1
|
Bu S, Liu M, Yang L, Lee P, Miller H, Park CS, Byazrova M, Filatov A, Benlagha K, Gaber T, Buttgereit F, Gong Q, Zhai Z, Liu C. The function of T cells in immune thrombocytopenia. Front Immunol 2025; 16:1499014. [PMID: 40061938 PMCID: PMC11885273 DOI: 10.3389/fimmu.2025.1499014] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Accepted: 01/20/2025] [Indexed: 05/13/2025] Open
Abstract
Immune thrombocytopenia (ITP) is an autoimmune disease, characterized by increased bleeding due to a reduced platelet count. The pathogenesis of ITP is very complex and involves autoantibody production and T-cell-mediated immune abnormalities. An imbalance of effector and regulatory CD4+ T cells and the breach of tolerance primarily cause ITP, leading to the dysfunctional development of autoreactive Th cells (including Th1, Th2, and Th17 cells) and Tregs. The loss of auto-platelet antigen tolerance in ITP results in autoantibody- and cytotoxic T-cell-mediated platelet clearance. T-cell-related genetic risk factors significantly influence the development and progression of this disease. New therapies targeting T cells have emerged as potentially effective cures for this disease. This review summarizes the role of T cells in ITP.
Collapse
Affiliation(s)
- Siyuan Bu
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Min Liu
- Department of Rheumatology and Clinical Immunology, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität Zu Berlin, Berlin, Germany
- German Rheumatism Research Centre (DRFZ) Berlin, Institute of the Leibniz Association, Berlin, Germany
| | - Lu Yang
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Pamela Lee
- Department of Paediatrics and Adolescent Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Heather Miller
- Cytek Biosciences, R&D Clinical Reagents, Fremont, CA, United States
| | - Chan-Sik Park
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Maria Byazrova
- Laboratory of Immunochemistry, National Research Center Institute of Immunology, Federal Medical Biological Agency of Russia, Moscow, Russia
| | - Alexander Filatov
- Laboratory of Immunochemistry, National Research Center Institute of Immunology, Federal Medical Biological Agency of Russia, Moscow, Russia
| | - Kamel Benlagha
- Institut de Recherche Saint-Louis, Université de Paris, Paris, France
| | - Timo Gaber
- Department of Rheumatology and Clinical Immunology, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität Zu Berlin, Berlin, Germany
- German Rheumatism Research Centre (DRFZ) Berlin, Institute of the Leibniz Association, Berlin, Germany
| | - Frank Buttgereit
- Department of Rheumatology and Clinical Immunology, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität Zu Berlin, Berlin, Germany
- German Rheumatism Research Centre (DRFZ) Berlin, Institute of the Leibniz Association, Berlin, Germany
| | - Quan Gong
- Department of Immunology, School of Medicine, Yangtze University, Jingzhou, Hubei, China
| | - Zhimin Zhai
- Department of Hematology, The Second Hospital of Anhui Medical University, Hefei, China
| | - Chaohong Liu
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, Hubei, China
| |
Collapse
|
2
|
Li LL, Luan ZQ, Tan Y, Wang H, Yu XJ, Qu Z, Yu F, Chen M. Anti-complement factor H (CFH) autoantibodies could delay pristane-induced lupus nephritis. Immunol Res 2023; 71:849-859. [PMID: 37322353 PMCID: PMC10667379 DOI: 10.1007/s12026-023-09396-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Accepted: 05/12/2023] [Indexed: 06/17/2023]
Abstract
PURPOSE Anti-complement factor H (CFH) autoantibodies could be detected in lupus and its significance remained to be elucidated. Herein, we aimed to explore the roles of anti-CFH autoantibodies based on pristane-induced lupus mice. METHODS Twenty-four female Balb/c mice were randomly divided into four groups, with one group injected with pristane (pristane group), one group with pristane and then human CFH (hCFH) (pristane-CFH group) 3 times, and the other two as vertical controls, PBS group and PBS-CFH group. Histopathological analysis was performed six months after pristane administration. Levels of hCFH, anti-CFH autoantibodies and anti-dsDNA antibody were detected. Murine IgG (mIgG) were purified and cross-reactivity, epitopes, subclasses and functional analysis were further evaluated in vitro. RESULTS Immunization with hCFH and subsequent development of anti-CFH autoantibodies significantly attenuated nephritis of pristane-induced lupus, including lower levels of urinary protein and serum creatinine, decreased levels of serum anti-dsDNA antibody, greatly ameliorated renal histopathologic damage, decreased IgG, complements (C1q, C3) deposits and lower inflammatory factor (IL-6) expression in glomerulus. Furthermore, the purified mIgG (contained anti-CFH autoantibodies) could recognize both hCFH and murine CFH, and the epitopes were predominantly located in hCFH short consensus repeats (SCRs) 1-4, 7 and 11-14. The IgG subclasses were predominant IgG1. The autoantibodies could enhance the binding between hCFH and C3b, and increase factor I mediated-C3b lysis in vitro. CONCLUSION Our results suggested that anti-CFH autoantibodies could attenuate pristane-induced lupus nephritis by increasing bio-functions of CFH on regulating complement activation and controlling inflammation.
Collapse
Affiliation(s)
- Lin-Lin Li
- Renal Division, Department of Medicine, Peking University First Hospital, Beijing, China
- Renal Division, Henan Provincial People's Hospital, Zhengzhou, China
| | - Zhong-Qiu Luan
- Department of Nephrology, First Affiliated Hospital of Heilongjiang, University of Chinese Medicine, Beijing, China
| | - Ying Tan
- Renal Division, Department of Medicine, Peking University First Hospital, Beijing, China
| | - Hui Wang
- Laboratory of Electron Microscopy, Pathological Centre, Peking University First Hospital, Beijing, China
| | - Xiao-Juan Yu
- Renal Division, Department of Medicine, Peking University First Hospital, Beijing, China
| | - Zhen Qu
- Department of Nephrology, Peking University International Hospital, Beijing, China.
| | - Feng Yu
- Department of Nephrology, Peking University International Hospital, Beijing, China
| | - Min Chen
- Renal Division, Department of Medicine, Peking University First Hospital, Beijing, China
| |
Collapse
|
3
|
Akiyama Y, Harada K, Miyakawa J, Kreder KJ, O’Donnell MA, Daichi M, Katoh H, Hori M, Owari K, Futami K, Ishikawa S, Ushiku T, Kume H, Homma Y, Luo Y. Th1/17 polarization and potential treatment by an anti-interferon-γ DNA aptamer in Hunner-type interstitial cystitis. iScience 2023; 26:108262. [PMID: 38026177 PMCID: PMC10663743 DOI: 10.1016/j.isci.2023.108262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 07/03/2023] [Accepted: 10/17/2023] [Indexed: 12/01/2023] Open
Abstract
Hunner-type interstitial cystitis (HIC) is a rare, enigmatic inflammatory disease of the urinary bladder with no curative treatments. In this study, we aimed to characterize the unique cellular and immunological factors specifically involved in HIC by comparing with cystitis induced by Mycobacterium bovis bacillus Calmette-Guérin, which presents similar clinicopathological features to HIC. Here, we show that T helper 1/17 +polarized immune responses accompanied by prominent overexpression of interferon (IFN)-γ, enhanced cGAS-STING cytosolic DNA sensing pathway, and increased plasma cell infiltration are the characteristic inflammatory features in HIC bladder. Further, we developed a mouse anti-IFN-γ DNA aptamer and observed that the intravesical instillation of the aptamer significantly ameliorated bladder inflammation, pelvic pain and voiding dysfunction in a recently developed murine HIC model with little migration into the blood. Our study provides the plausible basis for the clinical translation of the anti-IFN-γ DNA aptamer in the treatment of human HIC.
Collapse
Affiliation(s)
- Yoshiyuki Akiyama
- Department of Urology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- Department of Urology, University of Iowa, Iowa City, IA, USA
| | | | - Jimpei Miyakawa
- Department of Urology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Karl J. Kreder
- Department of Urology, University of Iowa, Iowa City, IA, USA
| | | | - Maeda Daichi
- Department of Molecular and Cellular Pathology, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Hiroto Katoh
- Department of Preventive Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | | | | | | | - Shumpei Ishikawa
- Department of Preventive Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Tetsuo Ushiku
- Department of Pathology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Haruki Kume
- Department of Urology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yukio Homma
- Department of Urology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- Department of Interstitial Cystitis Medicine, Faculty of Medicine, Kyorin University, Tokyo, Japan
| | - Yi Luo
- Department of Urology, University of Iowa, Iowa City, IA, USA
| |
Collapse
|
4
|
Association of the STAT4 Gene rs7574865 Polymorphism with IFN-γ Levels in Patients with Systemic Lupus Erythematosus. Genes (Basel) 2023; 14:genes14030537. [PMID: 36980810 PMCID: PMC10048585 DOI: 10.3390/genes14030537] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 02/13/2023] [Accepted: 02/17/2023] [Indexed: 02/24/2023] Open
Abstract
STAT4 plays an important role in disease activity in SLE patients. STAT4 particles have the capacity to activate the transcription of genes associated with the production of TH1 and Th17 lymphocytes, with a greater predominance on the production of IFN-γ and IL-17A. The presence of variants in STAT4 genes has a major impact on the generation of autoimmunity. However, there are few studies evaluating the impact of these variants on the production of proinflammatory cytokines such as IFN-γ and IL-17A. Methods—A case–control study was carried out with 206 Mexican mestizo patients residing in Western Mexico with a diagnosis of SLE and a group of 80 patients without autoimmune diseases was captured to determine the cut-off point for high IFN-γ levels. In this study, SLE patients with high IFN-γ levels were considered as cases (cut-off > 15.6 pg/mL), and SLE patients with normal IFN-γ levels were considered as controls (cut-off ≤ 15.6 pg/mL). Disease activity was identified from the systemic lupus erythematosus disease activity index (SLEDAI). For the determination of levels of cytokines IFN-γ, IL-12, and IL17A, commercial ELISA kits were used. Genotyping of STAT4 rs7574865 (G > T) was performed by quantitative polymerase chain reaction (qPCR) using TaqMan probes. Results—The patients with SLE had a median age of 45 years with a range of disease duration from 4 years to 18 years; 45.6% were identified as having disease activity. In this sample, we identified a high IFN-γ prevalence of 35.4%. The levels of IFN-γ were higher in the patients with genotype TT than GG. We found that TT genotype conferred a higher risk of high IFN-γ when compared to the GG and GT genotypes. Conclusions—In this study, we identified that the polymorphic genotype TT of the STAT4 gene rs7574865 polymorphism is associated with increased levels of IFN-γ. However, its strength of association was weak, so complementary studies are needed to evaluate its impact on SLE patients.
Collapse
|
5
|
Raveney BJE, El‐Darawish Y, Sato W, Arinuma Y, Yamaoka K, Hori S, Yamamura T, Oki S. Neuropilin-1 (NRP1) expression distinguishes self-reactive helper T cells in systemic autoimmune disease. EMBO Mol Med 2022; 14:e15864. [PMID: 36069030 PMCID: PMC9549730 DOI: 10.15252/emmm.202215864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 08/08/2022] [Accepted: 08/15/2022] [Indexed: 02/05/2023] Open
Abstract
Pathogenic T helper cells (Th cells) that respond to self-antigen cannot be easily distinguished from beneficial Th cells. These cells can generate systemic autoimmune disease in response to widely expressed self-antigens. In this study, we have identified neuropilin-1 (NRP1) as a cell surface marker of self-reactive Th cells. NRP1+ Th cells, absent in non-regulatory T cell subsets in normal mice, appeared in models of systemic autoimmune disease and strongly correlated with disease symptoms. NRP1+ Th cells were greatly reduced in Nr4a2 cKO mice, which have reduced self-reactive responses but showed normal responses against exogenous antigens. Transfer of NRP1+ Th cells was sufficient to initiate or accelerate systemic autoimmune disease, and targeting NRP1-expressing Th cells therapeutically ameliorated SLE-like autoimmune symptoms in BXSB-Yaa mice. Peripheral NRP1+ Th cells were significantly increased in human SLE patients. Our data suggest that self-reactive Th cells can be phenotypically distinguished within the Th cell pool. These findings offer a novel approach to identify self-reactive Th cells and target them to treat systemic autoimmune disease.
Collapse
Affiliation(s)
- Ben JE Raveney
- Department of ImmunologyNational Institute of NeuroscienceTokyoJapan
| | - Yosif El‐Darawish
- Department of ImmunologyNational Institute of NeuroscienceTokyoJapan
| | - Wakiro Sato
- Department of ImmunologyNational Institute of NeuroscienceTokyoJapan
| | - Yoshiyuki Arinuma
- Department of Rheumatology and Infectious DiseasesKitasato University School of MedicineSagamiharaJapan
| | - Kunihiro Yamaoka
- Department of Rheumatology and Infectious DiseasesKitasato University School of MedicineSagamiharaJapan
| | - Shohei Hori
- Laboratory for Immunology and MicrobiologyGraduate School of Pharmaceutical Sciences, The University of TokyoTokyoJapan
| | - Takashi Yamamura
- Department of ImmunologyNational Institute of NeuroscienceTokyoJapan
| | - Shinji Oki
- Department of ImmunologyNational Institute of NeuroscienceTokyoJapan
| |
Collapse
|
6
|
Ning W, Cheng D, Howe PH, Bian C, Kamen DL, Luo Z, Fu X, Ogunrinde E, Yang L, Wang X, Li QZ, Oates J, Zhang W, White D, Wan Z, Gilkeson GS, Jiang W. Staphylococcus aureus peptidoglycan (PGN) induces pathogenic autoantibody production via autoreactive B cell receptor clonal selection, implications in systemic lupus erythematosus. J Autoimmun 2022; 131:102860. [PMID: 35810689 PMCID: PMC9397544 DOI: 10.1016/j.jaut.2022.102860] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 06/24/2022] [Accepted: 06/26/2022] [Indexed: 10/17/2022]
Abstract
OBJECTIVES There is an intricate interplay between the microbiome and the immune response impacting development of normal immunity and autoimmunity. However, we do not fully understand how the microbiome affects production of natural-like and pathogenic autoantibodies. Peptidoglycan (PGN) is a component of the bacterial cell wall which is highly antigenic. PGNs from different bacteria can differ in their immune regulatory activities. METHODS C57BL/6 and MRL/lpr mice were intraperitoneally injected with saline or PGN from Staphylococcus aureus or Bacillus subtilis. Spleen anti-double-stranded DNA (dsDNA) IgG + B cells were sorted for B-cell receptor sequencing. Serum autoantibody levels and kidney damage were analyzed. Further, the association between plasma S. aureus translocation and systemic lupus erythematosus (SLE) pathogenesis was assessed in women. RESULTS Administration of B. subtilis PGN induced natural-like anti-dsDNA autoantibodies (e.g., IgM, short lived IgG response, and no tissue damage), whereas S. aureus PGN induced pathogenic anti-dsDNA autoantibodies (e.g., prolonged IgG production, low IgM, autoantibody-mediated kidney damage) in C57BL/6 and/or MRL/lpr mice. However, serum total IgG did not differ. S. aureus PGN induced antibodies with reduced clonality and greater hypermutation of IGHV3-74 in splenic anti-dsDNA IgG + B cells from C57BL/6 mice. Further, S. aureus PGN promoted IgG class switch recombination via toll-like receptor 2. Plasma S. aureus DNA levels were increased in women with SLE versus control women and correlated with levels of lupus-related autoantibodies and renal involvement. CONCLUSIONS S. aureus PGN induces pathogenic autoantibody production, whereas B. subtilis PGN drives production of natural nonpathogenic autoantibodies.
Collapse
Affiliation(s)
- Wangbin Ning
- Department of Microbiology and Immunology, Medical University of South Carolina, 173 Ashley Avenue, BSB208D, Charleston, SC, 29425, USA; Department of Rheumatology and Immunology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Da Cheng
- Department of Microbiology and Immunology, Medical University of South Carolina, 173 Ashley Avenue, BSB208D, Charleston, SC, 29425, USA; Department of Infectious Disease, Xiangya Hospital, Central South University, Changsha, China
| | - Philip H Howe
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, SC, USA
| | - Chuanxiu Bian
- Department of Microbiology and Immunology, Medical University of South Carolina, 173 Ashley Avenue, BSB208D, Charleston, SC, 29425, USA
| | - Diane L Kamen
- Division of Rheumatology and Immunology, Department of Medicine, Medical University of South Carolina, 114 Doughty Street, Strom Thurmond Research Building Room 416, Charleston, SC, 29403, USA
| | - Zhenwu Luo
- Department of Microbiology and Immunology, Medical University of South Carolina, 173 Ashley Avenue, BSB208D, Charleston, SC, 29425, USA
| | - Xiaoyu Fu
- Department of Microbiology and Immunology, Medical University of South Carolina, 173 Ashley Avenue, BSB208D, Charleston, SC, 29425, USA; Department of Infectious Disease, Xiangya Hospital, Central South University, Changsha, China
| | - Elizabeth Ogunrinde
- Department of Microbiology and Immunology, Medical University of South Carolina, 173 Ashley Avenue, BSB208D, Charleston, SC, 29425, USA
| | - Liuqing Yang
- Department of Microbiology and Immunology, Medical University of South Carolina, 173 Ashley Avenue, BSB208D, Charleston, SC, 29425, USA; The Third People's Hospital of Shenzhen, Guangdong, China
| | - Xu Wang
- Department of Microbiology and Immunology, Medical University of South Carolina, 173 Ashley Avenue, BSB208D, Charleston, SC, 29425, USA; Department of Urology, Capital Medical University Affiliated XuanWu Hospital, Beijing, China
| | - Quan-Zhen Li
- Department of Immunology and Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Jim Oates
- Division of Rheumatology and Immunology, Department of Medicine, Medical University of South Carolina, 114 Doughty Street, Strom Thurmond Research Building Room 416, Charleston, SC, 29403, USA; Ralph H. Johnson VA Medical Center, Charleston, SC, USA
| | - Weiru Zhang
- Department of Rheumatology and Immunology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - David White
- Department of Otolaryngology, Medical University of South Carolina, Charleston, SC, USA
| | - Zhuang Wan
- Department of Microbiology and Immunology, Medical University of South Carolina, 173 Ashley Avenue, BSB208D, Charleston, SC, 29425, USA
| | - Gary S Gilkeson
- Division of Rheumatology and Immunology, Department of Medicine, Medical University of South Carolina, 114 Doughty Street, Strom Thurmond Research Building Room 416, Charleston, SC, 29403, USA; Ralph H. Johnson VA Medical Center, Charleston, SC, USA.
| | - Wei Jiang
- Department of Microbiology and Immunology, Medical University of South Carolina, 173 Ashley Avenue, BSB208D, Charleston, SC, 29425, USA; Ralph H. Johnson VA Medical Center, Charleston, SC, USA; Division of Infectious Diseases, Department of Medicine, Medical University of South Carolina, Charleston, SC, USA.
| |
Collapse
|
7
|
Ferretti C, Horwitz DA, Bickerton S, La Cava A. Nanoparticle-mediated Delivery of IL-2 To T Follicular Helper Cells Protects BDF1 Mice from Lupus-like Disease. RHEUMATOLOGY AND IMMUNOLOGY RESEARCH 2021; 2:185-193. [PMID: 36465067 PMCID: PMC9524795 DOI: 10.2478/rir-2021-0024] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Accepted: 09/17/2021] [Indexed: 05/24/2023]
Abstract
We recently reported that poly lactic-co-glycolic acid (PLGA) nanoparticles (NPs) loaded with interleukin (IL)-2 and targeted to T cells inhibited the development of lupus-like disease in BDF1 mice by inducing functional T regulatory cells (Tregs). Here we show that the protection from disease and the extended survival of BDF1 mice provided by IL-2-loaded NPs targeted to T cells is not only due to an induction of Tregs but also contributed by an inhibition of T follicular helper (TFH) cells. These results identify a dual protective activity of IL-2 in the control of lupus autoimmunity, namely the inhibition of effector TFH cells, in addition to the previously known induction of Tregs. This newly recognized activity of IL-2 delivered by NPs can help better explain the beneficial effects of low-dose IL-2 immunotherapy in systemic lupus erythematosus (SLE), and might be considered as a new strategy to slow disease progression and improve outcomes in lupus patients.
Collapse
Affiliation(s)
- Concetta Ferretti
- Department of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - David A. Horwitz
- Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- General Nanotherapeutics, Santa Monica, CA, USA
| | - Sean Bickerton
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - Antonio La Cava
- Department of Medicine, University of California Los Angeles, Los Angeles, CA, USA
- Dipartimento di Biochimica e. Biotecnologie Mediche, University of Naples Federico II, Naples, Italy
| |
Collapse
|
8
|
Satta N, Frias MA, Vuilleumier N, Pagano S. Humoral Immunity Against HDL Particle: A New Perspective in Cardiovascular Diseases? Curr Pharm Des 2020; 25:3128-3146. [PMID: 31470782 DOI: 10.2174/1381612825666190830164917] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Accepted: 08/24/2019] [Indexed: 01/03/2023]
Abstract
BACKGROUND Autoimmune diseases are closely associated with cardiovascular diseases (CVD). Over the last decades, the comprehension of atherosclerosis, the principal initiator of CVD, evolved from a lipidcentered disease to a predominant inflammatory and immune response-driven disease displaying features of autoimmunity against a broad range of auto-antigens, including lipoproteins. Among them, high density lipoproteins (HDL) are important actors of cholesterol transport and bear several anti-atherogenic properties, raising a growing interest as therapeutic targets to decrease atherosclerosis and CVD burden, with nevertheless rather disappointing results so far. Reflecting HDL composition complexity, autoimmune responses and autoantibodies against various HDL components have been reported. RESULTS In this review, we addressed the important complexity of humoral autoimmunity towards HDL and particularly how this autoimmune response could help improving our understanding of HDL biological implication in atherosclerosis and CVD. We also discussed several issues related to specific HDL autoantibody subclasses characteristics, including etiology, prognosis and pathological mechanisms according to Rose criteria. CONCLUSION Finally, we addressed the possible clinical value of using these antibodies not only as potential biomarkers of atherogenesis and CVD, but also as a factor potentially mitigating the benefit of HDL-raising therapies.
Collapse
Affiliation(s)
- Nathalie Satta
- Division of Laboratory Medicine, Department of Diagnostic, Geneva University Hospitals, 4 rue Gabrielle Perret-Gentil, 1205 Geneva, Switzerland.,Department of Medical Specialties, Faculty of Medicine, University of Geneva, 1 rue Michel Servet, 1211 Geneva, Switzerland
| | - Miguel A Frias
- Division of Laboratory Medicine, Department of Diagnostic, Geneva University Hospitals, 4 rue Gabrielle Perret-Gentil, 1205 Geneva, Switzerland.,Department of Medical Specialties, Faculty of Medicine, University of Geneva, 1 rue Michel Servet, 1211 Geneva, Switzerland
| | - Nicolas Vuilleumier
- Division of Laboratory Medicine, Department of Diagnostic, Geneva University Hospitals, 4 rue Gabrielle Perret-Gentil, 1205 Geneva, Switzerland.,Department of Medical Specialties, Faculty of Medicine, University of Geneva, 1 rue Michel Servet, 1211 Geneva, Switzerland
| | - Sabrina Pagano
- Division of Laboratory Medicine, Department of Diagnostic, Geneva University Hospitals, 4 rue Gabrielle Perret-Gentil, 1205 Geneva, Switzerland.,Department of Medical Specialties, Faculty of Medicine, University of Geneva, 1 rue Michel Servet, 1211 Geneva, Switzerland
| |
Collapse
|
9
|
Liu A, Ferretti C, Shi FD, Cohen IR, Quintana FJ, La Cava A. DNA Vaccination With Hsp70 Protects Against Systemic Lupus Erythematosus in (NZB × NZW)F1 Mice. Arthritis Rheumatol 2020; 72:997-1002. [PMID: 31943822 DOI: 10.1002/art.41202] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Accepted: 01/07/2020] [Indexed: 12/30/2022]
Abstract
OBJECTIVE To address whether a targeted modulation of the abnormal expression of Hsp70 and autoantibodies against this molecule in systemic lupus erythematosus can influence disease. METHODS Lupus-prone (NZB × NZW)F1 mice that had been DNA-vaccinated with plasmids encoding Hsp70 and controls were monitored for lupus disease parameters including anti-double stranded DNA (anti-dsDNA) autoantibodies and cytokines using enzyme-linked immunosorbent assay, and for kidney function and pathology. The phenotypic and numerical changes in relevant immune cells were evaluated by flow cytometry, and cell function was assessed. RESULTS Mice that had been DNA-vaccinated with Hsp70 displayed marked suppression of anti-dsDNA antibody production, reduced renal disease, and antiinflammatory responses that are associated with a significantly extended survival, compared to controls. These protective effects in Hsp70-vaccinated mice were associated with an induction of tolerogenic immune responses and an expansion of functional Treg cells. CONCLUSION DNA vaccination with Hsp70 suppresses murine lupus by inducing tolerogenic immune responses and antiinflammatory immune responses associated with reduced disease manifestations and increased mouse survival.
Collapse
Affiliation(s)
| | | | - Fu-Dong Shi
- Barrow Neurological Institute, Phoenix, Arizona
| | - Irun R Cohen
- The Weizmann Institute of Science, Rehovot, Israel
| | | | | |
Collapse
|
10
|
Wang H, Lu M, Zhai S, Wu K, Peng L, Yang J, Xia Y. ALW peptide ameliorates lupus nephritis in MRL/lpr mice. Arthritis Res Ther 2019; 21:261. [PMID: 31791413 PMCID: PMC6889545 DOI: 10.1186/s13075-019-2038-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2019] [Accepted: 10/21/2019] [Indexed: 02/06/2023] Open
Abstract
Background Lupus nephritis (LN) is a common and serious complication of systemic lupus erythematosus. Anti-double-stranded (ds) DNA immunoglobulin G (IgG) plays a pivotal role in the pathogenesis of LN. Currently, there are various therapies for patients with LN; however, most of them are associated with considerable side effects. We confirmed previously that ALW (ALWPPNLHAWVP), a 12-amino acid peptide, inhibited the binding of polyclonal anti-dsDNA antibodies to mesangial cells and isolated glomeruli in vitro. In this study, we further investigate whether the administration of ALW peptide decreases renal IgG deposition and relevant damage in MRL/lpr lupus-prone mice. Methods Forty female MRL/lpr mice were randomly divided into four groups. The mice were intravenously injected with D-form ALW peptide (ALW group), scrambled peptide (PLP group), and normal saline (NaCl group) or were not treated (blank group). The IgG deposition, the histopathologic changes, and the expressions of profibrotic factors were analyzed in the kidney of MRL/lpr mice. Results Compared with the other groups, glomerular deposition of IgG, IgG2a, IgG2b, and IgG3 was decreased in the ALW group. Moreover, ALW administration attenuated renal histopathologic changes in MRL/lpr mice, including mesangial proliferation and infiltration of inflammatory cells. Furthermore, the expressions of profibrotic cytokines, such as transforming growth factor-beta1 (TGF-β1) and platelet-derived growth factor B (PDGF-B), decreased in the serum and kidney tissue of ALW-treated mice. Conclusions Our study demonstrated that ALW peptide ameliorates the murine model of LN, possibly through inhibiting renal IgG deposition and relevant tissue inflammation and fibrosis.
Collapse
Affiliation(s)
- Huixia Wang
- Department of Dermatology, The Second Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an, 710004, China
| | - Mei Lu
- Department of Dermatology, The Second Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an, 710004, China
| | - Siyue Zhai
- Department of Dermatology, The Second Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an, 710004, China
| | - Kunyi Wu
- Core Research Laboratory, The Second Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an, China
| | - Lingling Peng
- Department of Dermatology, The Second Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an, 710004, China
| | - Jie Yang
- Department of Nephrology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710032, China.
| | - Yumin Xia
- Department of Dermatology, The Second Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an, 710004, China.
| |
Collapse
|
11
|
Muhammad Yusoff F, Wong KK, Mohd Redzwan N. Th1, Th2, and Th17 cytokines in systemic lupus erythematosus. Autoimmunity 2019; 53:8-20. [PMID: 31771364 DOI: 10.1080/08916934.2019.1693545] [Citation(s) in RCA: 84] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Systemic lupus erythematosus (SLE) is an autoimmune disease characterized by the breakdown of immune tolerance leading to excessive inflammation and tissue damage. Imbalance in the levels of cytokines represents one of the multifactorial causes of SLE pathogenesis and it contributes to disease severity. Deregulated levels of T helper type 1 (Th1), type 2 (Th2), and type 17 (Th17) cytokines have been associated with autoimmune inflammation. Growing evidence has shown deregulated levels of Th1, Th2, and Th17 cytokines in SLE patients compared to healthy controls associated with disease activity and severity. In this review, we describe and discuss the levels of Th1, Th2, and Th17 cytokines in SLE patients, and clinical trials involving Th1, Th2, and Th17 cytokines in SLE patients. In particular, with the exception of IL-2, IL-4, and TGF-β1, the levels of Th1, Th2, and Th17 cytokines are increased in SLE patients associated with disease severity. Current phase II or III studies involve therapeutic antibodies targeting IFN-α and type I IFN receptor, while low-dose IL-2 therapy is assessed in phase II clinical trials.
Collapse
Affiliation(s)
- Farhana Muhammad Yusoff
- Department of Immunology, School of Medical Sciences, Universiti Sains Malaysia, Health Campus, Kota Bharu, Malaysia
| | - Kah Keng Wong
- Department of Immunology, School of Medical Sciences, Universiti Sains Malaysia, Health Campus, Kota Bharu, Malaysia
| | - Norhanani Mohd Redzwan
- Department of Immunology, School of Medical Sciences, Universiti Sains Malaysia, Health Campus, Kota Bharu, Malaysia
| |
Collapse
|
12
|
Wang X, Xia Y. Anti-double Stranded DNA Antibodies: Origin, Pathogenicity, and Targeted Therapies. Front Immunol 2019; 10:1667. [PMID: 31379858 PMCID: PMC6650533 DOI: 10.3389/fimmu.2019.01667] [Citation(s) in RCA: 112] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Accepted: 07/03/2019] [Indexed: 01/02/2023] Open
Abstract
Systemic lupus erythematosus (SLE) is characterized by high-titer serological autoantibodies, including antibodies that bind to double-stranded DNA (dsDNA). The origin, specificity, and pathogenicity of anti-dsDNA antibodies have been studied from a wider perspective. These autoantibodies have been suggested to contribute to multiple end-organ injuries, especially to lupus nephritis, in patients with SLE. Moreover, serum levels of anti-DNA antibodies fluctuate with disease activity in patients with SLE. By directly binding to self-antigens or indirectly forming immune complexes, anti-dsDNA antibodies can accumulate in the glomerular and tubular basement membrane. These autoantibodies can also trigger the complement cascade, penetrate into living cells, modulate gene expression, and even induce profibrotic phenotypes of renal cells. In addition, the expression of suppressor of cytokine signaling 1 is reduced by anti-DNA antibodies simultaneously with upregulation of profibrotic genes. Anti-dsDNA antibodies may even participate in the pathogenesis of SLE by catalyzing hydrolysis of certain DNA molecules or peptides in cells. Recently, anti-dsDNA antibodies have been explored in greater depth as a therapeutic target in the management of SLE. A substantial amount of data indicates that blockade of pathogenic anti-dsDNA antibodies can prevent or even reverse organ damage in murine models of SLE. This review focuses on the recent research advances regarding the origin, specificity, classification, and pathogenicity of anti-dsDNA antibodies and highlights the emerging therapies associated with them.
Collapse
Affiliation(s)
- Xiaoyu Wang
- Department of Dermatology, The Second Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an, China
| | - Yumin Xia
- Department of Dermatology, The Second Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
13
|
Yamanaka A, Konishi E. Key Amino Acid Substitution for Infection-Enhancing Activity-Free Designer Dengue Vaccines. iScience 2019; 13:125-137. [PMID: 30826727 PMCID: PMC6402262 DOI: 10.1016/j.isci.2019.02.012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Revised: 02/11/2019] [Accepted: 02/13/2019] [Indexed: 11/17/2022] Open
Abstract
Dengue is a globally important disease caused by four serotypes of dengue virus. Dengue vaccine development has been hampered by antigenic cross-reactivity among serotypes, which potentially causes antibody-dependent enhancement of infection and disease severity. Here we found that a single amino acid substitution in the envelope protein at position 87 from aspartic acid to asparagine or at position 107 from leucine to phenylalanine is critical for suppressing the induction of infection-enhancing antibody in a mouse model. The site and type of amino acid substitution were determined via neutralization escape using an enhancing-activity-only monoclonal antibody that was engineered to reveal neutralizing activity. Mutated dengue type 1 DNA vaccines containing either or both amino acid substitutions induced neutralizing antibodies devoid of enhancing activity against all serotypes. The effect of substitution was further demonstrated using other serotypes and a tetravalent formulation. This finding may contribute to the development of infection-enhancing-activity-free dengue vaccines. Amino acids at E87 or E107 are critical for dengue-enhancing antibody induction Neutralization escape is useful for identifying the key types or sites of amino acids Each substitution can be applied to antigens of all four dengue serotypes A modified tetravalent DNA vaccine suppresses enhancing antibody induction in mice
Collapse
Affiliation(s)
- Atsushi Yamanaka
- BIKEN Endowed Department of Dengue Vaccine Development, Faculty of Tropical Medicine, Mahidol University, 420/6 Ratchawithi Road, Ratchathewi, Bangkok 10400, Thailand; BIKEN Endowed Department of Dengue Vaccine Development, Research Institute for Microbial Diseases, Osaka University, 3-1 Yamada-oka, Suita, Osaka 565-0871, Japan.
| | - Eiji Konishi
- BIKEN Endowed Department of Dengue Vaccine Development, Faculty of Tropical Medicine, Mahidol University, 420/6 Ratchawithi Road, Ratchathewi, Bangkok 10400, Thailand; BIKEN Endowed Department of Dengue Vaccine Development, Research Institute for Microbial Diseases, Osaka University, 3-1 Yamada-oka, Suita, Osaka 565-0871, Japan
| |
Collapse
|
14
|
Shi M, Ouyang Y, Yang M, Yang M, Zhang X, Huang W, Wang W, Wang Z, Zhang W, Chen X, Pan X, Ren H, Chen N, Xie J. IgA Nephropathy Susceptibility Loci and Disease Progression. Clin J Am Soc Nephrol 2018; 13:1330-1338. [PMID: 30042224 PMCID: PMC6140573 DOI: 10.2215/cjn.13701217] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Accepted: 06/13/2018] [Indexed: 01/06/2023]
Abstract
BACKGROUND AND OBJECTIVES At least 20 susceptibility loci of IgA nephropathy have been identified by genome-wide association studies to date. Whether these loci were associated with disease progression is unclear. DESIGN, SETTING, PARTICIPANTS, & MEASUREMENTS We enrolled 613 adult patients with IgA nephropathy for a follow-up of ≥12 months. All 20 IgA nephropathy susceptibility loci were selected and their tag single nucleotide polymorphisms (SNPs) were genotyped. After strict quality control, 16 SNPs and 517 patients with IgA nephropathy were eligible for subsequent analysis. Progression was defined as ESKD or 50% decrease in eGFR. A stepwise Cox regression analysis of all SNPs on Akaike information criterion was performed to select the best model. RESULTS A four-SNP model, rs11150612 (ITGAM-ITGAX), rs7634389 (ST6GAL1), rs2412971 (HORMAD2), and rs2856717 (HLA-DQ/DR), was selected as the best predictive model. The genetic risk score calculated on the basis of the four SNPs was independently associated with disease progression before (hazard ratio [HR], 1.65; 95% confidence interval [95% CI], 1.29 to 2.12) and after adjustment by a recently reported clinical model (HR, 1.29; 95% CI, 1.03 to 1.62) or clinical-pathologic model (HR, 1.35; 95% CI, 1.03 to 1.77). Compared with low genetic risk, patients with middle genetic risk had a 2.12-fold (95% CI, 1.33 to 3.40) increase of progression risk, whereas patients with high genetic risk had 3.61-fold (95% CI, 2.00 to 6.52) progression risk increase. In addition, incorporation of genetic risk score could potentially increase discrimination of the clinical model (c-statistic increase from 0.83 to 0.86) or the clinical-pathologic model (c-statistic increase from 0.82 to 0.85) in predicting 5-year progression risk. CONCLUSIONS The four-SNP genetic risk score was independently associated with IgA nephropathy progression and could enhance the performance of clinical and clinical-pathologic risk models.
Collapse
Affiliation(s)
- Manman Shi
- Department of Nephrology, Institute of Nephrology, Shanghai Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Ryu H, Lim H, Choi G, Park YJ, Cho M, Na H, Ahn CW, Kim YC, Kim WU, Lee SH, Chung Y. Atherogenic dyslipidemia promotes autoimmune follicular helper T cell responses via IL-27. Nat Immunol 2018; 19:583-593. [DOI: 10.1038/s41590-018-0102-6] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Accepted: 03/15/2018] [Indexed: 01/01/2023]
|
16
|
Galectin-3 deficiency drives lupus-like disease by promoting spontaneous germinal centers formation via IFN-γ. Nat Commun 2018; 9:1628. [PMID: 29691398 PMCID: PMC5915532 DOI: 10.1038/s41467-018-04063-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2017] [Accepted: 03/30/2018] [Indexed: 01/14/2023] Open
Abstract
Germinal centers (GC) are important sites for high-affinity and long-lived antibody induction. Tight regulation of GC responses is critical for maintaining self-tolerance. Here, we show that Galectin-3 (Gal-3) is involved in GC development. Compared with WT mice, Gal-3 KO mice have more GC B cells and T follicular helper cells, increased percentages of antibody-secreting cells and higher concentrations of immunoglobulins and IFN-γ in serum, and develop a lupus-like disease. IFN-γ blockade in Gal-3 KO mice reduces spontaneous GC formation, class-switch recombination, autoantibody production and renal pathology, demonstrating that IFN-γ overproduction sustains autoimmunity. The results from chimeric mice show that intrinsic Gal-3 signaling in B cells controls spontaneous GC formation. Taken together, our data provide evidence that Gal-3 acts directly on B cells to regulate GC responses via IFN-γ and implicate the potential of Gal-3 as a therapeutic target in autoimmunity. Germinal center (GC) is where B cells interact with other immune cells for optimal induction of antibody responses. Here the authors show that galectin-3 regulates GC development by modulating interferon-γ and B cell-intrinsic signaling, such that galectin-3 deficiency mice exhibit lupus-like autoimmune symptoms.
Collapse
|
17
|
Kokic V, Martinovic Kaliterna D, Radic M, Tandara L, Perkovic D. Association between vitamin D, oestradiol and interferon-gamma in female patients with inactive systemic lupus erythematosus: A cross-sectional study. J Int Med Res 2017; 46:1162-1171. [PMID: 29235391 PMCID: PMC5972245 DOI: 10.1177/0300060517734686] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Objectives To investigate possible associations between 25-hydroxyvitamin D3 (25(OH)D3), oestradiol (E2) and IFN-gamma (IFNγ) in female patients with inactive systemic lupus erythematosus (SLE). Methods Female patients with inactive SLE and age-matched healthy controls were recruited into this cross-sectional study. Serum concentrations of 25(OH)D3, E2 and IFNγ were measured by radioimmunoassay with gamma-counters and enzyme-linked immunosorbent assay. Results 36 patients and 37 controls were enrolled. In patients with SLE, the concentration of 25(OH)D3 was lower and E2 was higher compared with controls. In vitamin D deficient (i.e., 25(OH)D3≤20 ng/ml) patients, IFNγ was 150% higher compared with patients with 25(OH)D3>20 ng/ml and controls. The concentration of E2 was higher in all patients compared with controls independently of the vitamin D level. A difference was found between patients and controls in the correlation of 25(OH)D3 with E2 and a positive correlation was found between E2 and IFNγ in all participants. Conclusions Our results suggest that E2 may have a strong modulating effect on vitamin D function which is significant only at low concentration of E2.
Collapse
Affiliation(s)
- Visnja Kokic
- 1 Division of Endocrinology, Diabetes and Metabolic Disease, University Hospital of Split, School of Medicine, Split, Croatia
| | | | - Mislav Radic
- 2 Division of Rheumatology and Clinical Immunology, University Hospital of Split, Split, Croatia
| | - Leida Tandara
- 3 Department of Medical Laboratory Diagnostic, University Hospital of Split, Split, Croatia
| | - Dijana Perkovic
- 2 Division of Rheumatology and Clinical Immunology, University Hospital of Split, Split, Croatia
| |
Collapse
|
18
|
Systemic Activation of NRF2 Alleviates Lethal Autoimmune Inflammation in Scurfy Mice. Mol Cell Biol 2017; 37:MCB.00063-17. [PMID: 28507037 DOI: 10.1128/mcb.00063-17] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2017] [Accepted: 05/11/2017] [Indexed: 12/20/2022] Open
Abstract
The transcription factor NRF2 (nuclear factor [erythroid-derived 2]-like 2) plays crucial roles in the defense mechanisms against oxidative stress and mediates anti-inflammatory actions under various pathological conditions. Recent studies showed that the dysfunction of regulatory T cells (Tregs) is directly linked to the initiation and progression of various autoimmune diseases. To determine the Treg-independent impact of NRF2 activation on autoimmune inflammation, we examined scurfy (Sf) mice, which are deficient in Tregs and succumb to severe multiorgan inflammation by 4 weeks of age. We found that systemic activation of NRF2 by Keap1 (Kelch-like ECH-associated protein 1) knockdown ameliorated tissue inflammation and lethality in Sf mice. Activated T cells and their cytokine production were accordingly decreased by Keap1 knockdown. In contrast, NRF2 activation through cell lineage-specific Keap1 disruption (i.e., in T cells, myeloid cells, and dendritic cells) achieved only partial or no improvement in the inflammatory status of Sf mice. Our results indicate that systemic activation of NRF2 suppresses effector T cell activities independently of Tregs and that NRF2 activation in multiple cell lineages appears to be required for sufficient anti-inflammatory effects. This study emphasizes the possible therapeutic application of NRF2 inducers in autoimmune diseases that are accompanied by Treg dysfunction.
Collapse
|
19
|
Recombinant Lactobacillus casei expressing Clostridium perfringens toxoids α, β2, ε and β1 gives protection against Clostridium perfringens in rabbits. Vaccine 2017. [DOI: 10.1016/j.vaccine.2017.05.076] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
|
20
|
Takahashi N, Saitoh T, Gotoh N, Nitta Y, Alkebsi L, Kasamatsu T, Minato Y, Yokohama A, Tsukamoto N, Handa H, Murakami H. The cytokine polymorphisms affecting Th1/Th2 increase the susceptibility to, and severity of, chronic ITP. BMC Immunol 2017; 18:26. [PMID: 28511637 PMCID: PMC5434613 DOI: 10.1186/s12865-017-0210-3] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Accepted: 04/21/2017] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND T-helper cell type 1 (Th1) polarization in chronic immune thrombocytopenia (cITP) has been reported at the protein and mRNA levels. We evaluated the impact of Th1/Th2 cytokine and cytokine receptor functional polymorphisms on both susceptibility to, and severity of, cITP. We analysed IFN-γ + 874 T/A, IFN-γR -611G/A, IL-4 -590C/T, and IL-4Rα Q576R polymorphisms in 126 cITP patients (male/female: 34/92; median age: 47.7 years) and 202 healthy control donors. Genotyping was determined by PCR and direct sequencing. The Th1/Th2 ratio was detected in peripheral blood mononuclear cells via flow cytometry. RESULTS cITP patients had a higher frequency of the IL-4Rα 576 non-QQ genotype compared to healthy subjects (P = 0.04). cITP patients with the IFN-γ +874 non-AA genotype (high expression type) showed more severe thrombocytopenia than those with the AA genotype (P < 0.05). cITP patients had a significantly higher Th1/Th2 ratio than control patients (P < 0.01); this ratio was inversely correlated with platelet counts. Furthermore, patients with both IFN-γ +874 non-AA genotype (high expression type) and IFN-γR -611 non-AA genotype (high-function type) had a significantly higher Th1/Th2 ratio (P < 0.05). CONCLUSIONS The cytokine polymorphisms affecting Th1/Th2 increase the susceptibility to, and severity of, chronic ITP.
Collapse
MESH Headings
- Adolescent
- Adult
- Aged
- Aged, 80 and over
- Cells, Cultured
- Child
- Child, Preschool
- Chronic Disease
- Female
- Genetic Predisposition to Disease
- Genotype
- Humans
- Interferon-gamma/genetics
- Interferon-gamma/metabolism
- Interleukin-4/genetics
- Interleukin-4/metabolism
- Interleukin-4 Receptor alpha Subunit/genetics
- Interleukin-4 Receptor alpha Subunit/metabolism
- Male
- Middle Aged
- Polymorphism, Single Nucleotide
- Purpura, Thrombocytopenic, Idiopathic/genetics
- Purpura, Thrombocytopenic, Idiopathic/immunology
- Receptors, Interferon/genetics
- Receptors, Interferon/metabolism
- Th1 Cells/immunology
- Th1-Th2 Balance/genetics
- Th2 Cells/immunology
- Young Adult
- Interferon gamma Receptor
Collapse
Affiliation(s)
- Noriyuki Takahashi
- Graduate School of Health Sciences, Gunma University, 3-39-22 Showa-machi, Maebashi, Gunma 371-8514 Japan
| | - Takayuki Saitoh
- Graduate School of Health Sciences, Gunma University, 3-39-22 Showa-machi, Maebashi, Gunma 371-8514 Japan
| | - Nanami Gotoh
- Graduate School of Health Sciences, Gunma University, 3-39-22 Showa-machi, Maebashi, Gunma 371-8514 Japan
| | - Yasuhiro Nitta
- Graduate School of Health Sciences, Gunma University, 3-39-22 Showa-machi, Maebashi, Gunma 371-8514 Japan
| | - Lobna Alkebsi
- Graduate School of Health Sciences, Gunma University, 3-39-22 Showa-machi, Maebashi, Gunma 371-8514 Japan
| | - Tetsuhiro Kasamatsu
- Graduate School of Health Sciences, Gunma University, 3-39-22 Showa-machi, Maebashi, Gunma 371-8514 Japan
| | - Yusuke Minato
- Department of Virology and Preventive Medicine, Gunma University, Gunma, Japan
| | - Akihiko Yokohama
- Division of Blood Transfusion Service, Gunma University Hospital, Gunma, Japan
| | | | - Hiroshi Handa
- Department of Medicine and Clinical Science, Gunma University Graduate School of Medicine, Gunma, Japan
| | - Hirokazu Murakami
- Graduate School of Health Sciences, Gunma University, 3-39-22 Showa-machi, Maebashi, Gunma 371-8514 Japan
| |
Collapse
|
21
|
Taylor EB, Ryan MJ. Immunosuppression With Mycophenolate Mofetil Attenuates Hypertension in an Experimental Model of Autoimmune Disease. J Am Heart Assoc 2017; 6:JAHA.116.005394. [PMID: 28242635 PMCID: PMC5524041 DOI: 10.1161/jaha.116.005394] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Background Systemic lupus erythematosus (SLE) is a chronic autoimmune disorder that predominantly affects women and is associated with prevalent hypertension, renal injury, and cardiovascular disease. Immune system dysfunction is recognized as an important factor in the pathogenesis of hypertension. We recently showed that preventing autoimmunity prevents the development of hypertension in an experimental model of SLE (female NZBWF1 mice). The present study tests the hypothesis that mycophenolate mofetil (MMF), an immunosuppressive therapy used clinically to treat SLE by depleting proliferating B and T lymphocytes, can improve blood pressure control. Methods and Results Female SLE and control (NZW/LacJ) mice were treated daily for 8 weeks with 60 mg/kg MMF. Circulating CD45R+ B cells were lower in MMF‐treated SLE mice after 4 weeks of treatment, but neither CD4+ nor CD8+ T cells were reduced by MMF. Plasma anti–double‐stranded DNA IgG autoantibodies, a marker of SLE disease activity, were higher in SLE mice compared with controls and were lower in SLE mice after 8 weeks of MMF. Mean arterial pressure was elevated in SLE mice compared with controls and lower in SLE mice treated with MMF compared with vehicle‐treated SLE mice. MMF also reduced both renal injury (urinary albumin excretion and glomerulosclerosis) and the infiltration of CD45R+ B cells and CD3+CD4+ T cells in kidneys from mice with SLE. Conclusions These data suggest that MMF selectively depleted CD45R+ B cells and lowered subsequent autoantibody production, furthering the concept that autoantibodies mechanistically contribute to the pathogenesis of hypertension.
Collapse
Affiliation(s)
- Erin B Taylor
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS
| | - Michael J Ryan
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS
| |
Collapse
|
22
|
Choi JY, Seth A, Kashgarian M, Terrillon S, Fung E, Huang L, Wang LC, Craft J. Disruption of Pathogenic Cellular Networks by IL-21 Blockade Leads to Disease Amelioration in Murine Lupus. THE JOURNAL OF IMMUNOLOGY 2017; 198:2578-2588. [PMID: 28219887 DOI: 10.4049/jimmunol.1601687] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Accepted: 01/26/2017] [Indexed: 12/31/2022]
Abstract
Systemic lupus erythematosus (lupus) is characterized by autoantibody-mediated organ injury. Follicular Th (Tfh) cells orchestrate physiological germinal center (GC) B cell responses, whereas in lupus they promote aberrant GC responses with autoreactive memory B cell development and plasma cell-derived autoantibody production. IL-21, a Tfh cell-derived cytokine, provides instructional cues for GC B cell maturation, with disruption of IL-21 signaling representing a potential therapeutic strategy for autoantibody-driven diseases such as systemic lupus erythematosus. We used blockade of IL-21 to dissect the mechanisms by which this cytokine promotes autoimmunity in murine lupus. Treatment of lupus-prone B6.Sle1.Yaa mice with an anti-IL-21 blocking Ab reduced titers of autoantibodies, delayed progression of glomerulonephritis and diminished renal-infiltrating Tfh and Th1 cells, and improved overall survival. Therapy inhibited excessive accumulation of Tfh cells coexpressing IL-21 and IFN-γ, and suppressed their production of the latter cytokine, albeit while not affecting their frequency. Anti-IL-21 treatment also led to a reduction in GC B cells, CD138hi plasmablasts, IFN-γ-dependent IgG2c production, and autoantibodies, indicating that Tfh cell-derived IL-21 is critical for pathological B cell cues in lupus. Normalization of GC responses was, in part, caused by uncoupling of Tfh-B cell interactions, as evidenced by reduced expression of CD40L on Tfh cells and reduced B cell proliferation in treated mice. Our work provides mechanistic insight into the contribution of IL-21 to the pathogenesis of murine lupus, while revealing the importance of T-B cellular cross-talk in mediating autoimmunity, demonstrating that its interruption impacts both cell types leading to disease amelioration.
Collapse
Affiliation(s)
- Jin-Young Choi
- Section of Rheumatology, Department of Internal Medicine, Yale School of Medicine, New Haven, CT 06520
| | - Abhinav Seth
- Section of Rheumatology, Department of Internal Medicine, Yale School of Medicine, New Haven, CT 06520
| | | | | | - Emma Fung
- AbbVie Bioresearch Center, Worcester, MA 01605; and
| | - Lili Huang
- AbbVie Bioresearch Center, Worcester, MA 01605; and
| | - Li Chun Wang
- AbbVie Bioresearch Center, Worcester, MA 01605; and
| | - Joe Craft
- Section of Rheumatology, Department of Internal Medicine, Yale School of Medicine, New Haven, CT 06520; .,Department of Immunobiology, Yale School of Medicine, New Haven, CT 06520
| |
Collapse
|
23
|
Nishimoto T, Okazaki Y, Numajiri M, Kuwana M. Mouse immune thrombocytopenia is associated with Th1 bias and expression of activating Fcγ receptors. Int J Hematol 2016; 105:598-605. [PMID: 28028748 DOI: 10.1007/s12185-016-2172-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2016] [Revised: 12/20/2016] [Accepted: 12/21/2016] [Indexed: 12/13/2022]
Abstract
Immune thrombocytopenia (ITP) is an autoimmune disease mediated by anti-platelet autoantibodies. We recently established a mouse ITP model exhibiting regulatory T-cell (Treg) deficiency, although only one-third of the Treg-deficient mice developed ITP. To clarify mechanisms involved in the emergence of platelet-specific autoimmunity in this model, we examined the T helper (Th)-cell balance and macrophage Fcγ receptor (FcγR) expression profiles in Treg-deficient mice with and without ITP. Splenocytes from both populations of Treg-deficient mice and control BALB/c mice were subjected to flow cytometry-based analyses to evaluate Th cell subset proportions and the expression of activating and inhibitory FcγRs on macrophages. In addition, IgG subclass distribution of anti-platelet autoantibodies in splenocyte culture supernatants was determined by flow cytometry using IgG subclass-specific antibodies. Treg-deficient ITP mice exhibited a significantly higher proportion of Th1 cells than either Treg-deficient non-ITP or control mice. The predominant anti-platelet autoantibody subclasses in the ITP mice were Th1-associated IgG2a and IgG2b. Furthermore, the FcγRI/FcγRIIB expression ratio in splenic macrophages was higher in the Treg-deficient ITP than in the Treg-deficient non-ITP and control mice. In summary, Th1 polarization and macrophages' activating FcγR expression profile are associated with the development of ITP in Treg-deficient mice.
Collapse
Affiliation(s)
- Tetsuya Nishimoto
- Division of Rheumatology, Department of Internal Medicine, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Yuka Okazaki
- Department of Allergy and Rheumatology, Nippon Medical School Graduate School of Medicine, 1-1-5 Sendagi, Bunkyo-ku, Tokyo, 113-8603, Japan
| | - Miku Numajiri
- Division of Rheumatology, Department of Internal Medicine, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Masataka Kuwana
- Division of Rheumatology, Department of Internal Medicine, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan. .,Department of Allergy and Rheumatology, Nippon Medical School Graduate School of Medicine, 1-1-5 Sendagi, Bunkyo-ku, Tokyo, 113-8603, Japan.
| |
Collapse
|
24
|
Mechanisms of disease: The human N-glycome. Biochim Biophys Acta Gen Subj 2016; 1860:1574-82. [DOI: 10.1016/j.bbagen.2015.10.016] [Citation(s) in RCA: 150] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Revised: 10/03/2015] [Accepted: 10/15/2015] [Indexed: 12/20/2022]
|
25
|
Gatto M, Ghirardello A, Luisetto R, Bassi N, Fedrigo M, Valente M, Valentino S, Del Prete D, Punzi L, Doria A. Immunization with pentraxin 3 (PTX3) leads to anti-PTX3 antibody production and delayed lupus-like nephritis in NZB/NZW F1 mice. J Autoimmun 2016; 74:208-216. [PMID: 27405845 DOI: 10.1016/j.jaut.2016.07.002] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2016] [Revised: 06/28/2016] [Accepted: 07/01/2016] [Indexed: 11/30/2022]
Abstract
BACKGROUND Anti-pentraxin 3 (PTX3) antibodies were associated with the absence of lupus glomerulonephritis in humans. AIM To explore the effects of anti-PTX3 antibodies in New Zealand Black/White (NZB/NZW F1) mice and their inherent mechanisms of action. MATERIALS AND METHODS 30 NZB/NZW F1 mice were subdivided into 3 groups of 10 mice each and subcutaneously injected with PTX3, alum and PBS (group 1), alum and PBS (group 2) or PBS alone (group 3), 3 times 3 weeks apart, before development of renal disease. Mice were followed until natural death. Histological analysis and immunohistochemistry were performed on harvested kidneys. Effects of anti-PTX3 antibodies on C1q binding to immobilized PTX3-anti-PTX3 immune complexes were evaluated in vitro using human SLE sera. Qualitative characterization of human IgG anti-PTX3 was performed. RESULTS Only group 1 mice developed anti-PTX3 antibodies. Anti-dsDNA and anti-C1q antibodies appeared significantly later and at lower levels in group 1 mice vs. controls (p < 0.0001). Proteinuria-free and overall survival were significantly increased in group 1 mice vs. controls (p < 0.05 and p = 0.03, respectively). Histopathological analysis showed that glomerular and tubular PTX3 staining and renal lesions were increased in controls compared with immunized mice. Addition of human SLE sera positive for anti-PTX3 antibodies to C1q and fixed PTX3 interfered with C1q binding to PTX3-anti-PTX3 immune complexes. Qualitative characterization of human IgG anti-PTX3 showed an increased proportion of IgG4. CONCLUSIONS Anti-PTX3 antibodies delay lupus-like nephritis and prolong survival of NZB/NZW F1 mice. In vitro observations suggest anti-PTX3 antibodies may dampen complement activation via their Fc fragment, likely hindering renal inflammation.
Collapse
Affiliation(s)
- Mariele Gatto
- Division of Rheumatology, Department of Medicine, University of Padova, Via Giustiniani 2, 35123, Padova, Italy
| | - Anna Ghirardello
- Division of Rheumatology, Department of Medicine, University of Padova, Via Giustiniani 2, 35123, Padova, Italy
| | - Roberto Luisetto
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Italy
| | - Nicola Bassi
- Division of Rheumatology, Department of Medicine, University of Padova, Via Giustiniani 2, 35123, Padova, Italy
| | - Marny Fedrigo
- Department of Cardiac, Thoracic and Vascular Sciences, University of Padova, Via Giustiniani 2, 35123, Padova, Italy
| | - Marialuisa Valente
- Department of Cardiac, Thoracic and Vascular Sciences, University of Padova, Via Giustiniani 2, 35123, Padova, Italy
| | - Sonia Valentino
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) -Humanitas Clinical and Research Center, 20089, Milan, Italy
| | - Dorella Del Prete
- Division of Nephrology, Department of Medicine DIMED, Laboratory of Histomorphology and Molecular Biology of the Kidney, University of Padua, Via Giustiniani 2, 35123, Padua, Italy
| | - Leonardo Punzi
- Division of Rheumatology, Department of Medicine, University of Padova, Via Giustiniani 2, 35123, Padova, Italy
| | - Andrea Doria
- Division of Rheumatology, Department of Medicine, University of Padova, Via Giustiniani 2, 35123, Padova, Italy.
| |
Collapse
|
26
|
Dema B, Charles N. Autoantibodies in SLE: Specificities, Isotypes and Receptors. Antibodies (Basel) 2016; 5:antib5010002. [PMID: 31557984 PMCID: PMC6698872 DOI: 10.3390/antib5010002] [Citation(s) in RCA: 118] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2015] [Revised: 12/09/2015] [Accepted: 12/11/2015] [Indexed: 12/23/2022] Open
Abstract
Systemic Lupus Erythematosus (SLE) is characterized by a wide spectrum of auto-antibodies which recognize several cellular components. The production of these self-reactive antibodies fluctuates during the course of the disease and the involvement of different antibody-secreting cell populations are considered highly relevant for the disease pathogenesis. These cells are developed and stimulated through different ways leading to the secretion of a variety of isotypes, affinities and idiotypes. Each of them has a particular mechanism of action binding to a specific antigen and recognized by distinct receptors. The effector responses triggered lead to a chronic tissue inflammation. DsDNA autoantibodies are the most studied as well as the first in being characterized for its pathogenic role in Lupus nephritis. However, others are of growing interest since they have been associated with other organ-specific damage, such as anti-NMDAR antibodies in neuropsychiatric clinical manifestations or anti-β2GP1 antibodies in vascular symptomatology. In this review, we describe the different auto-antibodies reported to be involved in SLE. How autoantibody isotypes and affinity-binding to their antigen might result in different pathogenic responses is also discussed.
Collapse
Affiliation(s)
- Barbara Dema
- Centre de Recherche sur l'Inflammation, INSERM UMR1149, CNRS ERL8252, Université Paris Diderot, Sorbonne Paris Cité, Faculté de Médecine site Bichat, Laboratoire d'Excellence Inflamex, DHU FIRE, Paris 75018, France.
| | - Nicolas Charles
- Centre de Recherche sur l'Inflammation, INSERM UMR1149, CNRS ERL8252, Université Paris Diderot, Sorbonne Paris Cité, Faculté de Médecine site Bichat, Laboratoire d'Excellence Inflamex, DHU FIRE, Paris 75018, France.
| |
Collapse
|
27
|
Xia Y, Eryilmaz E, Zhang Q, Cowburn D, Putterman C. Anti-DNA antibody mediated catalysis is isotype dependent. Mol Immunol 2015; 69:33-43. [PMID: 26655427 DOI: 10.1016/j.molimm.2015.11.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Revised: 10/29/2015] [Accepted: 11/01/2015] [Indexed: 12/27/2022]
Abstract
Anti-DNA antibodies are the serological hallmark of systemic lupus erythematosus, and participate in the pathogenesis of lupus nephritis by cross-reacting with multiple renal antigens. Previously, using a panel of murine anti-DNA IgGs that share identical variable regions but that differ in the constant regions, we demonstrated that the cross-reaction and renal pathogenicity of anti-DNA antibodies are isotype dependent. In this study, we investigated the catalytic potential of this anti-DNA antibody panel, and determined its isotype dependency. The three isotype switch variants (IgG1, IgG2a, IgG2b) and the parent IgG3 PL9-11 anti-DNA antibodies were compared in their catalysis of 500 base pair linear double stranded DNA and a 12-mer peptide (ALWPPNLHAWVP), by gel analysis, MALDI-TOF mass spectrometry, and nuclear magnetic resonance spectroscopy. The binding affinity of anti-DNA antibodies to double stranded DNA and peptide antigens were assessed by ELISA and surface plasmon resonance. We found that the PL9-11 antibody isotypes vary significantly in their potential to catalyze the cleavage of both linear and double stranded DNA and the proteolysis of peptides. The degree of the cleavage and proteolysis increases with the incubation temperature and time. While different PL9-11 isotypes have the same initial attack sites within the ALWPPNLHAWVP peptide, there was no correlation between binding affinity to the peptide and proteolysis rates. In conclusion, the catalytic properties of anti-DNA antibodies are isotype dependent. This finding provides further evidence that antibodies that share the same variable region, but which have different constant regions, are functionally distinct. The catalytic effects modulated by antibody constant regions need to be considered in the design of therapeutic antibodies (abzymes) and peptides designed to block pathogenic autoantibodies.
Collapse
Affiliation(s)
- Yumin Xia
- Department of Microbiology & Immunology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Division of Rheumatology, Albert Einstein College of Medicine, Bronx, NY 10461, USA.
| | - Ertan Eryilmaz
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY 10461, USA.
| | - Qiuting Zhang
- College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, Zhejiang 310058, China.
| | - David Cowburn
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY 10461, USA.
| | - Chaim Putterman
- Department of Microbiology & Immunology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Division of Rheumatology, Albert Einstein College of Medicine, Bronx, NY 10461, USA.
| |
Collapse
|
28
|
|
29
|
Wong EB, Soni C, Chan AY, Domeier PP, Shwetank, Abraham T, Limaye N, Khan TN, Elias MJ, Chodisetti SB, Wakeland EK, Rahman ZSM. B cell-intrinsic CD84 and Ly108 maintain germinal center B cell tolerance. THE JOURNAL OF IMMUNOLOGY 2015; 194:4130-43. [PMID: 25801429 DOI: 10.4049/jimmunol.1403023] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Accepted: 02/16/2015] [Indexed: 12/22/2022]
Abstract
Signaling lymphocyte activation molecules (SLAMs) play an integral role in immune regulation. Polymorphisms in the SLAM family receptors are implicated in human and mouse model of lupus disease. The lupus-associated, somatically mutated, and class-switched pathogenic autoantibodies are generated in spontaneously developed germinal centers (GCs) in secondary lymphoid organs. The role and mechanism of B cell-intrinsic expression of polymorphic SLAM receptors that affect B cell tolerance at the GC checkpoint are not clear. In this study, we generated several bacterial artificial chromosome-transgenic mice that overexpress C57BL/6 (B6) alleles of different SLAM family genes on an autoimmune-prone B6.Sle1b background. B6.Sle1b mice overexpressing B6-derived Ly108 and CD84 exhibit a significant reduction in the spontaneously developed GC response and autoantibody production compared with B6.Sle1b mice. These data suggest a prominent role for Sle1b-derived Ly108 and CD84 in altering the GC checkpoint. We further confirm that expression of lupus-associated CD84 and Ly108 specifically on GC B cells in B6.Sle1b mice is sufficient to break B cell tolerance, leading to an increase in autoantibody production. In addition, we observe that B6.Sle1b B cells have reduced BCR signaling and a lower frequency of B cell-T cell conjugates; the reverse is seen in B6.Sle1b mice overexpressing B6 alleles of CD84 and Ly108. Finally, we find a significant decrease in apoptotic GC B cells in B6.Sle1b mice compared with B6 controls. Our study establishes a central role for GC B cell-specific CD84 and Ly108 expression in maintaining B cell tolerance in GCs and in preventing autoimmunity.
Collapse
Affiliation(s)
- Eric B Wong
- Department of Microbiology and Immunology, Pennsylvania State University College of Medicine, Hershey, PA 17033
| | - Chetna Soni
- Department of Microbiology and Immunology, Pennsylvania State University College of Medicine, Hershey, PA 17033
| | - Alice Y Chan
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX 75390; and
| | - Phillip P Domeier
- Department of Microbiology and Immunology, Pennsylvania State University College of Medicine, Hershey, PA 17033
| | - Shwetank
- Department of Microbiology and Immunology, Pennsylvania State University College of Medicine, Hershey, PA 17033
| | - Thomas Abraham
- Department of Research Resources, Pennsylvania State University College of Medicine, Hershey, PA 17033
| | - Nisha Limaye
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX 75390; and
| | - Tahsin N Khan
- Department of Microbiology and Immunology, Pennsylvania State University College of Medicine, Hershey, PA 17033
| | - Melinda J Elias
- Department of Microbiology and Immunology, Pennsylvania State University College of Medicine, Hershey, PA 17033
| | - Sathi Babu Chodisetti
- Department of Microbiology and Immunology, Pennsylvania State University College of Medicine, Hershey, PA 17033
| | - Edward K Wakeland
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX 75390; and
| | - Ziaur S M Rahman
- Department of Microbiology and Immunology, Pennsylvania State University College of Medicine, Hershey, PA 17033;
| |
Collapse
|
30
|
Kitabatake M, Soma M, Zhang T, Kuwahara K, Fukushima Y, Nojima T, Kitamura D, Sakaguchi N. JNK regulatory molecule G5PR induces IgG autoantibody-producing plasmablasts from peritoneal B1a cells. THE JOURNAL OF IMMUNOLOGY 2015; 194:1480-8. [PMID: 25601926 DOI: 10.4049/jimmunol.1401127] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Peritoneal B1a cells expressing CD5 and CD11b generate autoantibody-producing precursors in autoimmune-prone mice. Previous studies show reduced JNK signaling in peritoneal B1a cells of female New Zealand Black mice and an abnormal increase of protein phosphatase 2A subunit G5PR that regulates BCR-mediated JNK signaling as a cause of autoimmunity. To investigate the mechanism regulating B1a differentiation into autoantibody-secreting plasmablasts (PBs), we applied an in vitro culture system that supports long-term growth of germinal center (GC) B cells (iGB) with IL-4, CD40L, and BAFF. Compared with spleen B2 cells, B1a cells differentiated into GC-like B cells, but more markedly into PBs, and underwent class switching toward IgG1. During iGB culture, B1a cells expressed GC-associated aicda, g5pr, and bcl6, and markedly PB-associated prdm1, irf4, and xbp1. B1a-derived iGB cells from New Zealand Black × New Zealand White F1 mice highly differentiated into autoantibody-secreting PBs in vitro and localized to the GC area in vivo. In iGB culture, JNK inhibitor SP600125 augmented the differentiation of C57BL/6 B1a cells into PBs. Furthermore, B1a cells from G5PR transgenic mice markedly differentiated into IgM and IgG autoantibody-secreting PBs. In conclusion, JNK regulation is critical to suppress autoantibody-secreting PBs from peritoneal B1a cells.
Collapse
Affiliation(s)
- Masahiro Kitabatake
- Department of Immunology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto 860-8556, Japan
| | - Miho Soma
- Department of Immunology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto 860-8556, Japan
| | - Tianli Zhang
- Department of Immunology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto 860-8556, Japan
| | - Kazuhiko Kuwahara
- Department of Immunology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto 860-8556, Japan
| | - Yoshimi Fukushima
- Department of Immunology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto 860-8556, Japan
| | - Takuya Nojima
- Division of Molecular Biology, Research Institute for Biomedical Sciences, Tokyo University of Science, Chiba 278-0022, Japan; and
| | - Daisuke Kitamura
- Division of Molecular Biology, Research Institute for Biomedical Sciences, Tokyo University of Science, Chiba 278-0022, Japan; and
| | - Nobuo Sakaguchi
- Department of Immunology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto 860-8556, Japan;
| |
Collapse
|
31
|
FcγRIIB regulates T-cell autoreactivity, ANCA production, and neutrophil activation to suppress anti-myeloperoxidase glomerulonephritis. Kidney Int 2014; 86:1140-9. [DOI: 10.1038/ki.2014.189] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2013] [Revised: 03/18/2014] [Accepted: 04/03/2014] [Indexed: 01/09/2023]
|
32
|
Hodge DL, Berthet C, Coppola V, Kastenmüller W, Buschman MD, Schaughency PM, Shirota H, Scarzello AJ, Subleski JJ, Anver MR, Ortaldo JR, Lin F, Reynolds DA, Sanford ME, Kaldis P, Tessarollo L, Klinman DM, Young HA. IFN-gamma AU-rich element removal promotes chronic IFN-gamma expression and autoimmunity in mice. J Autoimmun 2014; 53:33-45. [PMID: 24583068 PMCID: PMC4148478 DOI: 10.1016/j.jaut.2014.02.003] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2013] [Revised: 01/17/2014] [Accepted: 02/05/2014] [Indexed: 12/31/2022]
Abstract
We generated a mouse model with a 162 nt AU-rich element (ARE) region deletion in the 3' untranslated region (3'UTR) of the interferon-gamma (IFN-γ) gene that results in chronic circulating serum IFN-γ levels. Mice homozygous for the ARE deletion (ARE-Del) (-/-) present both serologic and cellular abnormalities typical of patients with systemic lupus erythematosus (SLE). ARE-Del(-/-) mice display increased numbers of pDCs in bone marrow and spleen. Addition of IFN-γ to Flt3-ligand (Flt3L) treated in vitro bone marrow cultures results in a 2-fold increase in pDCs with concurrent increases in IRF8 expression. Marginal zone B (MZB) cells and marginal zone macrophages (MZMs) are absent in ARE-Del(-/-) mice. ARE-Del(+/-) mice retain both MZB cells and MZMs and develop no or mild autoimmunity. However, low dose clodronate treatment in ARE-Del(+/-) mice specifically eliminates MZMs and promotes anti-DNA antibody development and glomerulonephritis. Our findings demonstrate the consequences of a chronic IFN-γ milieu on B220(+) cell types and in particular the impact of MZB cell loss on MZM function in autoimmunity. Furthermore, similarities between disease states in ARE-Del(-/-) mice and SLE patients suggest that IFN-γ may not only be a product of SLE but may be critical for disease onset and progression.
Collapse
Affiliation(s)
- Deborah L Hodge
- Laboratory of Experimental Immunology, Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute-Frederick, MD 21702, USA.
| | - Cyril Berthet
- Mouse Cancer Genetics Program, Center for Cancer Research, National Cancer Institute-Frederick, Frederick, MD 21702, USA; Oncodesign, 20 Rue Jean Mazen, Dijon 21076, France.
| | - Vincenzo Coppola
- Mouse Cancer Genetics Program, Center for Cancer Research, National Cancer Institute-Frederick, Frederick, MD 21702, USA; Department of Molecular Virology, Immunology and Medical Genetics at The Ohio State University, 460 W. 12th Street, Columbus, OH 43210, USA.
| | - Wolfgang Kastenmüller
- Lymphocyte Biology Section, Laboratory of Systems Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA; Cellular Interactions and Immunimaging Institutes of Molecular Medicine and Experimental Immunology (IMMEI), University of Bonn, Sigmund-Freud Str. 25, Bonn 53105, Germany.
| | - Matthew D Buschman
- Laboratory of Experimental Immunology, Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute-Frederick, MD 21702, USA; Division of Endocrinology and Metabolism, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA.
| | - Paul M Schaughency
- Laboratory of Experimental Immunology, Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute-Frederick, MD 21702, USA; Department of Molecular Biology and Genetics, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| | - Hidekazu Shirota
- Laboratory of Experimental Immunology, Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute-Frederick, MD 21702, USA; Department of Clinical Oncology, Tohoku University Hospital, 1-1 Seiryo-machi, Aoba-ku, Sendai 980-8574, Japan.
| | - Anthony J Scarzello
- Laboratory of Experimental Immunology, Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute-Frederick, MD 21702, USA.
| | - Jeff J Subleski
- Laboratory of Experimental Immunology, Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute-Frederick, MD 21702, USA.
| | - Miriam R Anver
- Laboratory Animal Science Program (LASP), Science Applications International Corporation (SAIC), National Cancer Institute, Frederick, MD 21702, USA.
| | - John R Ortaldo
- Laboratory of Experimental Immunology, Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute-Frederick, MD 21702, USA.
| | - Fanching Lin
- Laboratory of Experimental Immunology, Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute-Frederick, MD 21702, USA.
| | - Della A Reynolds
- Laboratory of Experimental Immunology, Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute-Frederick, MD 21702, USA.
| | - Michael E Sanford
- Laboratory of Experimental Immunology, Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute-Frederick, MD 21702, USA.
| | - Philipp Kaldis
- Mouse Cancer Genetics Program, Center for Cancer Research, National Cancer Institute-Frederick, Frederick, MD 21702, USA; Institute of Molecular and Cell Biology (IMCB), A*STAR (Agency for Science, Technology and Research), Proteos, 61 Biopolis Drive, Singapore 138673, Republic of Singapore.
| | - Lino Tessarollo
- Mouse Cancer Genetics Program, Center for Cancer Research, National Cancer Institute-Frederick, Frederick, MD 21702, USA.
| | - Dennis M Klinman
- Laboratory of Experimental Immunology, Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute-Frederick, MD 21702, USA.
| | - Howard A Young
- Laboratory of Experimental Immunology, Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute-Frederick, MD 21702, USA.
| |
Collapse
|
33
|
|
34
|
Yasuda K, Watkins AA, Kochar GS, Wilson GE, Laskow B, Richez C, Bonegio RG, Rifkin IR. Interferon regulatory factor-5 deficiency ameliorates disease severity in the MRL/lpr mouse model of lupus in the absence of a mutation in DOCK2. PLoS One 2014; 9:e103478. [PMID: 25076492 PMCID: PMC4116215 DOI: 10.1371/journal.pone.0103478] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2014] [Accepted: 07/01/2014] [Indexed: 11/18/2022] Open
Abstract
Interferon regulatory factor 5 (IRF5) polymorphisms are strongly associated with an increased risk of developing the autoimmune disease systemic lupus erythematosus. In mouse lupus models, IRF5-deficiency was shown to reduce disease severity consistent with an important role for IRF5 in disease pathogenesis. However these mouse studies were confounded by the recent demonstration that the IRF5 knockout mouse line contained a loss-of-function mutation in the dedicator of cytokinesis 2 (DOCK2) gene. As DOCK2 regulates lymphocyte trafficking and Toll-like receptor signaling, this raised the possibility that some of the protective effects attributed to IRF5 deficiency in the mouse lupus models may instead have been due to DOCK2 deficiency. We have therefore here evaluated the effect of IRF5-deficiency in the MRL/lpr mouse lupus model in the absence of the DOCK2 mutation. We find that IRF5-deficient (IRF5−/−) MRL/lpr mice develop much less severe disease than their IRF5-sufficient (IRF5+/+) littermates. Despite markedly lower serum levels of anti-nuclear autoantibodies and reduced total splenocyte and CD4+ T cell numbers, IRF5−/− MRL/lpr mice have similar numbers of all splenic B cell subsets compared to IRF5+/+ MRL/lpr mice, suggesting that IRF5 is not involved in B cell development up to the mature B cell stage. However, IRF5−/− MRL/lpr mice have greatly reduced numbers of spleen plasmablasts and bone marrow plasma cells. Serum levels of B lymphocyte stimulator (BLyS) were markedly elevated in the MRL/lpr mice but no effect of IRF5 on serum BLyS levels was seen. Overall our data demonstrate that IRF5 contributes to disease pathogenesis in the MRL/lpr lupus model and that this is due, at least in part, to the role of IRF5 in plasma cell formation. Our data also suggest that combined therapy targeting both IRF5 and BLyS might be a particularly effective therapeutic approach in lupus.
Collapse
Affiliation(s)
- Kei Yasuda
- Renal Section, Department of Medicine, Boston University School of Medicine, Boston, Massachusetts, United States of America
- * E-mail: (KY); (IRR)
| | - Amanda A. Watkins
- Renal Section, Department of Medicine, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Guneet S. Kochar
- Renal Section, Department of Medicine, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Gabriella E. Wilson
- Renal Section, Department of Medicine, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Bari Laskow
- Renal Section, Department of Medicine, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Christophe Richez
- Renal Section, Department of Medicine, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Ramon G. Bonegio
- Renal Section, Department of Medicine, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Ian R. Rifkin
- Renal Section, Department of Medicine, Boston University School of Medicine, Boston, Massachusetts, United States of America
- * E-mail: (KY); (IRR)
| |
Collapse
|
35
|
Goulabchand R, Vincent T, Batteux F, Eliaou JF, Guilpain P. Impact of autoantibody glycosylation in autoimmune diseases. Autoimmun Rev 2014; 13:742-50. [DOI: 10.1016/j.autrev.2014.02.005] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2014] [Accepted: 02/01/2014] [Indexed: 12/12/2022]
|
36
|
Metushi IG, Lee WM, Uetrecht J. IgG3 Is the Dominant Subtype of Anti-isoniazid Antibodies in Patients with Isoniazid-Induced Liver Failure. Chem Res Toxicol 2014; 27:738-40. [DOI: 10.1021/tx500108u] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Affiliation(s)
- Imir G. Metushi
- Department
of Pharmacology and Toxicology, Faculty of Medicine, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - William M. Lee
- Division
of Digestive and Liver Disease, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, Texas 75390, United States
| | - Jack Uetrecht
- Department
of Pharmacology and Toxicology, Faculty of Medicine, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| |
Collapse
|
37
|
Taher TE, Muhammad HA, Rahim A, Flores-Borja F, Renaudineau Y, Isenberg DA, Mageed RA. Aberrant B-lymphocyte responses in lupus: inherent or induced and potential therapeutic targets. Eur J Clin Invest 2013; 43:866-80. [PMID: 23701475 DOI: 10.1111/eci.12111] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2013] [Accepted: 04/29/2013] [Indexed: 12/11/2022]
Abstract
BACKGROUND Lupus is a prototype autoimmune disease of unknown aetiology. The disease is complex; manifest diverse clinical symptoms and disease mechanisms. This complexity has provided many leads to explore: from disease mechanisms to approaches for therapy. B-lymphocytes play a central role in the pathogenesis of the disease. However, the cause of aberrant B-lymphocyte responses in patients and, indeed, its causal relationship with the disease remain unclear. DESIGN This article provides a synopsis of current knowledge of immunological abnormalities in lupus with an emphasis on abnormalities in the B-lymphocyte compartment. RESULTS There is evidence for abnormalities in most compartments of the immune system in animal models and patients with lupus including an ever expanding list of abnormalities within the B-lymphocyte compartment. In addition, recent genome-wide linkage analyses in large cohorts of patients have identified new sets of genetic association factors some with potential links with defective B-lymphocyte responses although their full pathophysiological effects remain to be determined. The accumulating knowledge may help in the identification and application of new targeted therapies for treating lupus disease. CONCLUSIONS Cellular, molecular and genetic studies have provided significant insights into potential causes of immunological defects associated with lupus. Most of this insight relate to defects in B- and T-lymphocyte tolerance, signalling and responses. For B-lymphocytes, there is evidence for altered regulation of inter and intracellular signalling pathways at multiple levels. Some of these abnormalities will be discussed within the context of potential implications for disease pathogenesis and targeted therapies.
Collapse
Affiliation(s)
- Taher E Taher
- Bone & Joint Research Unit, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | | | | | | | | | | | | |
Collapse
|
38
|
de Salort J, Cuenca M, Terhorst C, Engel P, Romero X. Ly9 (CD229) Cell-Surface Receptor is Crucial for the Development of Spontaneous Autoantibody Production to Nuclear Antigens. Front Immunol 2013; 4:225. [PMID: 23914190 PMCID: PMC3728625 DOI: 10.3389/fimmu.2013.00225] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2013] [Accepted: 07/18/2013] [Indexed: 01/07/2023] Open
Abstract
The Signaling Lymphocyte Activation Molecule Family (SLAMF) genes, which encode cell-surface receptors that modulate innate and adaptive immune responses, lay within a genomic region of human and mouse chromosome 1 that confers a predisposition for the development of systemic lupus erythematosus (SLE). Herein, we demonstrate that the SLAMF member Ly9 arises as a novel receptor contributing to the reinforcement of tolerance. Specifically, Ly9-deficient mice spontaneously developed features of systemic autoimmunity such as the production of anti-nuclear antibodies (ANA), -dsDNA, and -nucleosome autoantibodies, independently of genetic background [(B6.129) or (BALB/c.129)]. In aged (10- to 12-month-old) Ly9−/− mice key cell subsets implicated in autoimmunity were expanded, e.g., T follicular helper (Tfh) as well as germinal center (GC) B cells. More importantly, in vitro functional experiments showed that Ly9 acts as an inhibitory receptor of IFN-γ producing CD4+ T cells. Taken together, our findings reveal that the Ly9 receptor triggers cell intrinsic safeguarding mechanisms to prevent a breach of tolerance, emerging as a new non-redundant inhibitory cell-surface receptor capable of disabling autoantibody responses.
Collapse
Affiliation(s)
- Jose de Salort
- Immunology Unit, Department of Cell Biology, Immunology and Neurosciences, Medical School, University of Barcelona , Barcelona , Spain
| | | | | | | | | |
Collapse
|
39
|
Flytzani S, Stridh P, Guerreiro-Cacais AO, Marta M, Hedreul MT, Jagodic M, Olsson T. Anti-MOG antibodies are under polygenic regulation with the most significant control coming from the C-type lectin-like gene locus. Genes Immun 2013; 14:409-19. [DOI: 10.1038/gene.2013.33] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2013] [Revised: 05/07/2013] [Accepted: 05/07/2013] [Indexed: 01/29/2023]
|
40
|
Giles BM, Boackle SA. Linking complement and anti-dsDNA antibodies in the pathogenesis of systemic lupus erythematosus. Immunol Res 2013; 55:10-21. [PMID: 22941560 DOI: 10.1007/s12026-012-8345-z] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Systemic lupus erythematosus is a severe autoimmune disease that affects multiple organ systems resulting in diverse symptoms and outcomes. It is characterized by antibody production to a variety of self-antigens, but it is specifically associated with those against anti-dsDNA. Anti-dsDNA antibodies are present before the onset of clinical disease and are associated with severe manifestations of lupus such as glomerulonephritis. Their levels fluctuate with changes in disease activity and, in combination with the levels of complement proteins C3 and C4, are strong indicators of disease flare and treatment response in patients with lupus. The decreased complement levels that are noted during flares of lupus activity are believed to be secondary to increased autoantibody production and immune complex formation that results in tissue damage; however, recent data suggest that complement activation can also drive development of these pathogenic autoantibodies. This review will explore the various roles of complement in the development and pathogenesis of anti-dsDNA antibodies.
Collapse
Affiliation(s)
- Brendan M Giles
- Division of Rheumatology, Department of Medicine, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | | |
Collapse
|
41
|
The constant region affects antigen binding of antibodies to DNA by altering secondary structure. Mol Immunol 2013; 56:28-37. [PMID: 23665381 DOI: 10.1016/j.molimm.2013.04.004] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2013] [Revised: 04/08/2013] [Accepted: 04/09/2013] [Indexed: 11/23/2022]
Abstract
We previously demonstrated an important role of the constant region in the pathogenicity of anti-DNA antibodies. To determine the mechanisms by which the constant region affects autoantibody binding, a panel of isotype-switch variants (IgG1, IgG2a, IgG2b) was generated from the murine PL9-11 IgG3 autoantibody. The affinity of the PL9-11 antibody panel for histone was measured by surface plasmon resonance (SPR). Tryptophan fluorescence was used to determine wavelength shifts of the antibody panel upon binding to DNA and histone. Finally, circular dichroism spectroscopy was used to measure changes in secondary structure. SPR analysis revealed significant differences in histone binding affinity between members of the PL9-11 panel. The wavelength shifts of tryptophan fluorescence emission were found to be dependent on the antibody isotype, while circular dichroism analysis determined that changes in antibody secondary structure content differed between isotypes upon antigen binding. Thus, the antigen binding affinity is dependent on the particular constant region expressed. Moreover, the effects of antibody binding to antigen were also constant region dependent. Alteration of secondary structures influenced by constant regions may explain differences in fine specificity of anti-DNA antibodies between antibodies with similar variable regions, as well as cross-reactivity of anti-DNA antibodies with non-DNA antigens.
Collapse
|
42
|
Lacotte S, Decossas M, Le Coz C, Brun S, Muller S, Dumortier H. Early differentiated CD138(high) MHCII+ IgG+ plasma cells express CXCR3 and localize into inflamed kidneys of lupus mice. PLoS One 2013; 8:e58140. [PMID: 23520491 PMCID: PMC3592892 DOI: 10.1371/journal.pone.0058140] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2012] [Accepted: 02/04/2013] [Indexed: 11/30/2022] Open
Abstract
Humoral responses are central to the development of chronic autoimmune diseases such as systemic lupus erythematosus. Indeed, autoantibody deposition is responsible for tissue damage, the kidneys being one of the main target organs. As the source of pathogenic antibodies, plasma cells are therefore critical players in this harmful scenario, both at systemic and local levels. The aim of the present study was to analyze plasma cells in NZB/W lupus mice and to get a better understanding of the mechanisms underlying their involvement in the renal inflammation process. Using various techniques (i.e. flow cytometry, quantitative PCR, ELISpot), we identified and extensively characterized three plasma cell intermediates, according to their B220/CD138/MHCII expression levels. Each of these cell subsets displays specific proliferation and antibody secretion capacities. Moreover, we evidenced that the inflammation-related CXCR3 chemokine receptor is uniquely expressed by CD138highMHCII+ plasma cells, which encompass both short- and long-lived cells and mostly produce IgG (auto)antibodies. Expression of CXCR3 allows efficient chemotactic responsiveness of these cells to cognate chemokines, which production is up-regulated in the kidneys of diseased NZB/W mice. Finally, using fluorescence and electron microscopy, we demonstrated the presence of CD138+CXCR3+IgG+ cells in inflammatory areas in the kidneys, where they are very likely involved in the injury process. Thus, early differentiated CD138highMHCII+ rather than terminally differentiated CD138highMHCIIlow plasma cells may be involved in the renal inflammatory injury in lupus, due to CXCR3 expression and IgG secretion.
Collapse
Affiliation(s)
- Stéphanie Lacotte
- CNRS, Institut de Biologie Moléculaire et Cellulaire, Immunopathologie et Chimie Thérapeutique/Laboratory of Excellence Medalis, Strasbourg, France
| | - Marion Decossas
- CNRS, Institut de Biologie Moléculaire et Cellulaire, Immunopathologie et Chimie Thérapeutique/Laboratory of Excellence Medalis, Strasbourg, France
| | - Carole Le Coz
- CNRS, Institut de Biologie Moléculaire et Cellulaire, Immunopathologie et Chimie Thérapeutique/Laboratory of Excellence Medalis, Strasbourg, France
| | - Susana Brun
- CNRS, Institut de Biologie Moléculaire et Cellulaire, Immunopathologie et Chimie Thérapeutique/Laboratory of Excellence Medalis, Strasbourg, France
| | - Sylviane Muller
- CNRS, Institut de Biologie Moléculaire et Cellulaire, Immunopathologie et Chimie Thérapeutique/Laboratory of Excellence Medalis, Strasbourg, France
| | - Hélène Dumortier
- CNRS, Institut de Biologie Moléculaire et Cellulaire, Immunopathologie et Chimie Thérapeutique/Laboratory of Excellence Medalis, Strasbourg, France
- * E-mail:
| |
Collapse
|
43
|
Loci associated with N-glycosylation of human immunoglobulin G show pleiotropy with autoimmune diseases and haematological cancers. PLoS Genet 2013; 9:e1003225. [PMID: 23382691 PMCID: PMC3561084 DOI: 10.1371/journal.pgen.1003225] [Citation(s) in RCA: 281] [Impact Index Per Article: 23.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2012] [Accepted: 11/21/2012] [Indexed: 12/22/2022] Open
Abstract
Glycosylation of immunoglobulin G (IgG) influences IgG effector function by modulating binding to Fc receptors. To identify genetic loci associated with IgG glycosylation, we quantitated N-linked IgG glycans using two approaches. After isolating IgG from human plasma, we performed 77 quantitative measurements of N-glycosylation using ultra-performance liquid chromatography (UPLC) in 2,247 individuals from four European discovery populations. In parallel, we measured IgG N-glycans using MALDI-TOF mass spectrometry (MS) in a replication cohort of 1,848 Europeans. Meta-analysis of genome-wide association study (GWAS) results identified 9 genome-wide significant loci (P<2.27×10−9) in the discovery analysis and two of the same loci (B4GALT1 and MGAT3) in the replication cohort. Four loci contained genes encoding glycosyltransferases (ST6GAL1, B4GALT1, FUT8, and MGAT3), while the remaining 5 contained genes that have not been previously implicated in protein glycosylation (IKZF1, IL6ST-ANKRD55, ABCF2-SMARCD3, SUV420H1, and SMARCB1-DERL3). However, most of them have been strongly associated with autoimmune and inflammatory conditions (e.g., systemic lupus erythematosus, rheumatoid arthritis, ulcerative colitis, Crohn's disease, diabetes type 1, multiple sclerosis, Graves' disease, celiac disease, nodular sclerosis) and/or haematological cancers (acute lymphoblastic leukaemia, Hodgkin lymphoma, and multiple myeloma). Follow-up functional experiments in haplodeficient Ikzf1 knock-out mice showed the same general pattern of changes in IgG glycosylation as identified in the meta-analysis. As IKZF1 was associated with multiple IgG N-glycan traits, we explored biomarker potential of affected N-glycans in 101 cases with SLE and 183 matched controls and demonstrated substantial discriminative power in a ROC-curve analysis (area under the curve = 0.842). Our study shows that it is possible to identify new loci that control glycosylation of a single plasma protein using GWAS. The results may also provide an explanation for the reported pleiotropy and antagonistic effects of loci involved in autoimmune diseases and haematological cancer. After analysing glycans attached to human immunoglobulin G in 4,095 individuals, we performed the first genome-wide association study (GWAS) of the glycome of an individual protein. Nine genetic loci were found to associate with glycans with genome-wide significance. Of these, four were enzymes that directly participate in IgG glycosylation, thus the observed associations were biologically founded. The remaining five genetic loci were not previously implicated in protein glycosylation, but the most of them have been reported to be relevant for autoimmune and inflammatory conditions and/or haematological cancers. A particularly interesting gene, IKZF1 was found to be associated with multiple IgG N-glycans. This gene has been implicated in numerous diseases, including systemic lupus erythematosus (SLE). We analysed N-glycans in 101 cases with SLE and 183 matched controls and demonstrated their substantial biomarker potential. Our study shows that it is possible to identify new loci that control glycosylation of a single plasma protein using GWAS. Our results may also provide an explanation for opposite effects of some genes in autoimmune diseases and haematological cancer.
Collapse
|
44
|
Lee SK, Silva DG, Martin JL, Pratama A, Hu X, Chang PP, Walters G, Vinuesa CG. Interferon-γ excess leads to pathogenic accumulation of follicular helper T cells and germinal centers. Immunity 2012; 37:880-92. [PMID: 23159227 DOI: 10.1016/j.immuni.2012.10.010] [Citation(s) in RCA: 207] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2011] [Accepted: 07/23/2012] [Indexed: 12/20/2022]
Abstract
Overactivity of the germinal center (GC) pathway resulting from accumulation of follicular helper T (Tfh) cells causes autoimmunity, underscoring the need to understand the factors that control Tfh cell homeostasis. Here we have identifed posttranscriptional repression of interferon-γ (Ifng) mRNA as a mechanism to limit Tfh cell formation. By using the sanroque lupus model, we have shown that decreased Ifng mRNA decay caused excessive IFN-γ signaling in T cells and led to accumulation of Tfh cells, spontaneous GC, autoantibody formation, and nephritis. Unlike ICOS and T-bet deficiency that failed to rescue several autoimmune manifestations, interferon-γ receptor (IFN-γR) deficiency prevented lupus development. IFN-γ blockade reduced Tfh cells and autoantibodies, demonstrating that IFN-γ overproduction was required to sustain lupus-associated pathology. Increased IFN-γR signaling caused Bcl-6 overexpression in Tfh cells and their precursors. This link between IFN-γ and aberrant Tfh cell formation provides a rationale for IFN-γ blockade in lupus patients with an overactive Tfh cell-associated pathway.
Collapse
Affiliation(s)
- Sau K Lee
- Department of Pathogens and Immunity, John Curtin School of Medical Research, The Australian National University, Acton, ACT 2601, Australia
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Sweet RA, Lee SK, Vinuesa CG. Developing connections amongst key cytokines and dysregulated germinal centers in autoimmunity. Curr Opin Immunol 2012; 24:658-64. [PMID: 23123277 DOI: 10.1016/j.coi.2012.10.003] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2012] [Accepted: 10/09/2012] [Indexed: 10/27/2022]
Abstract
Systemic autoimmunity owing to overactivity of Tfh and dysregulated germinal centers has been described in mice and humans. Cytokines such as IL-21, IFN-γ, IL-6 and IL-17 are elevated in the plasma of mouse models of lupus, arthritis, and multiple sclerosis, and in subsets of patients with autoimmune disease. Monoclonal antibodies targeting these cytokines are entering clinical trials. While these cytokines exert pleiotropic effects on immune cells and organs, it is becoming clear that each and all of them can profoundly regulate Tfh numbers and/or function and induce or maintain the aberrant germinal center reactions that lead to pathogenic autoantibody formation. Here we review recent discoveries into the roles of IL-21, IFN-γ, IL-6, and IL-17 in germinal center responses and antibody-driven autoimmunity. These new insights used in conjunction with biomarkers of an overactive Tfh pathway may help stratify patients to rationalize the use of emerging monoclonal anti-cytokine antibody therapies.
Collapse
Affiliation(s)
- Rebecca A Sweet
- Department of Pathogens and Immunity, John Curtin School of Medical Research, The Australian National University, Canberra, ACT, Australia
| | | | | |
Collapse
|
46
|
Chang SH, Johns M, Boyle JJ, McConnell E, Kirkham PA, Bicknell C, Zahoor-ul-Hassan Dogar M, Edwards RJ, Gale-Grant O, Khamis R, Ramkhelawon KVV, Haskard DO. Model IgG monoclonal autoantibody-anti-idiotype pair for dissecting the humoral immune response to oxidized low density lipoprotein. Hybridoma (Larchmt) 2012; 31:87-98. [PMID: 22509912 DOI: 10.1089/hyb.2011.0058] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Increasing evidence implicates IgG autoantibodies against oxidized forms of low density lipoprotein (oxLDL) in the pathophysiology of atherosclerotic arterial disease. However, insufficient knowledge of their structure and function is a key gap. Using an elderly LDL receptor-deficient atherosclerotic mouse, we isolated a novel IgG3k against oxLDL (designated MAb LO1). LO1 reacts with copper-oxidized LDL, but minimally with native LDL. Further analysis showed that MAb LO1 also reacts in vitro with malondialdehyde-conjugated LDL (MDA-LDL), a known key epitope in copper-oxidized LDL preparations. By screening a phage library expressing single chain variable region antibodies (scFv), we selected an anti-idiotype scFv (designated H3) that neutralizes MAb LO1 binding to MDA-LDL. Amino acid substitutions between H3 and an irrelevant control scFv C12 showed that residues in the H3 CDRH2, CDRH3, and CDRL2 are all critical for MAb LO1 binding, consistent with a conformational epitope on H3 involving both heavy and light chains. Comparison of amino acids in H3 CDRH2 and CDRL2 with apoB, the major LDL protein, showed homologous sequences, suggesting H3 has structural similarities to the MAb LO1 binding site on MDA-LDL. Immunocytochemical staining showed that MAb LO1 binds epitopes in mouse and human atherosclerotic lesions. The MAb LO1-H3 combination therefore provides a very promising model for analyzing the structure and function of an individual IgG autoantibody in relation to atherosclerosis.
Collapse
Affiliation(s)
- Shang-Hung Chang
- Vascular Sciences Section, Department of Medicine, Imperial College, Hammersmith Hospital, London, United Kingdom
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Xia Y, Campbell SR, Broder A, Herlitz L, Abadi M, Wu P, Michaelson JS, Burkly LC, Putterman C. Inhibition of the TWEAK/Fn14 pathway attenuates renal disease in nephrotoxic serum nephritis. Clin Immunol 2012; 145:108-21. [PMID: 22982296 DOI: 10.1016/j.clim.2012.08.008] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2012] [Revised: 08/07/2012] [Accepted: 08/08/2012] [Indexed: 02/08/2023]
Abstract
Previously it was shown that the TNF superfamily member TWEAK (TNFSF12) acts through its receptor, Fn14, to promote proinflammatory responses in kidney cells, including the production of MCP-1, RANTES, IP-10 and KC. In addition, the TWEAK/Fn14 pathway promotes mesangial cell proliferation, vascular cell activation, and renal cell death. To study the relevance of the TWEAK/Fn14 pathway in the pathogenesis of antibody-induced nephritis using the mouse model of nephrotoxic serum nephritis (NTN), we induced NTN by passive transfer of rabbit anti-glomerular antibodies into Fn14 knockout (KO) and wild type (WT) mice. Severe proteinuria as well as renal histopathology were induced in WT but not in Fn14 KO mice. Similarly, a pharmacologic approach of anti-TWEAK mAb administration into WT mice in the NTN model significantly ameliorated proteinuria and improved kidney histology. Anti-TWEAK treatment did not affect the generation of mouse anti-rabbit antibodies; however, within the kidney there was a significant decrease in glomerular immunoglobulin deposition, as well as macrophage infiltrates and tubulointerstitial fibrosis. The mechanism of action is most likely due to reductions in downstream targets of TWEAK/Fn14 signaling, including reduced renal expression of MCP-1, VCAM-1, IP-10, RANTES as well as Fn14 itself, and other molecular pathways associated with fibrosis in anti-TWEAK treated mice. Thus, TWEAK/Fn14 interactions are instrumental in the pathogenesis of nephritis in the NTN model, apparently mediating a cascade of pathologic events locally in the kidney rather than by impacting the systemic immune response. Disrupting TWEAK/Fn14 interactions may be an innovative kidney-protective approach for the treatment of lupus nephritis and other antibody-induced renal diseases.
Collapse
Affiliation(s)
- Yumin Xia
- Division of Rheumatology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Xia Y, Pawar RD, Nakouzi AS, Herlitz L, Broder A, Liu K, Goilav B, Fan M, Wang L, Li QZ, Casadevall A, Putterman C. The constant region contributes to the antigenic specificity and renal pathogenicity of murine anti-DNA antibodies. J Autoimmun 2012; 39:398-411. [PMID: 22841793 DOI: 10.1016/j.jaut.2012.06.005] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2012] [Revised: 06/26/2012] [Accepted: 06/28/2012] [Indexed: 12/26/2022]
Abstract
Affinity for DNA and cross-reactivity with renal antigens are associated with enhanced renal pathogenicity of lupus autoantibodies. In addition, certain IgG subclasses are enriched in nephritic kidneys, suggesting that isotype may determine the outcome of antibody binding to renal antigens. To investigate if the isotype of DNA antibodies affects renal pathogenicity by influencing antigen binding, we derived IgM, IgG1, IgG2b and IgG2a forms of the PL9-11 antibody (IgG3 anti-DNA) by in vitro class switching or PCR cloning. The affinity and specificity of PL9-11 antibodies for nuclear and renal antigens were analyzed using ELISA, Western blotting, surface plasmon resonance (SPR), binding to mesangial cells, and glomerular proteome arrays. Renal deposition and pathogenicity were assayed in mice injected with PL9-11 hybridomas. We found that PL9-11 and its isotype-switched variants had differential binding to DNA and chromatin (IgG3>IgG2a>IgG1>IgG2b>IgM) by direct and competition ELISA, and SPR. In contrast, in binding to laminin and collagen IV the IgG2a isotype actually had the highest affinity. Differences in affinity of PL9-11 antibodies for renal antigens were mirrored in analysis of specificity for glomeruli, and were associated with significant differences in renal pathogenicity in vivo and survival. Our novel findings indicate that the constant region plays an important role in the nephritogenicity of antibodies to DNA by affecting immunoglobulin affinity and specificity. Increased binding to multiple glomerular and/or nuclear antigens may contribute to the renal pathogenicity of anti-DNA antibodies of the IgG2a and IgG3 isotype. Finally, class switch recombination may be another mechanism by which B cell autoreactivity is generated.
Collapse
Affiliation(s)
- Yumin Xia
- The Department of Microbiology & Immunology, Albert Einstein College of Medicine (AECOM), Bronx, NY 10461, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Definition of IFN-γ-related pathways critical for chemically-induced systemic autoimmunity. J Autoimmun 2012; 39:323-31. [PMID: 22578563 DOI: 10.1016/j.jaut.2012.04.003] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2012] [Revised: 04/12/2012] [Accepted: 04/15/2012] [Indexed: 12/24/2022]
Abstract
IFN-γ is essential for idiopathic and murine mercury-induced systemic autoimmunity (mHgIA), and heterozygous IFN-γ(+/-) mice also exhibit reduced disease. This suggests that blocking specific IFN-γ-related pathways that may only partially inhibit IFN-γ production or function will also suppress autoimmunity. To test this hypothesis, mice deficient in genes regulating IFN-γ expression (Casp1, Nlrp3, Il12a, Il12b, Stat4) or function (Ifngr1, Irf1) were examined for mHgIA susceptibility. Absence of either Ifngr1 or Irf1 resulted in a striking reduction of disease, while deficiency of genes promoting IFN-γ expression had modest to no effect. Furthermore, both Irf1- and Ifng-deficiency only modestly reduced the expansion of CD44(hi) and CD44(hi)CD55(lo) CD4(+) T cells, indicating that they are not absolutely required for T cell activation. Thus, there is substantial redundancy in genes that regulate IFN-γ expression in contrast to those that mediate later signaling events. These findings have implications for the therapeutic targeting of IFN-γ pathways in systemic autoimmunity.
Collapse
|
50
|
Kang HK, Chiang MY, Ecklund D, Zhang L, Ramsey-Goldman R, Datta SK. Megakaryocyte progenitors are the main APCs inducing Th17 response to lupus autoantigens and foreign antigens. THE JOURNAL OF IMMUNOLOGY 2012; 188:5970-80. [PMID: 22561152 DOI: 10.4049/jimmunol.1200452] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
In search of autoantigen-presenting cells that prime the pathogenic autoantibody-inducing Th cells of lupus, we found that CD41(+)CD151(+) cells among Lineage(-) (Lin(-)) CD117(+) (c-Kit(+)) CX3CR1(-) splenocytes depleted of known APCs were most proficient in presenting nuclear autoantigens from apoptotic cells to induce selectively an autoimmune Th17 response in different lupus-prone mouse strains. The new APCs have properties resembling megakaryocyte and/or bipotent megakaryocyte/erythroid progenitors of bone marrow, hence they are referred to as MM cells in this study. The MM cells produce requisite cytokines, but they require contact for optimal Th17 induction upon nucleosome feeding, and can induce Th17 only before undergoing differentiation to become c-Kit(-)CD41(+) cells. The MM cells expand up to 10-fold in peripheral blood of lupus patients and 49-fold in spleens of lupus mice preceding disease activity; they accelerate lupus in vivo and break tolerance in normal mice, inducing autoimmune Th17 cells. MM cells also cause Th17 skewing to foreign Ag in normal mice without Th17-polarizing culture conditions. Several molecules in MM cells are targets for blocking of autoimmunization. This study advances our understanding of lupus pathogenesis and Th17 differentiation biology by characterizing a novel category of APC.
Collapse
Affiliation(s)
- Hee-Kap Kang
- Division of Rheumatology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | | | | | | | | | | |
Collapse
|