):6509–6516PubMed
40.
go back to reference Chesnutt MS, Finck BK, Killeen N, Connolly MK, Goodman H, Wofsy D (1998) Enhanced lymphoproliferation and diminished autoimmunity in CD4-deficient MRL/lpr mice. Clin Immunol Immunopathol 87(1):23–32PubMedCrossRef Chesnutt MS, Finck BK, Killeen N, Connolly MK, Goodman H, Wofsy D (1998) Enhanced lymphoproliferation and diminished autoimmunity in CD4-deficient MRL/lpr mice. Clin Immunol Immunopathol 87(1):23–32PubMedCrossRef
41.
go back to reference Mohan C, Alas E, Morel L, Yang P, Wakeland EK (1998) Genetic dissection of SLE pathogenesis. Sle1 on murine chromosome 1 leads to a selective loss of tolerance to H2A/H2B/DNA subnucleosomes. J Clin Invest 101(6):1362–1372PubMedPubMedCentral Mohan C, Alas E, Morel L, Yang P, Wakeland EK (1998) Genetic dissection of SLE pathogenesis. Sle1 on murine chromosome 1 leads to a selective loss of tolerance to H2A/H2B/DNA subnucleosomes. J Clin Invest 101(6):1362–1372PubMedPubMedCentral
42.
go back to reference Sobel ES, Satoh M, Chen Y, Wakeland EK, Morel L (2002) The major murine systemic lupus erythematosus susceptibility locus Sle1 results in abnormal functions of both B and T cells. J Immunol 169(5):2694–2700PubMedCrossRef Sobel ES, Satoh M, Chen Y, Wakeland EK, Morel L (2002) The major murine systemic lupus erythematosus susceptibility locus Sle1 results in abnormal functions of both B and T cells. J Immunol 169(5):2694–2700PubMedCrossRef
43.
go back to reference Sobel ES, Mohan C, Morel L, Schiffenbauer J, Wakeland EK (1999) Genetic dissection of SLE pathogenesis: adoptive transfer of Sle1 mediates the loss of tolerance by bone marrow-derived B cells. J Immunol 162(4):2415–2421PubMed Sobel ES, Mohan C, Morel L, Schiffenbauer J, Wakeland EK (1999) Genetic dissection of SLE pathogenesis: adoptive transfer of Sle1 mediates the loss of tolerance by bone marrow-derived B cells. J Immunol 162(4):2415–2421PubMed
44.
go back to reference Wandstrat AE, Nguyen C, Limaye N, Chan AY, Subramanian S, Tian XH, Yim YS, Pertsemlidis A, Garner HR Jr, Morel L, Wakeland EK (2004) Association of extensive polymorphisms in the SLAM/CD2 gene cluster with murine lupus. Immunity 21(6):769–780. doi:10.1016/j.immuni.2004.10.009 PubMedCrossRef Wandstrat AE, Nguyen C, Limaye N, Chan AY, Subramanian S, Tian XH, Yim YS, Pertsemlidis A, Garner HR Jr, Morel L, Wakeland EK (2004) Association of extensive polymorphisms in the SLAM/CD2 gene cluster with murine lupus. Immunity 21(6):769–780. doi:10.​1016/​j.​immuni.​2004.​10.​009 PubMedCrossRef
46.
go back to reference Simpson N, Gatenby PA, Wilson A, Malik S, Fulcher DA, Tangye SG, Manku H, Vyse TJ, Roncador G, Huttley GA, Goodnow CC, Vinuesa CG, Cook MC (2010) Expansion of circulating T cells resembling follicular helper T cells is a fixed phenotype that identifies a subset of severe systemic lupus erythematosus. Arthritis & Rheumatism 62(1):234–244. doi:10.1002/art.25032 CrossRef Simpson N, Gatenby PA, Wilson A, Malik S, Fulcher DA, Tangye SG, Manku H, Vyse TJ, Roncador G, Huttley GA, Goodnow CC, Vinuesa CG, Cook MC (2010) Expansion of circulating T cells resembling follicular helper T cells is a fixed phenotype that identifies a subset of severe systemic lupus erythematosus. Arthritis & Rheumatism 62(1):234–244. doi:10.​1002/​art.​25032 CrossRef
47.
go back to reference Jacquemin C, Schmitt N, Contin-Bordes C, Liu Y, Narayanan P, Seneschal J, Maurouard T, Dougall D, Davizon ES, Dumortier H, Douchet I, Raffray L, Richez C, Lazaro E, Duffau P, Truchetet ME, Khoryati L, Mercie P, Couzi L, Merville P, Schaeverbeke T, Viallard JF, Pellegrin JL, Moreau JF, Muller S, Zurawski S, Coffman RL, Pascual V, Ueno H, Blanco P (2015) OX40 ligand contributes to human lupus pathogenesis by promoting T follicular helper response. Immunity 42(6):1159–1170. doi:10.1016/j.immuni.2015.05.012 PubMedPubMedCentralCrossRef Jacquemin C, Schmitt N, Contin-Bordes C, Liu Y, Narayanan P, Seneschal J, Maurouard T, Dougall D, Davizon ES, Dumortier H, Douchet I, Raffray L, Richez C, Lazaro E, Duffau P, Truchetet ME, Khoryati L, Mercie P, Couzi L, Merville P, Schaeverbeke T, Viallard JF, Pellegrin JL, Moreau JF, Muller S, Zurawski S, Coffman RL, Pascual V, Ueno H, Blanco P (2015) OX40 ligand contributes to human lupus pathogenesis by promoting T follicular helper response. Immunity 42(6):1159–1170. doi:10.​1016/​j.​immuni.​2015.​05.​012 PubMedPubMedCentralCrossRef
48.
go back to reference Mohan C, Yu Y, Morel L, Yang P, Wakeland EK (1999) Genetic dissection of Sle pathogenesis: Sle3 on murine chromosome 7 impacts T cell activation, differentiation, and cell death. J Immunol 162(11):6492–6502PubMed Mohan C, Yu Y, Morel L, Yang P, Wakeland EK (1999) Genetic dissection of Sle pathogenesis: Sle3 on murine chromosome 7 impacts T cell activation, differentiation, and cell death. J Immunol 162(11):6492–6502PubMed
49.
go back to reference Bethunaickan R, Berthier CC, Zhang W, Eksi R, Li HD, Guan Y, Kretzler M, Davidson A (2014) Identification of stage-specific genes associated with lupus nephritis and response to remission induction in (NZB x NZW)F1 and NZM2410 mice. Arthritis & Rheumatology 66(8):2246–2258. doi:10.1002/art.38679 CrossRef Bethunaickan R, Berthier CC, Zhang W, Eksi R, Li HD, Guan Y, Kretzler M, Davidson A (2014) Identification of stage-specific genes associated with lupus nephritis and response to remission induction in (NZB x NZW)F1 and NZM2410 mice. Arthritis & Rheumatology 66(8):2246–2258. doi:10.​1002/​art.​38679 CrossRef
52.
go back to reference Hwang SH, Lee H, Yamamoto M, Jones LA, Dayalan J, Hopkins R, Zhou XJ, Yarovinsky F, Connolly JE, Curotto de Lafaille MA, Wakeland EK, Fairhurst AM (2012) B cell TLR7 expression drives anti-RNA autoantibody production and exacerbates disease in systemic lupus erythematosus-prone mice. J Immunol 189(12):5786–5796. doi:10.4049/jimmunol.1202195 PubMedPubMedCentralCrossRef Hwang SH, Lee H, Yamamoto M, Jones LA, Dayalan J, Hopkins R, Zhou XJ, Yarovinsky F, Connolly JE, Curotto de Lafaille MA, Wakeland EK, Fairhurst AM (2012) B cell TLR7 expression drives anti-RNA autoantibody production and exacerbates disease in systemic lupus erythematosus-prone mice. J Immunol 189(12):5786–5796. doi:10.​4049/​jimmunol.​1202195 PubMedPubMedCentralCrossRef
53.
go back to reference Scalapino KJ, Tang Q, Bluestone JA, Bonyhadi ML, Daikh DI (2006) Suppression of disease in New Zealand black/New Zealand White lupus-prone mice by adoptive transfer of ex vivo expanded regulatory T cells. J Immunol 177(3):1451–1459PubMedCrossRef Scalapino KJ, Tang Q, Bluestone JA, Bonyhadi ML, Daikh DI (2006) Suppression of disease in New Zealand black/New Zealand White lupus-prone mice by adoptive transfer of ex vivo expanded regulatory T cells. J Immunol 177(3):1451–1459PubMedCrossRef
54.
go back to reference Humrich JY, Morbach H, Undeutsch R, Enghard P, Rosenberger S, Weigert O, Kloke L, Heimann J, Gaber T, Brandenburg S, Scheffold A, Huehn J, Radbruch A, Burmester GR, Riemekasten G (2010) Homeostatic imbalance of regulatory and effector T cells due to IL-2 deprivation amplifies murine lupus. Proc Natl Acad Sci U S A 107(1):204–209. doi:10.1073/pnas.0903158107 PubMedCrossRef Humrich JY, Morbach H, Undeutsch R, Enghard P, Rosenberger S, Weigert O, Kloke L, Heimann J, Gaber T, Brandenburg S, Scheffold A, Huehn J, Radbruch A, Burmester GR, Riemekasten G (2010) Homeostatic imbalance of regulatory and effector T cells due to IL-2 deprivation amplifies murine lupus. Proc Natl Acad Sci U S A 107(1):204–209. doi:10.​1073/​pnas.​0903158107 PubMedCrossRef
55.
go back to reference Yang CH, Tian L, Ling GS, Trendell-Smith NJ, Ma L, Lo CK, Stott DI, Liew FY, Huang FP (2008) Immunological mechanisms and clinical implications of regulatory T cell deficiency in a systemic autoimmune disorder: roles of IL-2 versus IL-15. Eur J Immunol 38(6):1664–1676. doi:10.1002/eji.200838190 PubMedCrossRef Yang CH, Tian L, Ling GS, Trendell-Smith NJ, Ma L, Lo CK, Stott DI, Liew FY, Huang FP (2008) Immunological mechanisms and clinical implications of regulatory T cell deficiency in a systemic autoimmune disorder: roles of IL-2 versus IL-15. Eur J Immunol 38(6):1664–1676. doi:10.​1002/​eji.​200838190 PubMedCrossRef
56.
go back to reference Divekar AA, Dubey S, Gangalum PR, Singh RR (2011) Dicer insufficiency and microRNA-155 overexpression in lupus regulatory T cells: an apparent paradox in the setting of an inflammatory milieu. J Immunol 186(2):924–930. doi:10.4049/jimmunol.1002218 PubMedCrossRef Divekar AA, Dubey S, Gangalum PR, Singh RR (2011) Dicer insufficiency and microRNA-155 overexpression in lupus regulatory T cells: an apparent paradox in the setting of an inflammatory milieu. J Immunol 186(2):924–930. doi:10.​4049/​jimmunol.​1002218 PubMedCrossRef
57.
go back to reference Cuda CM, Wan S, Sobel ES, Croker BP, Morel L (2007) Murine lupus susceptibility locus Sle1a controls regulatory T cell number and function through multiple mechanisms. J Immunol 179(11):7439–7447PubMedCrossRef Cuda CM, Wan S, Sobel ES, Croker BP, Morel L (2007) Murine lupus susceptibility locus Sle1a controls regulatory T cell number and function through multiple mechanisms. J Immunol 179(11):7439–7447PubMedCrossRef
58.
go back to reference Wan S, Xia C, Morel L (2007) IL-6 produced by dendritic cells from lupus-prone mice inhibits CD4+CD25+ T cell regulatory functions. J Immunol 178(1):271–279PubMedCrossRef Wan S, Xia C, Morel L (2007) IL-6 produced by dendritic cells from lupus-prone mice inhibits CD4+CD25+ T cell regulatory functions. J Immunol 178(1):271–279PubMedCrossRef
59.
go back to reference Weigert O, von Spee C, Undeutsch R, Kloke L, Humrich JY, Riemekasten G (2013) CD4+Foxp3+ regulatory T cells prolong drug-induced disease remission in (NZBxNZW) F1 lupus mice. Arthritis research & therapy 15(1):R35. doi:10.1186/ar4188 CrossRef Weigert O, von Spee C, Undeutsch R, Kloke L, Humrich JY, Riemekasten G (2013) CD4+Foxp3+ regulatory T cells prolong drug-induced disease remission in (NZBxNZW) F1 lupus mice. Arthritis research & therapy 15(1):R35. doi:10.​1186/​ar4188 CrossRef
61.
go back to reference von Spee-Mayer C, Siegert E, Abdirama D, Rose A, Klaus A, Alexander T, Enghard P, Sawitzki B, Hiepe F, Radbruch A, Burmester GR, Riemekasten G, Humrich JY (2016) Low-dose interleukin-2 selectively corrects regulatory T cell defects in patients with systemic lupus erythematosus. Ann Rheum Dis 75(7):1407–1415. doi:10.1136/annrheumdis-2015-207776 CrossRef von Spee-Mayer C, Siegert E, Abdirama D, Rose A, Klaus A, Alexander T, Enghard P, Sawitzki B, Hiepe F, Radbruch A, Burmester GR, Riemekasten G, Humrich JY (2016) Low-dose interleukin-2 selectively corrects regulatory T cell defects in patients with systemic lupus erythematosus. Ann Rheum Dis 75(7):1407–1415. doi:10.​1136/​annrheumdis-2015-207776 CrossRef
67.
68.
71.
go back to reference Tsumiyama K, Hashiramoto A, Takimoto M, Tsuji-Kawahara S, Miyazawa M, Shiozawa S (2013) IFN-gamma-producing effector CD8 T lymphocytes cause immune glomerular injury by recognizing antigen presented as immune complex on target tissue. J Immunol 191(1):91–96. doi:10.4049/jimmunol.1203217 PubMedCrossRef Tsumiyama K, Hashiramoto A, Takimoto M, Tsuji-Kawahara S, Miyazawa M, Shiozawa S (2013) IFN-gamma-producing effector CD8 T lymphocytes cause immune glomerular injury by recognizing antigen presented as immune complex on target tissue. J Immunol 191(1):91–96. doi:10.​4049/​jimmunol.​1203217 PubMedCrossRef
72.
go back to reference Heymann F, Meyer-Schwesinger C, Hamilton-Williams EE, Hammerich L, Panzer U, Kaden S, Quaggin SE, Floege J, Grone HJ, Kurts C (2009) Kidney dendritic cell activation is required for progression of renal disease in a mouse model of glomerular injury. J Clin Invest 119(5):1286–1297. doi:10.1172/JCI38399 PubMedPubMedCentralCrossRef Heymann F, Meyer-Schwesinger C, Hamilton-Williams EE, Hammerich L, Panzer U, Kaden S, Quaggin SE, Floege J, Grone HJ, Kurts C (2009) Kidney dendritic cell activation is required for progression of renal disease in a mouse model of glomerular injury. J Clin Invest 119(5):1286–1297. doi:10.​1172/​JCI38399 PubMedPubMedCentralCrossRef
73.
go back to reference Couzi L, Merville P, Deminiere C, Moreau JF, Combe C, Pellegrin JL, Viallard JF, Blanco P (2007) Predominance of CD8+ T lymphocytes among periglomerular infiltrating cells and link to the prognosis of class III and class IV lupus nephritis. Arthritis & Rheumatism 56(7):2362–2370. doi:10.1002/art.22654 CrossRef Couzi L, Merville P, Deminiere C, Moreau JF, Combe C, Pellegrin JL, Viallard JF, Blanco P (2007) Predominance of CD8+ T lymphocytes among periglomerular infiltrating cells and link to the prognosis of class III and class IV lupus nephritis. Arthritis & Rheumatism 56(7):2362–2370. doi:10.​1002/​art.​22654 CrossRef
75.
go back to reference Taher TE, Muhammad HA, Rahim A, Flores-Borja F, Renaudineau Y, Isenberg DA, Mageed RA (2013) Aberrant B-lymphocyte responses in lupus: inherent or induced and potential therapeutic targets. Eur J Clin Investig 43(8):866–880. doi:10.1111/eci.12111 CrossRef Taher TE, Muhammad HA, Rahim A, Flores-Borja F, Renaudineau Y, Isenberg DA, Mageed RA (2013) Aberrant B-lymphocyte responses in lupus: inherent or induced and potential therapeutic targets. Eur J Clin Investig 43(8):866–880. doi:10.​1111/​eci.​12111 CrossRef
76.
go back to reference Youinou P, Taher TE, Pers JO, Mageed RA, Renaudineau Y (2009) B lymphocyte cytokines and rheumatic autoimmune disease. Arthritis & Rheumatism 60(7):1873–1880. doi:10.1002/art.24665 CrossRef Youinou P, Taher TE, Pers JO, Mageed RA, Renaudineau Y (2009) B lymphocyte cytokines and rheumatic autoimmune disease. Arthritis & Rheumatism 60(7):1873–1880. doi:10.​1002/​art.​24665 CrossRef
78.
go back to reference Jacobi AM, Reiter K, Mackay M, Aranow C, Hiepe F, Radbruch A, Hansen A, Burmester GR, Diamond B, Lipsky PE, Dorner T (2008) Activated memory B cell subsets correlate with disease activity in systemic lupus erythematosus: delineation by expression of CD27, IgD, and CD95. Arthritis & Rheumatism 58(6):1762–1773. doi:10.1002/art.23498 CrossRef Jacobi AM, Reiter K, Mackay M, Aranow C, Hiepe F, Radbruch A, Hansen A, Burmester GR, Diamond B, Lipsky PE, Dorner T (2008) Activated memory B cell subsets correlate with disease activity in systemic lupus erythematosus: delineation by expression of CD27, IgD, and CD95. Arthritis & Rheumatism 58(6):1762–1773. doi:10.​1002/​art.​23498 CrossRef
79.
go back to reference Simonin L, Pasquier E, Leroyer C, Cornec D, Lemerle J, Bendaoud B, Hillion S, Pers J-O, Couturaud F, Renaudineau Y (2016) Lymphocyte disturbances in primary antiphospholipid syndrome and application to venous thromboembolism follow-up. Clinical reviews in allergy & immunology:1–14. doi:10.1007/s12016-016-8568-1 Simonin L, Pasquier E, Leroyer C, Cornec D, Lemerle J, Bendaoud B, Hillion S, Pers J-O, Couturaud F, Renaudineau Y (2016) Lymphocyte disturbances in primary antiphospholipid syndrome and application to venous thromboembolism follow-up. Clinical reviews in allergy & immunology:1–14. doi:10.​1007/​s12016-016-8568-1
82.
go back to reference Youinou P, Renaudineau Y (2011) CD5 expression in B cells from patients with systemic lupus erythematosus. Crit Rev Immunol 31(1):31–42PubMedCrossRef Youinou P, Renaudineau Y (2011) CD5 expression in B cells from patients with systemic lupus erythematosus. Crit Rev Immunol 31(1):31–42PubMedCrossRef
86.
go back to reference Xu Z, Butfiloski EJ, Sobel ES, Morel L (2004) Mechanisms of peritoneal B-1a cells accumulation induced by murine lupus susceptibility locus Sle2. J Immunol 173(10):6050–6058PubMedCrossRef Xu Z, Butfiloski EJ, Sobel ES, Morel L (2004) Mechanisms of peritoneal B-1a cells accumulation induced by murine lupus susceptibility locus Sle2. J Immunol 173(10):6050–6058PubMedCrossRef
87.
go back to reference Xu Z, Duan B, Croker BP, Wakeland EK, Morel L (2005) Genetic dissection of the murine lupus susceptibility locus Sle2: contributions to increased peritoneal B-1a cells and lupus nephritis map to different loci. J Immunol 175(2):936–943PubMedCrossRef Xu Z, Duan B, Croker BP, Wakeland EK, Morel L (2005) Genetic dissection of the murine lupus susceptibility locus Sle2: contributions to increased peritoneal B-1a cells and lupus nephritis map to different loci. J Immunol 175(2):936–943PubMedCrossRef
89.
go back to reference Ye YL, Chuang YH, Chiang BL (1996) In vitro and in vivo functional analysis of CD5+ and CD5- B cells of autoimmune NZB x NZW F1 mice. Clin Exp Immunol 106(2):253–258PubMedPubMedCentralCrossRef Ye YL, Chuang YH, Chiang BL (1996) In vitro and in vivo functional analysis of CD5+ and CD5- B cells of autoimmune NZB x NZW F1 mice. Clin Exp Immunol 106(2):253–258PubMedPubMedCentralCrossRef
91.
go back to reference Garaud S, Taher TE, Debant M, Burgos M, Melayah S, Berthou C, Parikh K, Pers JO, Luque-Paz D, Chiocchia G, Peppelenbosch M, Isenberg DA, Youinou P, Mignen O, Renaudineau Y, Mageed RA (2016) CD5 expression promotes IL-10 production through activation of the MAPK/Erk pathway and upregulation of TRPC1 channels in B lymphocytes. Cellular & molecular immunology. doi:10.1038/cmi.2016.42 Garaud S, Taher TE, Debant M, Burgos M, Melayah S, Berthou C, Parikh K, Pers JO, Luque-Paz D, Chiocchia G, Peppelenbosch M, Isenberg DA, Youinou P, Mignen O, Renaudineau Y, Mageed RA (2016) CD5 expression promotes IL-10 production through activation of the MAPK/Erk pathway and upregulation of TRPC1 channels in B lymphocytes. Cellular & molecular immunology. doi:10.​1038/​cmi.​2016.​42
92.
go back to reference Garaud S, Morva A, Lemoine S, Hillion S, Bordron A, Pers JO, Berthou C, Mageed RA, Renaudineau Y, Youinou P (2011) CD5 promotes IL-10 production in chronic lymphocytic leukemia B cells through STAT3 and NFAT2 activation. J Immunol 186(8):4835–4844. doi:10.4049/jimmunol.1003050 PubMedCrossRef Garaud S, Morva A, Lemoine S, Hillion S, Bordron A, Pers JO, Berthou C, Mageed RA, Renaudineau Y, Youinou P (2011) CD5 promotes IL-10 production in chronic lymphocytic leukemia B cells through STAT3 and NFAT2 activation. J Immunol 186(8):4835–4844. doi:10.​4049/​jimmunol.​1003050 PubMedCrossRef
93.
go back to reference Roy V, Chang NH, Cai Y, Bonventi G, Wither J (2005) Aberrant IgM signaling promotes survival of transitional T1 B cells and prevents tolerance induction in lupus-prone New Zealand black mice. J Immunol 175(11):7363–7371PubMedCrossRef Roy V, Chang NH, Cai Y, Bonventi G, Wither J (2005) Aberrant IgM signaling promotes survival of transitional T1 B cells and prevents tolerance induction in lupus-prone New Zealand black mice. J Immunol 175(11):7363–7371PubMedCrossRef
94.
go back to reference Giltiay NV, Chappell CP, Sun X, Kolhatkar N, Teal TH, Wiedeman AE, Kim J, Tanaka L, Buechler MB, Hamerman JA, Imanishi-Kari T, Clark EA, Elkon KB (2013) Overexpression of TLR7 promotes cell-intrinsic expansion and autoantibody production by transitional T1 B cells. J Exp Med 210(12):2773–2789. doi:10.1084/jem.20122798 PubMedPubMedCentralCrossRef Giltiay NV, Chappell CP, Sun X, Kolhatkar N, Teal TH, Wiedeman AE, Kim J, Tanaka L, Buechler MB, Hamerman JA, Imanishi-Kari T, Clark EA, Elkon KB (2013) Overexpression of TLR7 promotes cell-intrinsic expansion and autoantibody production by transitional T1 B cells. J Exp Med 210(12):2773–2789. doi:10.​1084/​jem.​20122798 PubMedPubMedCentralCrossRef
95.
go back to reference Kumar KR, Li L, Yan M, Bhaskarabhatla M, Mobley AB, Nguyen C, Mooney JM, Schatzle JD, Wakeland EK, Mohan C (2006) Regulation of B cell tolerance by the lupus susceptibility gene Ly108. Science 312(5780):1665–1669. doi:10.1126/science.1125893 PubMedCrossRef Kumar KR, Li L, Yan M, Bhaskarabhatla M, Mobley AB, Nguyen C, Mooney JM, Schatzle JD, Wakeland EK, Mohan C (2006) Regulation of B cell tolerance by the lupus susceptibility gene Ly108. Science 312(5780):1665–1669. doi:10.​1126/​science.​1125893 PubMedCrossRef
96.
go back to reference Batten M, Groom J, Cachero TG, Qian F, Schneider P, Tschopp J, Browning JL, Mackay F (2000) BAFF mediates survival of peripheral immature B lymphocytes. J Exp Med 192(10):1453–1466PubMedPubMedCentralCrossRef Batten M, Groom J, Cachero TG, Qian F, Schneider P, Tschopp J, Browning JL, Mackay F (2000) BAFF mediates survival of peripheral immature B lymphocytes. J Exp Med 192(10):1453–1466PubMedPubMedCentralCrossRef
97.
go back to reference Teague BN, Pan Y, Mudd PA, Nakken B, Zhang Q, Szodoray P, Kim-Howard X, Wilson PC, Farris AD (2007) Cutting edge: transitional T3 B cells do not give rise to mature B cells, have undergone selection, and are reduced in murine lupus. J Immunol 178(12):7511–7515PubMedCrossRef Teague BN, Pan Y, Mudd PA, Nakken B, Zhang Q, Szodoray P, Kim-Howard X, Wilson PC, Farris AD (2007) Cutting edge: transitional T3 B cells do not give rise to mature B cells, have undergone selection, and are reduced in murine lupus. J Immunol 178(12):7511–7515PubMedCrossRef
98.
99.
go back to reference Mumtaz IM, Hoyer BF, Panne D, Moser K, Winter O, Cheng QY, Yoshida T, Burmester GR, Radbruch A, Manz RA, Hiepe F (2012) Bone marrow of NZB/W mice is the major site for plasma cells resistant to dexamethasone and cyclophosphamide: implications for the treatment of autoimmunity. J Autoimmun 39(3):180–188. doi:10.1016/j.jaut.2012.05.010 PubMedCrossRef Mumtaz IM, Hoyer BF, Panne D, Moser K, Winter O, Cheng QY, Yoshida T, Burmester GR, Radbruch A, Manz RA, Hiepe F (2012) Bone marrow of NZB/W mice is the major site for plasma cells resistant to dexamethasone and cyclophosphamide: implications for the treatment of autoimmunity. J Autoimmun 39(3):180–188. doi:10.​1016/​j.​jaut.​2012.​05.​010 PubMedCrossRef
101.
go back to reference Neubert K, Meister S, Moser K, Weisel F, Maseda D, Amann K, Wiethe C, Winkler TH, Kalden JR, Manz RA, Voll RE (2008) The proteasome inhibitor bortezomib depletes plasma cells and protects mice with lupus-like disease from nephritis. Nat Med 14(7):748–755. doi:10.1038/nm1763 PubMedCrossRef Neubert K, Meister S, Moser K, Weisel F, Maseda D, Amann K, Wiethe C, Winkler TH, Kalden JR, Manz RA, Voll RE (2008) The proteasome inhibitor bortezomib depletes plasma cells and protects mice with lupus-like disease from nephritis. Nat Med 14(7):748–755. doi:10.​1038/​nm1763 PubMedCrossRef
102.
103.
go back to reference Taddeo A, Khodadadi L, Voigt C, Mumtaz IM, Cheng Q, Moser K, Alexander T, Manz RA, Radbruch A, Hiepe F, Hoyer BF (2015) Long-lived plasma cells are early and constantly generated in New Zealand black/New Zealand White F1 mice and their therapeutic depletion requires a combined targeting of autoreactive plasma cells and their precursors. Arthritis research & therapy 17:39. doi:10.1186/s13075-015-0551-3 CrossRef Taddeo A, Khodadadi L, Voigt C, Mumtaz IM, Cheng Q, Moser K, Alexander T, Manz RA, Radbruch A, Hiepe F, Hoyer BF (2015) Long-lived plasma cells are early and constantly generated in New Zealand black/New Zealand White F1 mice and their therapeutic depletion requires a combined targeting of autoreactive plasma cells and their precursors. Arthritis research & therapy 17:39. doi:10.​1186/​s13075-015-0551-3 CrossRef
104.
go back to reference Seret G, Hanrotel C, Bendaoud B, Le Meur Y, Renaudineau Y (2013) Homozygous FCGR3A-158F mutation is associated with delayed B-cell depletion following rituximab but with preserved efficacy in a patient with refractory lupus nephritis. Clin Kidney J 6(1):74–76. doi:10.1093/ckj/sfs162 PubMedCrossRef Seret G, Hanrotel C, Bendaoud B, Le Meur Y, Renaudineau Y (2013) Homozygous FCGR3A-158F mutation is associated with delayed B-cell depletion following rituximab but with preserved efficacy in a patient with refractory lupus nephritis. Clin Kidney J 6(1):74–76. doi:10.​1093/​ckj/​sfs162 PubMedCrossRef
105.
go back to reference Devauchelle-Pensec V, Morvan J, Rat AC, Jousse-Joulin S, Pennec Y, Pers JO, Jamin C, Renaudineau Y, Quintin-Roue I, Cochener B, Youinou P, Saraux A (2011) Effects of rituximab therapy on quality of life in patients with primary Sjogren’s syndrome. Clin Exp Rheumatol 29(1):6–12PubMed Devauchelle-Pensec V, Morvan J, Rat AC, Jousse-Joulin S, Pennec Y, Pers JO, Jamin C, Renaudineau Y, Quintin-Roue I, Cochener B, Youinou P, Saraux A (2011) Effects of rituximab therapy on quality of life in patients with primary Sjogren’s syndrome. Clin Exp Rheumatol 29(1):6–12PubMed
107.
go back to reference Jousse-Joulin S, Devauchelle-Pensec V, Morvan J, Guias B, Pennec Y, Pers JO, Daridon C, Jamin C, Renaudineau Y, Roue IQ, Cochener B, Bressollette L, Youinou P, Saraux A (2007) Ultrasound assessment of salivary glands in patients with primary Sjogren’s syndrome treated with rituximab: quantitative and Doppler waveform analysis. Biologics : targets & therapy 1(3):311–319 Jousse-Joulin S, Devauchelle-Pensec V, Morvan J, Guias B, Pennec Y, Pers JO, Daridon C, Jamin C, Renaudineau Y, Roue IQ, Cochener B, Bressollette L, Youinou P, Saraux A (2007) Ultrasound assessment of salivary glands in patients with primary Sjogren’s syndrome treated with rituximab: quantitative and Doppler waveform analysis. Biologics : targets & therapy 1(3):311–319
108.
go back to reference Devauchelle-Pensec V, Pennec Y, Morvan J, Pers JO, Daridon C, Jousse-Joulin S, Roudaut A, Jamin C, Renaudineau Y, Roue IQ, Cochener B, Youinou P, Saraux A (2007) Improvement of Sjogren’s syndrome after two infusions of rituximab (anti-CD20). Arthritis & Rheumatism 57(2):310–317. doi:10.1002/art.22536 CrossRef Devauchelle-Pensec V, Pennec Y, Morvan J, Pers JO, Daridon C, Jousse-Joulin S, Roudaut A, Jamin C, Renaudineau Y, Roue IQ, Cochener B, Youinou P, Saraux A (2007) Improvement of Sjogren’s syndrome after two infusions of rituximab (anti-CD20). Arthritis & Rheumatism 57(2):310–317. doi:10.​1002/​art.​22536 CrossRef
109.
go back to reference Pers JO, Devauchelle V, Daridon C, Bendaoud B, Le Berre R, Bordron A, Hutin P, Renaudineau Y, Dueymes M, Loisel S, Berthou C, Saraux A, Youinou P (2007) BAFF-modulated repopulation of B lymphocytes in the blood and salivary glands of rituximab-treated patients with Sjogren’s syndrome. Arthritis & Rheumatism 56(5):1464–1477. doi:10.1002/art.22603 CrossRef Pers JO, Devauchelle V, Daridon C, Bendaoud B, Le Berre R, Bordron A, Hutin P, Renaudineau Y, Dueymes M, Loisel S, Berthou C, Saraux A, Youinou P (2007) BAFF-modulated repopulation of B lymphocytes in the blood and salivary glands of rituximab-treated patients with Sjogren’s syndrome. Arthritis & Rheumatism 56(5):1464–1477. doi:10.​1002/​art.​22603 CrossRef
110.
go back to reference Mahevas M, Patin P, Huetz F, Descatoire M, Cagnard N, Bole-Feysot C, Le Gallou S, Khellaf M, Fain O, Boutboul D, Galicier L, Ebbo M, Lambotte O, Hamidou M, Bierling P, Godeau B, Michel M, Weill JC, Reynaud CA (2013) B cell depletion in immune thrombocytopenia reveals splenic long-lived plasma cells. J Clin Invest 123(1):432–442. doi:10.1172/JCI65689 PubMedCrossRef Mahevas M, Patin P, Huetz F, Descatoire M, Cagnard N, Bole-Feysot C, Le Gallou S, Khellaf M, Fain O, Boutboul D, Galicier L, Ebbo M, Lambotte O, Hamidou M, Bierling P, Godeau B, Michel M, Weill JC, Reynaud CA (2013) B cell depletion in immune thrombocytopenia reveals splenic long-lived plasma cells. J Clin Invest 123(1):432–442. doi:10.​1172/​JCI65689 PubMedCrossRef
111.
go back to reference Mahevas M, Michel M, Vingert B, Moroch J, Boutboul D, Audia S, Cagnard N, Ripa J, Menard C, Tarte K, Megret J, Le Gallou S, Patin P, Thai L, Galicier L, Bonnotte B, Godeau B, Noizat-Pirenne F, Weill JC, Reynaud CA (2015) Emergence of long-lived autoreactive plasma cells in the spleen of primary warm auto-immune hemolytic anemia patients treated with rituximab. J Autoimmun 62:22–30. doi:10.1016/j.jaut.2015.05.006 PubMedCrossRef Mahevas M, Michel M, Vingert B, Moroch J, Boutboul D, Audia S, Cagnard N, Ripa J, Menard C, Tarte K, Megret J, Le Gallou S, Patin P, Thai L, Galicier L, Bonnotte B, Godeau B, Noizat-Pirenne F, Weill JC, Reynaud CA (2015) Emergence of long-lived autoreactive plasma cells in the spleen of primary warm auto-immune hemolytic anemia patients treated with rituximab. J Autoimmun 62:22–30. doi:10.​1016/​j.​jaut.​2015.​05.​006 PubMedCrossRef
112.
go back to reference Morell M, Varela N, Maranon C (2017) Myeloid populations are altered in systemic autoimmune diseases. Clinical reviews in allergy & immunology Morell M, Varela N, Maranon C (2017) Myeloid populations are altered in systemic autoimmune diseases. Clinical reviews in allergy & immunology
114.
116.
go back to reference Celhar T, Hopkins R, Thornhill SI, De Magalhaes R, Hwang SH, Lee HY, Yasuga H, Jones LA, Casco J, Lee B, Thamboo TP, Zhou XJ, Poidinger M, Connolly JE, Wakeland EK, Fairhurst AM (2015) RNA sensing by conventional dendritic cells is central to the development of lupus nephritis. Proc Natl Acad Sci U S A 112(45):E6195–E6204. doi:10.1073/pnas.1507052112 PubMedPubMedCentralCrossRef Celhar T, Hopkins R, Thornhill SI, De Magalhaes R, Hwang SH, Lee HY, Yasuga H, Jones LA, Casco J, Lee B, Thamboo TP, Zhou XJ, Poidinger M, Connolly JE, Wakeland EK, Fairhurst AM (2015) RNA sensing by conventional dendritic cells is central to the development of lupus nephritis. Proc Natl Acad Sci U S A 112(45):E6195–E6204. doi:10.​1073/​pnas.​1507052112 PubMedPubMedCentralCrossRef
118.
go back to reference Sahu R, Bethunaickan R, Singh S, Davidson A (2014) Structure and function of renal macrophages and dendritic cells from lupus-prone mice. Arthritis & Rheumatology 66(6):1596–1607. doi:10.1002/art.38410 CrossRef Sahu R, Bethunaickan R, Singh S, Davidson A (2014) Structure and function of renal macrophages and dendritic cells from lupus-prone mice. Arthritis & Rheumatology 66(6):1596–1607. doi:10.​1002/​art.​38410 CrossRef
Metadata
Title
Dysregulated Lymphoid Cell Populations in Mouse Models of Systemic Lupus Erythematosus
Authors
Aurélie De Groof
Patrice Hémon
Olivier Mignen
Jacques-Olivier Pers
Edward K. Wakeland
Yves Renaudineau
Bernard R. Lauwerys
Publication date
01-10-2017
Publisher
Springer US
Published in
Clinical Reviews in Allergy & Immunology / Issue 2/2017
Print ISSN: 1080-0549
Electronic ISSN: 1559-0267
DOI
https://doi.org/10.1007/s12016-017-8605-8

Other articles of this Issue 2/2017

Clinical Reviews in Allergy & Immunology 2/2017 Go to the issue
Live Webinar | 27-06-2024 | 18:00 (CEST)

Keynote webinar | Spotlight on medication adherence

Live: Thursday 27th June 2024, 18:00-19:30 (CEST)

WHO estimates that half of all patients worldwide are non-adherent to their prescribed medication. The consequences of poor adherence can be catastrophic, on both the individual and population level.

Join our expert panel to discover why you need to understand the drivers of non-adherence in your patients, and how you can optimize medication adherence in your clinics to drastically improve patient outcomes.

Prof. Kevin Dolgin
Prof. Florian Limbourg
Prof. Anoop Chauhan
Developed by: Springer Medicine
Obesity Clinical Trial Summary

At a glance: The STEP trials

A round-up of the STEP phase 3 clinical trials evaluating semaglutide for weight loss in people with overweight or obesity.

Developed by: Springer Medicine