1
|
La Gualana F, Olivieri G, Petriti B, Picciariello L, Natalucci F, Sciannamea M, Gragnani L, Basile U, Casato M, Spinelli FR, Stefanini L, Basili S, Visentini M, Ceccarelli F, Conti F. Early decrease of T-bet + B cells during subcutaneous belimumab predicts response to therapy in systemic lupus erythematosus patients. Immunol Lett 2025; 272:106962. [PMID: 39643119 DOI: 10.1016/j.imlet.2024.106962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Revised: 11/26/2024] [Accepted: 12/03/2024] [Indexed: 12/09/2024]
Abstract
Systemic lupus erythematosus (SLE) is characterized by B cell dysregulation and expansion of atypical B cells that may correlate with disease manifestations and activity. This study investigated the impact of subcutaneous (sc) Belimumab (BLM) on the peripheral B cell compartment and on the functional properties of CD21low, T-bet+ and CD11c+ atypical B cells, in 21 active SLE patients over a 12-month period. At baseline, active SLE patients displayed reduced unswitched IgM memory B cells and expansion of atypical B cells, compared to healthy donors and to SLE patients in remission. sc BLM therapy promptly restored B cell homeostasis with a reduction of T-bet+ B cells, observed early in patients responsive to therapy. These findings highlight the pathogenic role of T-bet+ B cells in SLE disease and suggest their potential utility as biomarker of clinical response.
Collapse
Affiliation(s)
- Francesca La Gualana
- Department of Translational and Precision Medicine, Sapienza University, Rome, Italy
| | - Giulio Olivieri
- Research Unit of Clinical Immunology and Vaccinology, Bambino Gesù Children's Hospital, IRCCS, Rome
| | - Begi Petriti
- Department of Translational and Precision Medicine, Sapienza University, Rome, Italy
| | - Licia Picciariello
- Lupus Clinic, Rheumatology, Dipartimento di Scienze Cliniche Internistiche Anestesiologiche e Cardiovascolari, Sapienza Università di Roma, Viale del Policlinico 155 00161 Rome, Italy
| | - Francesco Natalucci
- Lupus Clinic, Rheumatology, Dipartimento di Scienze Cliniche Internistiche Anestesiologiche e Cardiovascolari, Sapienza Università di Roma, Viale del Policlinico 155 00161 Rome, Italy
| | - Maddalena Sciannamea
- Department of Translational and Precision Medicine, Sapienza University, Rome, Italy
| | - Laura Gragnani
- Department of Translational Research and New Technologies in Medicine and Surgery, Medical School, University of Pisa, Pisa, Italy
| | - Umberto Basile
- Department of Clinical Pathology, Santa Maria Goretti HospitalAUSL Latina, Latina, Italy
| | - Milvia Casato
- Department of Translational and Precision Medicine, Sapienza University, Rome, Italy
| | - Francesca Romana Spinelli
- Lupus Clinic, Rheumatology, Dipartimento di Scienze Cliniche Internistiche Anestesiologiche e Cardiovascolari, Sapienza Università di Roma, Viale del Policlinico 155 00161 Rome, Italy
| | - Lucia Stefanini
- Department of Translational and Precision Medicine, Sapienza University, Rome, Italy
| | - Stefania Basili
- Department of Translational and Precision Medicine, Sapienza University, Rome, Italy
| | - Marcella Visentini
- Department of Translational and Precision Medicine, Sapienza University, Rome, Italy.
| | - Fulvia Ceccarelli
- Lupus Clinic, Rheumatology, Dipartimento di Scienze Cliniche Internistiche Anestesiologiche e Cardiovascolari, Sapienza Università di Roma, Viale del Policlinico 155 00161 Rome, Italy
| | - Fabrizio Conti
- Lupus Clinic, Rheumatology, Dipartimento di Scienze Cliniche Internistiche Anestesiologiche e Cardiovascolari, Sapienza Università di Roma, Viale del Policlinico 155 00161 Rome, Italy
| |
Collapse
|
2
|
Bang SY, Joh CSY, Itamiya T, Jeong S, Lee JH, Kwon H, Jin H, Jung J, Chung H, Lee BH, Gong JR, Ishigaki K, Fujio K, Bae SC, Kim HJ, Lee HS. Unveiling the dynamics of B lymphocytes in systemic lupus erythematosus patients treated with belimumab through longitudinal single-cell RNA sequencing. Rheumatology (Oxford) 2025; 64:2220-2226. [PMID: 39037931 DOI: 10.1093/rheumatology/keae364] [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: 03/08/2024] [Revised: 05/17/2024] [Accepted: 06/18/2024] [Indexed: 07/24/2024] Open
Abstract
OBJECTIVES Unravelling the mechanisms underlying treatment response for targeted therapeutics in systemic lupus erythematosus (SLE) patients is challenging due to the limited understanding of diverse responses of circulating immune cells, particularly B cells. We investigated B lymphocyte dynamics during anti-BAFF treatment, utilizing longitudinal single-cell transcriptome data. METHODS We conducted single-cell RNA sequencing on peripheral blood mononuclear cells (PBMCs) in four Korean SLE patients before and after belimumab treatment at the following time points: 2 weeks, 1, 3, 6 and 12 months. RESULTS Analysing over 73 000 PBMCs, we identified eight distinct subsets of B cells and plasmablasts and analysed dynamic changes within these cell subsets: initial declines in naïve and transitional B cells followed by an increase at 3 months, contrasted by an initial increase and subsequent decrease in memory B cells by the third month. Meanwhile, plasmablasts exhibited a consistent decline throughout the treatment. B cell activation pathways, specifically in naïve and memory B cells, were downregulated during the third and sixth months. These findings were validated at the protein level throughout the first 4 weeks of treatment using flow cytometry. Comparative analysis with bulk transcriptome data from 22 Japanese SLE patients showed increased NR4A1 expression 6 months post-belimumab treatment, indicating its role in restricting self-reactive B cells, thereby contributing to the biological responses of anti-BAFF treatment. CONCLUSION The observed B cell dynamics provided insights into the immunological mechanisms underlying the therapeutic effects of anti-BAFF in SLE patients. Furthermore, it underscores the need for research in predicting drug responses based on immune profiling.
Collapse
Affiliation(s)
- So-Young Bang
- Department of Rheumatology, Hanyang University Hospital for Rheumatic Diseases and Hanyang University Institute for Rheumatology Research, Seoul, South Korea
| | - Christine Suh-Yun Joh
- Department of Biomedical Sciences, Seoul National University Graduate School, Seoul, South Korea
| | - Takahiro Itamiya
- Department of Allergy and Rheumatology, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
- Department of Functional Genomics and Immunological Diseases, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| | - Soyoung Jeong
- Department of Biomedical Sciences, Seoul National University Graduate School, Seoul, South Korea
| | - Jung-Ho Lee
- Department of Biomedical Sciences, Seoul National University Graduate School, Seoul, South Korea
| | - Haeyoon Kwon
- Department of Medicine, Seoul National University College of Medicine, Seoul, South Korea
| | - Hyunjin Jin
- Transplantation Research Institute, Medical Research Center, Seoul National University College of Medicine, Seoul, South Korea
| | - Jaewon Jung
- Department of Biomedical Sciences, Seoul National University Graduate School, Seoul, South Korea
| | - Hyeyeon Chung
- Department of Biomedical Sciences, Seoul National University Graduate School, Seoul, South Korea
| | - Brian H Lee
- Department of Biomedical Sciences, Seoul National University Graduate School, Seoul, South Korea
| | - Jeong-Ryeol Gong
- Transplantation Research Institute, Medical Research Center, Seoul National University College of Medicine, Seoul, South Korea
| | - Kazuyoshi Ishigaki
- Laboratory for Human Immunogenetics, RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa, Japan
| | - Keishi Fujio
- Department of Allergy and Rheumatology, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| | - Sang-Cheol Bae
- Department of Rheumatology, Hanyang University Hospital for Rheumatic Diseases and Hanyang University Institute for Rheumatology Research, Seoul, South Korea
| | - Hyun Je Kim
- Department of Biomedical Sciences, Seoul National University Graduate School, Seoul, South Korea
- Genomic Medicine Institute, Seoul National University College of Medicine, Seoul, South Korea
- Department of Dermatology, Seoul National University Hospital, South Korea
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, South Korea
| | - Hye-Soon Lee
- Department of Rheumatology, Hanyang University Hospital for Rheumatic Diseases and Hanyang University Institute for Rheumatology Research, Seoul, South Korea
| |
Collapse
|
3
|
Xie M, Tsai CY, Woo J, Nuritdinov F, Cristaldo M, Odjourian NM, Antilus-Sainte R, Dougher M, Gengenbacher M. BAFF and APRIL immunotherapy following Bacille Calmette-Guérin vaccination enhances protection against pulmonary tuberculosis in mice. Front Immunol 2025; 16:1551183. [PMID: 39981256 PMCID: PMC11839638 DOI: 10.3389/fimmu.2025.1551183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Accepted: 01/21/2025] [Indexed: 02/22/2025] Open
Abstract
Introduction Bacille Calmette-Guérin (BCG), the only tuberculosis vaccine currently in clinical use, provides inadequate long-term protection. Administered at birth, BCG induces broad immune responses against a large number of antigens shared with Mycobacterium tuberculosis (Mtb), but protection wanes over time. We have previously shown that unconventional B cell subsets play a role in tuberculosis control. Methods High-dimensional flow cytometry and multiplex cytokine analysis were employed to investigate the effects of immunotherapy on BCG-vaccinated mice in an Mtb challenge model. Results In this study, we investigate the potential of recombinant cytokines targeting B cells - B-cell activating factor (BAFF) and A proliferation-inducing ligand (APRIL) - to modulate BCG immunity and improve protection in mice. Both cytokines play overlapping roles in B cell development and peripheral survival. Following subcutaneous BCG vaccination, immunotherapy with BAFF or APRIL resulted in an increased frequency of unconventional B cells potentially transitioning into antibody-producing plasma cells. Concurrently, we observed an increased frequency of central memory T cells, a subset critical for protective immunity. Changes in cellular immune responses were accompanied by reduced pro-inflammatory cytokine profiles and a contraction of the leukocyte population in lungs. Importantly, mice receiving BCG vaccination followed by BAFF or APRIL immunotherapy exhibited superior long-term protection against pulmonary tuberculosis relative to controls that received only BCG. Conclusion In summary, our findings demonstrate that combining BCG vaccination with B cell targeted immunomodulatory therapies can improve long-term protection against pulmonary tuberculosis, highlighting the continued relevance and underutilized potential of BCG as a vaccine platform.
Collapse
Affiliation(s)
- Min Xie
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, NJ, United States
| | - Chen-Yu Tsai
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, NJ, United States
| | - Joshua Woo
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, NJ, United States
| | - Frank Nuritdinov
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, NJ, United States
| | - Melissa Cristaldo
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, NJ, United States
| | - Narineh M. Odjourian
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, NJ, United States
| | | | - Maureen Dougher
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, NJ, United States
| | - Martin Gengenbacher
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, NJ, United States
- Department of Medical Sciences, Hackensack Meridian School of Medicine, Nutley, NJ, United States
| |
Collapse
|
4
|
Al-Aubodah TA, Piccirillo CA, Trachtman H, Takano T. The autoimmune architecture of childhood idiopathic nephrotic syndrome. Kidney Int 2025; 107:271-279. [PMID: 39571906 DOI: 10.1016/j.kint.2024.10.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 09/26/2024] [Accepted: 10/17/2024] [Indexed: 12/10/2024]
Abstract
Idiopathic nephrotic syndrome, the most common glomerular disorder in children, has long been considered an immune-mediated disease based on the efficacy of glucocorticoids at inducing remission. Nevertheless, the immune processes leading to podocytopathy have largely remained elusive. The success of B-cell depletion with rituximab, descriptions of B-cell dysregulation during active disease, and the most recent discovery of autoantibodies targeting the major podocyte antigen nephrin point to an autoimmune humoral etiology for idiopathic nephrotic syndrome. Investigations of the immune factors involved in idiopathic nephrotic syndrome pathogenesis have uncovered common features with other autoimmune disorders that will aid in prognostication and in guiding the expansion of our glucocorticoid-sparing therapeutic arsenal. In this review, we discuss the emerging autoimmune architecture of idiopathic nephrotic syndrome, with a specific focus on pediatric steroid-sensitive disease, including the podocyte-reactive B-cell response that causes anti-podocyte antibodies, the predisposing genetic factors that shape the podocyte-reactive immune landscape, and the immune triggers driving active disease.
Collapse
Affiliation(s)
- Tho-Alfakar Al-Aubodah
- Department of Microbiology and Immunology, Faculty of Medicine and Health Sciences, McGill University, Montréal, Québec, Canada; Infectious Diseases and Immunity in Global Health Program, Centre for Translational Biology, Research Institute of the McGill University Health Centre, Montréal, Québec, Canada; Metabolic Disorders and Complications Program, Centre for Translational Biology, Research Institute of the McGill University Health Centre, Montréal, Québec, Canada
| | - Ciriaco A Piccirillo
- Department of Microbiology and Immunology, Faculty of Medicine and Health Sciences, McGill University, Montréal, Québec, Canada; Infectious Diseases and Immunity in Global Health Program, Centre for Translational Biology, Research Institute of the McGill University Health Centre, Montréal, Québec, Canada
| | - Howard Trachtman
- Division of Nephrology, Department of Pediatrics, University of Michigan, Ann Arbor, Michigan, USA
| | - Tomoko Takano
- Metabolic Disorders and Complications Program, Centre for Translational Biology, Research Institute of the McGill University Health Centre, Montréal, Québec, Canada; Division of Nephrology, Faculty of Medicine and Health Sciences, McGill University, Montréal, Québec, Canada.
| |
Collapse
|
5
|
Scaletti C, Pratesi S, Bellando Randone S, Di Pietro L, Campochiaro C, Annunziato F, Matucci Cerinic M. The B-cells paradigm in systemic sclerosis: an update on pathophysiology and B-cell-targeted therapies. Clin Exp Immunol 2025; 219:uxae098. [PMID: 39498828 PMCID: PMC11754866 DOI: 10.1093/cei/uxae098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 10/05/2024] [Accepted: 11/04/2024] [Indexed: 11/07/2024] Open
Abstract
Systemic sclerosis (SSc) is considered a rare autoimmune disease in which there are alterations of both the innate and adaptive immune response resulting in the production of autoantibodies. Abnormalities of the immune system compromise the normal function of blood vessels leading to a vasculopathy manifested by Raynaud's phenomenon, an early sign of SSc . As a consequence of this reactive picture, the disease can evolve leading to tissue fibrosis. Several SSc-specific autoantibodies are currently known and are associated with specific clinical manifestations and prognosis. Although the pathogenetic role of these autoantibodies is still unclear, their production by B cells and plasma cells suggests the importance of these cells in the development of SSc. This review narratively examines B-cell dysfunctions and their role in the pathogenesis of SSc and discusses B-cell-targeted therapies currently used or potentially useful for the management of end-organ complications.
Collapse
Affiliation(s)
- Cristina Scaletti
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Sara Pratesi
- Flow Cytometry Diagnostic Center and Immunotherapy, University Hospital Careggi, Florence, Italy
| | - Silvia Bellando Randone
- Department of Clinical and Experimental Medicine, Rheumatology Unit, University of Florence, and Scleroderma Unit, University Hospital Careggi, Florence, Italy
| | - Linda Di Pietro
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Corrado Campochiaro
- Unit of Immunology, Rheumatology, Allergy and Rare diseases (UnIRAR), IRCCS San Raffaele Hospital, Milan, Italy
| | - Francesco Annunziato
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
- Flow Cytometry Diagnostic Center and Immunotherapy, University Hospital Careggi, Florence, Italy
| | - Marco Matucci Cerinic
- Unit of Immunology, Rheumatology, Allergy and Rare diseases (UnIRAR), IRCCS San Raffaele Hospital, Milan, Italy
- Vita Salute San Raffaele University, Milan, Italy
| |
Collapse
|
6
|
Hu SZ, Yuan ZY, Zhang XX, Yu XJ, Ni HY, Sun SJ, Xu T, Zhan HQ. The emerging role of BLyS/APRIL in autoimmune diseases: Biological characteristics, functions, and therapeutic potential. J Autoimmun 2024; 149:103329. [PMID: 39504927 DOI: 10.1016/j.jaut.2024.103329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 10/26/2024] [Accepted: 10/26/2024] [Indexed: 11/08/2024]
Abstract
Autoimmune diseases (AIDs) are common diseases in the world. Some cases are difficult to cure and can only delay the progression of the diseases. The B lymphocyte stimulator (BLyS)/a proliferation-inducing ligand (APRIL) plays an important role in B cell homeostasis, regulation of both innate and adaptive immune responses. After binding to their receptors, BLyS/APRIL primarily affects the survival and development of marginal, transitional, and mature B cells. Of note, elevated BLyS/APRIL is seen in many AIDs, such as systemic lupus erythematosus, rheumatoid arthritis, immunoglobulin A nephropathy, etc. Moreover, there is evidence that blocking these two cytokines can control the number of serum autoantibodies, promote the depletion of B lymphocytes, inhibit the activation of T cells and dendritic lymphocytes, and reduce inflammatory stress. Currently, some clinical studies are underway targeting BLyS/APRIL inhibitors for the treatment of AIDs. However, due to the scattered knowledge on the relationship between BLyS/APRIL and AIDs, it is necessary to sort out the existing data. Therefore, in this review, we describe the basic biological characteristics and functions of BLyS/APRIL in AIDs, summarize the potential clinical applications of related inhibitors, especially monoclonal antibodies and recombinant fusion proteins targeting BLyS/APRIL in AIDs, and also outline promising research directions.
Collapse
Affiliation(s)
- Shi-Zhi Hu
- Department of Pathology, The Second Affiliated Hospital of Anhui Medical University, 678 Furong Road, Hefei, Anhui, 230601, China; Department of Pathology, School of Basic Medical Sciences, Anhui Medical University, 81 Meishan Road, Hefei, Anhui, 230032, China
| | - Zhan-Yuan Yuan
- Department of Plastic and Reconstructive Surgery, The Second Affiliated Hospital of Anhui Medical University, 678 Furong Road, Hefei, Anhui, 230601, China
| | - Xiao-Xun Zhang
- Department of Pathology, School of Basic Medical Sciences, Anhui Medical University, 81 Meishan Road, Hefei, Anhui, 230032, China
| | - Xiao-Jing Yu
- Department of Pathology, The Second Affiliated Hospital of Anhui Medical University, 678 Furong Road, Hefei, Anhui, 230601, China
| | - Hai-Yan Ni
- Department of Pathology, School of Basic Medical Sciences, Anhui Medical University, 81 Meishan Road, Hefei, Anhui, 230032, China
| | - Sheng-Jia Sun
- Clinical Medical College of Anhui Medical University, 1166 Wangjiang West Road, Hefei, Anhui, 230031, China
| | - Tao Xu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, 81 Meishan Road, Hefei, Anhui, 230032, China.
| | - He-Qin Zhan
- Department of Pathology, The Second Affiliated Hospital of Anhui Medical University, 678 Furong Road, Hefei, Anhui, 230601, China; Department of Pathology, School of Basic Medical Sciences, Anhui Medical University, 81 Meishan Road, Hefei, Anhui, 230032, China.
| |
Collapse
|
7
|
Felten R, Foray AP, Schneider P, Marquet C, Pecquet C, Monneaux F, Dumortier H, Sibilia J, Valette F, Chatenoud L, Gottenberg JE. Efficacy of BAFF inhibition and B-cell depletion in non-obese diabetic mice as a spontaneous model for Sjögren's disease. RMD Open 2024; 10:e004112. [PMID: 39209370 PMCID: PMC11367362 DOI: 10.1136/rmdopen-2024-004112] [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: 01/12/2024] [Accepted: 07/19/2024] [Indexed: 09/04/2024] Open
Abstract
INTRODUCTION The therapeutic interest of targeting B-cell activating factor (BAFF) in Sjögren's disease (SjD) can be suspected from the results of two phase II clinical trials but has not been evaluated in an animal model of the disease. We aimed to evaluate the therapeutic efficacy of this strategy on dryness and salivary gland (SG) infiltrates in the NOD mouse model of SjD. MATERIAL AND METHODS Female NOD mice between ages 10 and 18 weeks were treated with a BAFF-blocking monoclonal antibody, Sandy-2 or an isotype control. Dryness was measured by the stimulated salivary flow. Salivary lymphocytic infiltrates were assessed by immunohistochemistry. Blood, SGs, spleen and lymph-node lymphocyte subpopulations were analysed by flow cytometry. SG mRNA expression was analysed by transcriptomic analysis. RESULTS BAFF inhibition significantly decreased SG lymphocytic infiltrates, which was inversely correlated with salivary flow. The treatment markedly decreased B-cell number in SGs, blood, lymph nodes and spleen and increased Foxp3+ regulatory and CD3+CD4-CD8- double negative T-cell numbers in SGs. CONCLUSION A monoclonal antibody blocking BAFF and depleting B cells had therapeutic effectiveness in the NOD mouse model of SjD. The increase in regulatory T-lymphocyte populations might underlie the efficacy of this treatment.
Collapse
Affiliation(s)
- Renaud Felten
- Service de Rhumatologie - Centre National de Référence des maladies auto-immuneset et auto-inflammatoires systémiques rares RESO, Hopitaux universitaires de Strasbourg, Strasbourg, France
- Immunologie, Immunopathologie et Chimie Thérapeutique, CNRS UPR 3572, IBMC, Strasbourg, France
| | - Anne-Perrine Foray
- Université Paris Cité, CNRS UMR 8253, INSERM UMR 1151, Institut Necker Enfants Malades-INEM, Paris, France
| | - Pascal Schneider
- Department of Immunobiology, University of Lausanne, Lausanne, Switzerland
| | - Cindy Marquet
- Université Paris Cité, CNRS UMR 8253, INSERM UMR 1151, Institut Necker Enfants Malades-INEM, Paris, France
| | - Coralie Pecquet
- Université Paris Cité, CNRS UMR 8253, INSERM UMR 1151, Institut Necker Enfants Malades-INEM, Paris, France
| | - Fanny Monneaux
- Immunologie, Immunopathologie et Chimie Thérapeutique, CNRS UPR 3572, IBMC, Strasbourg, France
| | - Hélène Dumortier
- Immunologie, Immunopathologie et Chimie Thérapeutique, CNRS UPR 3572, IBMC, Strasbourg, France
| | - Jean Sibilia
- Service de Rhumatologie - Centre National de Référence des maladies auto-immuneset et auto-inflammatoires systémiques rares RESO, Hopitaux universitaires de Strasbourg, Strasbourg, France
| | - Fabrice Valette
- Université Paris Cité, CNRS UMR 8253, INSERM UMR 1151, Institut Necker Enfants Malades-INEM, Paris, France
| | - Lucienne Chatenoud
- Université Paris Cité, CNRS UMR 8253, INSERM UMR 1151, Institut Necker Enfants Malades-INEM, Paris, France
| | - Jacques-Eric Gottenberg
- Service de Rhumatologie - Centre National de Référence des maladies auto-immuneset et auto-inflammatoires systémiques rares RESO, Hopitaux universitaires de Strasbourg, Strasbourg, France
- Immunologie, Immunopathologie et Chimie Thérapeutique, CNRS UPR 3572, IBMC, Strasbourg, France
| |
Collapse
|
8
|
Kato H, Kahlenberg JM. Emerging biologic therapies for systemic lupus erythematosus. Curr Opin Rheumatol 2024; 36:169-175. [PMID: 38299618 DOI: 10.1097/bor.0000000000001003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2024]
Abstract
PURPOSE OF REVIEW The approval of belimumab and anifrolumab has expanded the scope of treatment for systemic lupus erythematosus (SLE) patients. However, many patients remain refractory to currently available therapies and suffer from drug toxicities. This review will discuss approved and target-specific therapeutics in development that bring hope for better SLE treatments. RECENT FINDINGS Since the last review on this subject in the journal, the FDA has approved anifrolumab and belimumab for SLE and lupus nephritis (LN), respectively. A fully humanized anti-CD20, obinutuzumab, met the primary end point in a phase II trial in LN. A Tyk2 inhibitor, deucravacitinib, and an antibody targeting plasmacytoid dendritic cells, litifilimab, met the primary end point in phase II trials in SLE and cutaneous lupus erythematosus (CLE). Ustekinumab and baricitinib met the primary end point in phase II but not in phase III trials. SUMMARY While many drug candidates which met the end points in phase II trials have failed phase III trials, the number of target-specific therapies for SLE has continued to expand.
Collapse
Affiliation(s)
- Hiroshi Kato
- University of Michigan Lupus Program, Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | | |
Collapse
|
9
|
Cui W, Tian Y, Huang G, Zhang X, Li F, Liu X. Clinical research progress of novel biologics for the treatment of lupus nephritis. Clin Exp Med 2023; 23:4153-4162. [PMID: 37481481 DOI: 10.1007/s10238-023-01143-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Accepted: 07/12/2023] [Indexed: 07/24/2023]
Abstract
Systemic lupus erythematosus (SLE) is a chronic autoimmune disease characterized by the loss of immune tolerance. Lupus nephritis (LN) is one of the most common manifestations of severe organ damage in SLE, and also an important cause of disability and death. Its pathogenesis is associated with immune abnormalities such as immune cells, cytokines, and immune complex deposition. Traditional immunosuppressive therapy has been unable to meet the treatment needs of patients while bringing them toxic effects. In recent years, targeted therapies have emerged, and several novel biologics have gradually entered people's sight. This review will briefly introduce the pathogenesis of LN and the mechanism of biological targets, and summarize and analyze the clinical trials of new biologics for treating LN. Although not all biologics show positive results in clinical trials, the experience learned from these trials can help researchers adjust and plan future trial programs to seek better treatment methods.
Collapse
Affiliation(s)
- Wenyan Cui
- The Second Hospital of Hebei Medical University, Shijiazhuang City, Hebei Province, China
| | - Yunfei Tian
- The University of Hong Kong, Pok Fu Lam, Hong Kong, China
| | - Guangliang Huang
- The Second Hospital of Hebei Medical University, Shijiazhuang City, Hebei Province, China
| | - Xinhui Zhang
- The Second Hospital of Hebei Medical University, Shijiazhuang City, Hebei Province, China
| | - Feigao Li
- The Second Hospital of Hebei Medical University, Shijiazhuang City, Hebei Province, China
| | - Xiuju Liu
- The Second Hospital of Hebei Medical University, Shijiazhuang City, Hebei Province, China.
| |
Collapse
|
10
|
Li F, Cui W, Huang G, Tian Y, Zhang X, He W, Sun Q, Zhao X, Zhao Y, Li D, Liu X, Liu X. Efficacy and safety of novel biologics in the treatment of lupus nephritis based on registered clinical trials: a systematic review and network meta-analysis. Clin Exp Med 2023; 23:3011-3018. [PMID: 37462818 DOI: 10.1007/s10238-023-01132-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Accepted: 06/26/2023] [Indexed: 11/02/2023]
Abstract
To compare the clinical effectiveness and safety of novel biologics for the treatment of lupus nephritis based on a reticulated meta-analysis approach. Registered clinical trials in 4 major databases (PubMed, Embase, Web of Science, The Cochrane Register of Clinical Trials) and ClinicalTrials.gov were systematically searched with a search time frame of build to June 2022. And we screened registered randomized controlled clinical trials of biologics for the treatment of lupus nephritis according to the protocol's nadir criteria. Trials were evaluated for quality using the Cochrane Risk of Bias Assessment Tool, and data were statistically analyzed using Stata 16.0 and Review Manager 5.3 software to compare and rank differences in effectiveness and safety between the biologics. A total of 10 registered randomized controlled clinical trials involving 2148 subjects were included in this study. The interventions were ranked from best to worst in terms of the primary outcome indicator of effectiveness, renal complete remission: belimumab > anifrolumab (900 + 300) mg > obinutuzumab > ocrelizumab 400 mg > abatacept 30/10 mg/kg > belimumab + rituximab > abatacept 10/10 mg/kg > abatacept (30/10 + 10/10) mg/kg > placeo > ocrelizumab 1000 mg > rituximab > anifrolumab 300 mg, belimumab was superior to placebo [OR = 1.75, 95% CI (1.13, 2.70)] and anifrolumab 300 mg [OR = 3.27, 95% CI (1.05, 10.14)], anifrolumab (900 + 300) mg was superior to anifrolumab 300 mg [OR = 3.56, 95% CI (1.30, 9.76)], and all were statistically significant. The ranking of each intervention in terms of overall renal remission for secondary outcome indicators from best to worst was: obinutuzumab > belimumab + rituximab > anifrolumab (900 + 300) mg > ocrelizumab 1000 mg > ocrelizumab 400 mg > belimumab > rituximab 1000 mg > abatacept 30/10 mg/kg > abatacept (30/10 + 10/10) mg/kg > placeo > abatacept 10/10 mg/kg > anifrolumab 300 mg, obinutuzumab was superior to placebo [OR = 2.27, 95% CI (1.11, 4.67)] and belimumab was also superior to placebo [OR = 1.56, 95% CI (1.07, 2.27)], and all were statistically significant. In terms of safety, with a focus on serious adverse events and serious infections, the results were: Serious adverse events at 1 year of monitoring occurred better with ocrelizumab 1000 mg than ocrelizumab 400 mg [OR = 0.51, 95% CI (0.29, 0.89)] and were statistically different; serious adverse events at 2 years of monitoring infection adverse events occurred better with obinutuzumab than with abatacept (30/10 + 10/10) mg/kg [OR = 0.24, 95% CI (0.07, 0.81)] and were statistically different. The safety of the new biologics in combination with conventional standard therapies is generally good, but it is belimumab and obinutuzumab that are most effective in achieving complete and overall remission in the kidney. This study protocol has been registered with PROSPERO, with a registration number of CRD42021262498.
Collapse
Affiliation(s)
- Feigao Li
- The Second Hospital of Hebei Medical University, No. 215, Heping West Road, Shijiazhuang, 050000, People's Republic of China
| | - Wenyan Cui
- The Second Hospital of Hebei Medical University, No. 215, Heping West Road, Shijiazhuang, 050000, People's Republic of China
| | - Guangliang Huang
- The Second Hospital of Hebei Medical University, No. 215, Heping West Road, Shijiazhuang, 050000, People's Republic of China
| | - Yunfei Tian
- The University of Hong Kong, Hong Kong, China
| | - Xinhui Zhang
- The Second Hospital of Hebei Medical University, No. 215, Heping West Road, Shijiazhuang, 050000, People's Republic of China
| | - Wenjuan He
- The Second Hospital of Hebei Medical University, No. 215, Heping West Road, Shijiazhuang, 050000, People's Republic of China
| | - Qian Sun
- The Second Hospital of Hebei Medical University, No. 215, Heping West Road, Shijiazhuang, 050000, People's Republic of China
| | - Xiaojuan Zhao
- The Second Hospital of Hebei Medical University, No. 215, Heping West Road, Shijiazhuang, 050000, People's Republic of China
| | - Yonghong Zhao
- The Second Hospital of Hebei Medical University, No. 215, Heping West Road, Shijiazhuang, 050000, People's Republic of China
| | - Dan Li
- The Second Hospital of Hebei Medical University, No. 215, Heping West Road, Shijiazhuang, 050000, People's Republic of China
| | - Xizhe Liu
- Hebei Medical University Third Affiliated Hospital, Shijiazhuang, China
| | - Xiuju Liu
- The Second Hospital of Hebei Medical University, No. 215, Heping West Road, Shijiazhuang, 050000, People's Republic of China.
| |
Collapse
|
11
|
Maeda S, Hashimoto H, Maeda T, Tamechika SY, Isogai S, Naniwa T, Niimi A. High-dimensional analysis of T-cell profiling variations following belimumab treatment in systemic lupus erythematosus. Lupus Sci Med 2023; 10:e000976. [PMID: 37802602 PMCID: PMC10565340 DOI: 10.1136/lupus-2023-000976] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2023] [Accepted: 09/06/2023] [Indexed: 10/10/2023]
Abstract
OBJECTIVE This study sought to elucidate the molecular impacts of belimumab (BEL) treatment on T-cell immune profiling in SLE. METHODS We used mass cytometry with 25 marker panels for T-cell immune profiling in peripheral blood T cells (CD3+) from 22 patients with BEL-treated SLE and 20 controls with non-BEL-treated SLE. An unsupervised machine-learning clustering, FlowSOM, was used to identify 39 T-cell clusters (TCLs; TCL01-TCL39). TCLs (% of CD3+) showing significant (p<0.05) associations with BEL treatment (BEL-TCL) were selected by a linear mixed-effects model for comparing groups of time-series data. Furthermore, we analysed the association between BEL treatment and variations in regulatory T-cell (Treg) phenotypes, and the ratio of other T-cell subsets to Treg as secondary analysis. RESULTS Clinical outcomes: BEL treatment was associated with a decrease in daily prednisolone use (coef=-0.1769, p=0.00074), and an increase in serum CH50 (coef=0.4653, p=0.003), C3 (coef=1.1047, p=0.00001) and C4 (coef=0.2990, p=0.00157) levels. Molecular effects: five distinct BEL-TCLs (TCL 04, 07, 11, 12 and 27) were identified. Among these, BEL-treated patients exhibited increased proportions in the Treg-like cluster TCL11 (coef=0.404, p=0.037) and two naïve TCLs (TCL04 and TCL07). TCL27 showed increased levels (coef=0.222, p=0.037) inversely correlating with baseline C3 levels. Secondary analyses revealed associations between BEL treatment and an increase in Tregs (coef=1.749, p=0.0044), elevated proportions of the fraction of Tregs with inhibitory function (fTregs, coef=0.7294, p=0.0178) and changes in peripheral helper T cells/fTreg (coef=-4.475, p=0.0319) and T helper 17/fTreg ratios (coef=-6.7868, p=0.0327). Additionally, BEL was linked to variations in T-cell immunoglobulin and mucin domain-containing protein-3 expression (coef=0.2422, p=0.039). CONCLUSIONS The study suggests an association between BEL treatment and variations in T cells, particularly Tregs, in SLE pathologies involving various immune cells.
Collapse
Affiliation(s)
- Shinji Maeda
- Department of Respiratory Medicine, Allergy and Clinical Immunology, Nagoya City University Graduate School of Medical Sciences and Medical School, Nagoya, Japan
| | - Hiroya Hashimoto
- Clinical Research Management Center, Nagoya City University Hospital, Nagoya, Japan
| | - Tomoyo Maeda
- Department of Respiratory Medicine, Allergy and Clinical Immunology, Nagoya City University Graduate School of Medical Sciences and Medical School, Nagoya, Japan
| | - Shin-Ya Tamechika
- Department of Respiratory Medicine, Allergy and Clinical Immunology, Nagoya City University Graduate School of Medical Sciences and Medical School, Nagoya, Japan
| | - Shuntaro Isogai
- Department of Respiratory Medicine, Allergy and Clinical Immunology, Nagoya City University Graduate School of Medical Sciences and Medical School, Nagoya, Japan
| | - Taio Naniwa
- Department of Respiratory Medicine, Allergy and Clinical Immunology, Nagoya City University Graduate School of Medical Sciences and Medical School, Nagoya, Japan
| | - Akio Niimi
- Department of Respiratory Medicine, Allergy and Clinical Immunology, Nagoya City University Graduate School of Medical Sciences and Medical School, Nagoya, Japan
| |
Collapse
|
12
|
Bogers L, Kuiper KL, Smolders J, Rip J, van Luijn MM. Epstein-Barr virus and genetic risk variants as determinants of T-bet + B cell-driven autoimmune diseases. Immunol Lett 2023; 261:66-74. [PMID: 37451321 DOI: 10.1016/j.imlet.2023.07.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Revised: 06/07/2023] [Accepted: 07/10/2023] [Indexed: 07/18/2023]
Abstract
B cells expressing the transcription factor T-bet are found to have a protective role in viral infections, but are also considered major players in the onset of different types of autoimmune diseases. Currently, the exact mechanisms driving such 'atypical' memory B cells to contribute to protective immunity or autoimmunity are unclear. In addition to general autoimmune-related factors including sex and age, the ways T-bet+ B cells instigate autoimmune diseases may be determined by the close interplay between genetic risk variants and Epstein-Barr virus (EBV). The impact of EBV on T-bet+ B cells likely relies on the type of risk variants associated with each autoimmune disease, which may affect their differentiation, migratory routes and effector function. In this hypothesis-driven review, we discuss the lines of evidence pointing to such genetic and/or EBV-mediated influence on T-bet+ B cells in a range of autoimmune diseases, including systemic lupus erythematosus (SLE) and multiple sclerosis (MS). We provide examples of how genetic risk variants can be linked to certain signaling pathways and are differentially affected by EBV to shape T-bet+ B-cells. Finally, we propose options to improve current treatment of B cell-related autoimmune diseases by more selective targeting of pathways that are critical for pathogenic T-bet+ B-cell formation.
Collapse
Affiliation(s)
- Laurens Bogers
- MS Center ErasMS, Department of Immunology, Erasmus MC, University Medical Center Rotterdam, Wytemaweg 80, Rotterdam 3015 CN, The Netherlands
| | - Kirsten L Kuiper
- MS Center ErasMS, Department of Immunology, Erasmus MC, University Medical Center Rotterdam, Wytemaweg 80, Rotterdam 3015 CN, The Netherlands
| | - Joost Smolders
- MS Center ErasMS, Department of Immunology, Erasmus MC, University Medical Center Rotterdam, Wytemaweg 80, Rotterdam 3015 CN, The Netherlands; MS Center ErasMS, Department of Neurology, Erasmus MC, University Medical Center Rotterdam, Rotterdam 3015 CN, The Netherlands; Netherlands Institute for Neuroscience, Neuroimmunology research group, Amsterdam 1105 BA, The Netherlands
| | - Jasper Rip
- MS Center ErasMS, Department of Immunology, Erasmus MC, University Medical Center Rotterdam, Wytemaweg 80, Rotterdam 3015 CN, The Netherlands
| | - Marvin M van Luijn
- MS Center ErasMS, Department of Immunology, Erasmus MC, University Medical Center Rotterdam, Wytemaweg 80, Rotterdam 3015 CN, The Netherlands.
| |
Collapse
|
13
|
Iwata S, Hajime Sumikawa M, Tanaka Y. B cell activation via immunometabolism in systemic lupus erythematosus. Front Immunol 2023; 14:1155421. [PMID: 37256149 PMCID: PMC10225689 DOI: 10.3389/fimmu.2023.1155421] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 04/27/2023] [Indexed: 06/01/2023] Open
Abstract
Systemic lupus erythematosus (SLE) is an inflammatory autoimmune disease involving multiple organs in which B cells perform important functions such as antibody and cytokine production and antigen presentation. B cells are activated and differentiated by the primary B cell receptor, co-stimulatory molecule signals-such as CD40/CD40L-, the Toll-like receptors 7,9, and various cytokine signals. The importance of immunometabolism in the activation, differentiation, and exerting functions of B cells and other immune cells has been widely reported in recent years. However, the regulatory mechanism of immunometabolism in B cells and its involvement in SLE pathogenesis remain elusive. Similarly, the importance of the PI3K-Akt-mTOR signaling pathway, glycolytic system, and oxidative phosphorylation has been demonstrated in the mechanisms of B cell immunometabolic activation, mainly in mouse studies. However, the activation of the mTOR pathway in B cells in patients with SLE, the induction of plasmablast differentiation through metabolic and transcription factor regulation by mTOR, and the involvement of this phenomenon in SLE pathogenesis are unclear. In our studies using activated B cells derived from healthy donors and from patients with SLE, we observed that methionine, an essential amino acid, is important for mTORC1 activation. Further, we observed that splenic tyrosine kinase and mTORC1 activation synergistically induce EZH2 expression and plasmablasts by suppressing BACH2 expression through epigenomic modification. Additionally, we identified another mechanism by which the glutaminolysis-induced enhancement of mitochondrial function promotes plasmablast differentiation in SLE. In this review, we focused on the SLE exacerbation mechanisms related to the activation of immune cells-especially B cells-and immunometabolism and reported the latest findings in the field.
Collapse
Affiliation(s)
- Shigeru Iwata
- Department of Rheumatology and Clinical Immunology, Wakayama Medical University, Wakayama, Japan
- First Department of Internal Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Maiko Hajime Sumikawa
- First Department of Internal Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Yoshiya Tanaka
- First Department of Internal Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| |
Collapse
|
14
|
Deng L, Xu G. Update on the Application of Monoclonal Antibody Therapy in Primary Membranous Nephropathy. Drugs 2023; 83:507-530. [PMID: 37017915 DOI: 10.1007/s40265-023-01855-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/27/2023] [Indexed: 04/06/2023]
Abstract
When first introduced, rituximab (RTX), a chimeric anti-CD20 monoclonal antibody, brought about an alternative therapeutic paradigm for primary membranous nephropathy (PMN). Rituximab was shown to be effective and safe in PMN patients with kidney dysfunction, with. patients receiving second-line rituximab therapy achieving remission as effectively as those patients who had not previously received immunotherapy. No safety issues were reported. The B cell-driven protocol seems to be as efficient as the 375 mg/m2 × 4 regimen or 1 g × 2 regimen in achieving B cell depletion and remission, but patients with high M-type phospholipase A2 receptor (PLA2R) antibody levels may benefit from a higher dose of rituximab. While rituximab added another therapeutic option to the treatment regimen, it does have limitations as 20 to 40% of patients do not respond. Not all patients respond to RTX therapy for lymphoproliferative disorders either, therefore further novel anti-CD20 monoclonal antibodies have been developed and these may provide alternative therapeutic options for PMN. Ofatumumab, a fully human monoclonal antibody, specifically recognizes an epitope encompassing both the small and large extracellular loops of the CD20 molecule, resulting in increased complement-dependent cytotoxic activity. Ocrelizumab binds an alternative but overlapping epitope region to rituximab and displays enhanced antibody-dependent cellular cytotoxic (ADCC) activities. Obinutuzumab is designed to have a modified elbow-hinge amino acid sequence, leading to increased direct cell death induction and ADCC activities. In PMN clinical studies, ocrelizumab and obinutuzumab showed promising results, while ofatumumab displayed mixed results. However, there is a lack of randomized controlled trials with large samples, especially direct head-to-head comparisons. Alternative molecular mechanisms have been suggested in this context to explore novel therapeutic strategies. B cell activator-targeted, plasma cell-targeted and complement-directed treatments may lead to novel therapy paradigms for PMN. Exploratory strategies for the use of drugs with different mechanisms, such as a combination of rituximab and cyclophosphamide and a steroid, a combination of rituximab and a calcineurin inhibitor, may provide more rapid and efficient remission, but the combination of standard immunosuppression with rituximab could increase infection risk.
Collapse
Affiliation(s)
- Le Deng
- Department of Nephrology, The Second Affiliated Hospital of Nanchang University, No. 1, Minde Road, Donghu District, Nanchang, 330006, Jiangxi, China
| | - Gaosi Xu
- Department of Nephrology, The Second Affiliated Hospital of Nanchang University, No. 1, Minde Road, Donghu District, Nanchang, 330006, Jiangxi, China.
| |
Collapse
|
15
|
Mustafa AJ, Balaky HM, Ismail PA, Abdalla HO, Muhammed KM. Serum Calprotectin and B-cell activating factor are potential biomarkers for Helicobacter pylori infection. JOURNAL OF BIOLOGICAL RESEARCH - BOLLETTINO DELLA SOCIETÀ ITALIANA DI BIOLOGIA SPERIMENTALE 2023. [DOI: 10.4081/jbr.2023.10803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/03/2023]
Abstract
Humans always mount a robust immune response to the bacterial infection caused by Helicobacter pylori, which causes various gastrointestinal tract infections. Calprotectin (CALP) and B-Cell Activating Factor (BAFF) are inflammatory biomarkers having a role in the gastrointestinal neutrophilic response to bacterial infection. The study was designed to assess serum CALP and BAFF as inflammatory biomarkers in H. pylori infection and peptic ulcer patients. The current study comprised 112 people, including 62 H. pylori-infected patients (34 men and 28 women) who were clinically diagnosed with H. pylori infection via testing positive for the H. pylori stool antigen test; they were compared to a control group of 50 healthy people (34 men and 16 women) who were age and gender-matched to H. pylori-infected patients. The serum level of CALP and BAFF were assayed using the ELISA technique. The biochemical parameters were statistically compared between patients and controls by unpaired Man-Whitney U t-test and Receiver Operating Characteristic (ROC) curve analysis. There was a significant elevation of serum CALP in H. pylori-infected patients [116.4(120.7), p=0.0132] in comparison to healthy controls [99.50(115.8)]. Similarly, there was a significant elevation of serum BAFF concentration in H. pylori-infected patients [485.7(367.1), p=0.0014] in comparison to healthy controls [444.5(513.0)]. The ROC curve analysis suggests serum CALP and BAFF as reasonable inflammatory biomarkers for H. pylori infection with statistically significant (p=0.0135, p=0.0015) area under the ROC curve of (0.6361, 0.6748), respectively. CALP and BAFF are potent inflammatory biomarkers involved in the development and etiology of H. pylori infection. Serum CALP and BAFF levels could be used as biomarkers for chronic inflammation induced by H. pylori. CALP and BAFF biomarkers can be combined to diagnose and predict the prognosis of H. pylori infection.
Collapse
|
16
|
Harsini S, Rezaei N. Autoimmune diseases. Clin Immunol 2023. [DOI: 10.1016/b978-0-12-818006-8.00001-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
|
17
|
Scurt FG, Bose K, Hammoud B, Brandt S, Bernhardt A, Gross C, Mertens PR, Chatzikyrkou C. Old known and possible new biomarkers of ANCA-associated vasculitis. J Autoimmun 2022; 133:102953. [PMID: 36410262 DOI: 10.1016/j.jaut.2022.102953] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 11/06/2022] [Accepted: 11/06/2022] [Indexed: 11/19/2022]
Abstract
Antineutrophil cytoplasm antibody (ANCA)-associated vasculitis (AAV) comprises a group of multisystem disorders involving severe, systemic, small-vessel vasculitis with short- and long term serious and life-threating complications. Despite the simplification of treatment, fundamental aspects concerning assessment of its efficacy and its adaptation to encountered complications or to the relapsing/remitting/subclinical disease course remain still unknown. The pathogenesis of AAV is complex and unique, and despite the progress achieved in the last years, much has not to be learnt. Foremost, there is still no accurate marker enabling us to monitoring disease and guide therapy. Therefore, the disease management relays often on clinical judgment and follows a" trial and error approach". In the recent years, an increasing number of new molecules s have been explored and used for this purpose including genomics, B- and T-cell subpopulations, complement system factors, cytokines, metabolomics, biospectroscopy and components of our microbiome. The aim of this review is to discuss both the role of known historical and clinically established biomarkers of AAV, as well as to highlight potential new ones, which could be used for timely diagnosis and monitoring of this devastating disease, with the goal to improve the effectiveness and ameliorate the complications of its demanding therapy.
Collapse
Affiliation(s)
- Florian G Scurt
- University Clinic for Nephrology and Hypertension, Diabetology and Endocrinology, University Hospital Magdeburg, Otto-von-Guericke University Magdeburg, Germany.
| | - K Bose
- Department of Gastroenterology, Hepatology and Infectious Diseases, University Hospital Magdeburg, Otto-von-Guericke University Magdeburg, Germany
| | - Ben Hammoud
- University Clinic for Nephrology and Hypertension, Diabetology and Endocrinology, University Hospital Magdeburg, Otto-von-Guericke University Magdeburg, Germany
| | - S Brandt
- University Clinic for Nephrology and Hypertension, Diabetology and Endocrinology, University Hospital Magdeburg, Otto-von-Guericke University Magdeburg, Germany
| | - A Bernhardt
- University Clinic for Nephrology and Hypertension, Diabetology and Endocrinology, University Hospital Magdeburg, Otto-von-Guericke University Magdeburg, Germany
| | - C Gross
- University Clinic for Nephrology and Hypertension, Diabetology and Endocrinology, University Hospital Magdeburg, Otto-von-Guericke University Magdeburg, Germany
| | - Peter R Mertens
- University Clinic for Nephrology and Hypertension, Diabetology and Endocrinology, University Hospital Magdeburg, Otto-von-Guericke University Magdeburg, Germany
| | | |
Collapse
|
18
|
Madayag K, Incrocci R, Swanson‐Mungerson M. The impact of Epstein-Barr virus latent membrane protein 2A on the production of B cell activating factor of the tumor necrosis factor family (BAFF), APRIL and their receptors. Immun Inflamm Dis 2022; 10:e729. [PMID: 36301035 PMCID: PMC9597489 DOI: 10.1002/iid3.729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 10/05/2022] [Accepted: 10/13/2022] [Indexed: 11/06/2022] Open
Abstract
INTRODUCTION Epstein-Barr virus (EBV) establishes a lifelong infection in human B cells where the virus consistently expresses Latent Membrane Protein 2A (LMP2A) to promote B cell survival. A prior study indicates that LMP2A may increase the production of the pro-survival factor, B cell Activating Factor of the tumor necrosis factor family (BAFF), which could also indirectly increase B cell survival. The current study sought to extend these findings and determine if LMP2A increased BAFF production and/or the responsiveness of LMP2A-expressing cells to this cytokine. METHODS Four independently derived LMP2A-negative and -positive B cell lymphoma cell lines were analyzed for BAFF and APRIL levels by both ELISA and Western Blot analysis. Additionally, flow cytometric analysis measured any LMP2A-dependent changes in the receptors for BAFF and APRIL (BAFF-R, transmembrane activator and calcium-modulator and cyclophilin ligand interactor [TACI], B cell maturation antigen [BCMA]) in both LMP2A-negative and -positive B cell lymphoma cell lines. RESULTS In contrast to previous reports, our data indicate that LMP2A does not increase the expression of BAFF or APRIL by Western blot analysis or ELISA. Additionally, flow cytometric analysis indicates that LMP2A does not influence the expression of the receptors for BAFF and APRIL: TACI, BAFF-R, and BCMA. CONCLUSION Therefore, these data suggest that while EBV utilizes other latency proteins to regulate BAFF production, EBV does not appear to use LMP2A to enhance BAFF-or APRIL-dependent survival to promote EBV latency.
Collapse
Affiliation(s)
- Kevin Madayag
- Department of Biomedical SciencesCollege of Graduate StudiesDowners GroveIllinoisUSA
| | - Ryan Incrocci
- Department of Microbiology and Immunology, College of Graduate StudiesMidwestern UniversityDowners GroveIllinoisUSA
| | - Michelle Swanson‐Mungerson
- Department of Microbiology and Immunology, College of Graduate StudiesMidwestern UniversityDowners GroveIllinoisUSA
| |
Collapse
|
19
|
Lee S, Yang JI, Lee JH, Lee HW, Kim TJ. Low-Level Expression of CD138 Marks Naturally Arising Anergic B Cells. Immune Netw 2022; 22:e50. [PMID: 36627940 PMCID: PMC9807963 DOI: 10.4110/in.2022.22.e50] [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: 08/30/2022] [Revised: 10/07/2022] [Accepted: 10/14/2022] [Indexed: 12/30/2022] Open
Abstract
Autoreactive B cells are not entirely deleted, but some remain as immunocompetent or anergic B cells. Although the persistence of autoreactive B cells as anergic cells has been shown in transgenic mouse models with the expression of B cell receptor (BCR) reactive to engineered self-antigen, the characterization of naturally occurring anergic B cells is important to identify them and understand their contribution to immune regulation or autoimmune diseases. We report here that a low-level expression of CD138 in the splenic B cells marks naturally arising anergic B cells, not plasma cells. The CD138int B cells consisted of IgMlowIgDhigh follicular (FO) B cells and transitional 3 B cells in homeostatic conditions. The CD138int FO B cells showed an anergic gene expression profile shared with that of monoclonal anergic B cells expressing engineered BCRs and the gene expression profile was different from those of plasma cells, age-associated B cells, or germinal center B cells. The anergic state of the CD138int FO B cells was confirmed by attenuated Ca2+ response and failure to upregulate CD69 upon BCR engagement with anti-IgM, anti-IgD, anti-Igκ, or anti-IgG. The BCR repertoire of the CD138int FO B cells was distinct from that of the CD138- FO B cells and included some class-switched B cells with low-level somatic mutations. These findings demonstrate the presence of polyclonal anergic B cells in the normal mice that are characterized by low-level expression of CD138, IgM downregulation, reduced Ca2+ and CD69 responses upon BCR engagement, and distinct BCR repertoire.
Collapse
Affiliation(s)
| | | | - Joo Hee Lee
- Department of Immunology, Sungkyunkwan University School of Medicine, Suwon 16419, Korea
| | - Hyun Woo Lee
- Department of Immunology, Sungkyunkwan University School of Medicine, Suwon 16419, Korea
| | - Tae Jin Kim
- Department of Immunology, Sungkyunkwan University School of Medicine, Suwon 16419, Korea
| |
Collapse
|
20
|
Chawla S, Barman P, Tyagi R, Jindal AK, Sharma S, Rawat A, Singh S. Autoimmune Cytopenias in Common Variable Immunodeficiency Are a Diagnostic and Therapeutic Conundrum: An Update. Front Immunol 2022; 13:869466. [PMID: 35795667 PMCID: PMC9251126 DOI: 10.3389/fimmu.2022.869466] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Accepted: 05/20/2022] [Indexed: 11/13/2022] Open
Abstract
Common variable immunodeficiency (CVID) is the most common symptomatic primary immunodeficiency (PID). CVID is a heterogenous condition and clinical manifestations may vary from increased susceptibility to infections to autoimmune manifestations, granulomatous disease, polyclonal lymphoproliferation, and increased risk of malignancy. Autoimmune manifestations may, at times, be the first and only clinical presentation of CVID, resulting in diagnostic dilemma for the treating physician.Autoimmune cytopenias (autoimmune haemolytic anaemia and/or thrombocytopenia) are the most common autoimmune complications seen in patients with CVID. Laboratory investigations such as antinuclear antibodies, direct Coomb’s test and anti-platelet antibodies may not be useful in patients with CVID because of lack of specific antibody response. Moreover, presence of autoimmune cytopenias may pose a significant therapeutic challenge as use of immunosuppressive agents can be contentious in these circumstances. It has been suggested that serum immunoglobulins must be checked in all patients presenting with autoimmune cytopenia such as immune thrombocytopenia or autoimmune haemolytic anaemia.It has been observed that patients with CVID and autoimmune cytopenias have a different clinical and immunological profile as compared to patients with CVID who do not have an autoimmune footprint. Monogenic defects have been identified in 10-50% of all patients with CVID depending upon the population studied. Monogenic defects are more likely to be identified in patients with CVID with autoimmune complications. Common genetic defects that may lead to CVID with an autoimmune phenotype include nuclear factor kappa B subunit 1 (NF-kB1), Lipopolysaccharide (LPS)-responsive beige-like anchor protein (LRBA), cytotoxic T lymphocyte antigen 4 (CTLA4), Phosphoinositide 3-kinase (PI3K), inducible T-cell costimulatory (ICOS), IKAROS and interferon regulatory factor-2 binding protein 2 (IRF2BP2).In this review, we update on recent advances in pathophysiology and management of CVID with autoimmune cytopenias.
Collapse
|
21
|
Merino-Vico A, van Hamburg JP, Tas SW. B Lineage Cells in ANCA-Associated Vasculitis. Int J Mol Sci 2021; 23:387. [PMID: 35008813 PMCID: PMC8745114 DOI: 10.3390/ijms23010387] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 12/21/2021] [Accepted: 12/28/2021] [Indexed: 12/18/2022] Open
Abstract
Anti-neutrophil cytoplasmic antibody (ANCA)-associated vasculitis (AAV) is a systemic autoimmune disease that affects small sized blood vessels and can lead to serious complications in the lungs and kidneys. The prominent presence of ANCA autoantibodies in this disease implicates B cells in its pathogenesis, as these are the precursors of the ANCA-producing plasma cells (PCs). Further evidence supporting the potential role of B lineage cells in vasculitis are the increased B cell cytokine levels and the dysregulated B cell populations in patients. Confirmation of the contribution of B cells to pathology arose from the beneficial effect of anti-CD20 therapy (i.e., rituximab) in AAV patients. These anti-CD20 antibodies deplete circulating B cells, which results in amelioration of disease. However, not all patients respond completely, and this treatment does not target PCs, which can maintain ANCA production. Hence, it is important to develop more specific therapies for AAV patients. Intracellular signalling pathways may be potential therapeutic targets as they can show (disease-specific) alterations in certain B lineage cells, including pathogenic B cells, and contribute to differentiation and survival of PCs. Preliminary data on the inhibition of certain signalling molecules downstream of receptors specific for B lineage cells show promising therapeutic effects. In this narrative review, B cell specific receptors and their downstream signalling molecules that may contribute to pathology in AAV are discussed, including the potential to therapeutically target these pathways.
Collapse
Affiliation(s)
- Ana Merino-Vico
- Department of Rheumatology and Clinical Immunology, Amsterdam Rheumatology and Immunology Center, Amsterdam University Medical Centers, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands; (A.M.-V.); (J.P.v.H.)
- Department of Experimental Immunology, Amsterdam University Medical Centers, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| | - Jan Piet van Hamburg
- Department of Rheumatology and Clinical Immunology, Amsterdam Rheumatology and Immunology Center, Amsterdam University Medical Centers, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands; (A.M.-V.); (J.P.v.H.)
- Department of Experimental Immunology, Amsterdam University Medical Centers, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| | - Sander W. Tas
- Department of Rheumatology and Clinical Immunology, Amsterdam Rheumatology and Immunology Center, Amsterdam University Medical Centers, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands; (A.M.-V.); (J.P.v.H.)
- Department of Experimental Immunology, Amsterdam University Medical Centers, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| |
Collapse
|
22
|
Role of B-Cell Activating Factor (BAFF) in Inflammatory Bowel Disease. Diagnostics (Basel) 2021; 12:diagnostics12010045. [PMID: 35054212 PMCID: PMC8774757 DOI: 10.3390/diagnostics12010045] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 12/21/2021] [Accepted: 12/21/2021] [Indexed: 12/12/2022] Open
Abstract
As early commencement of inflammatory bowel disease (IBD) treatment has been shown to substantially improve outcomes, it is of utmost importance to make a timely diagnosis of this disease. Despite undisputed sensitivity of fecal calprotectin, the most widely accepted IBD biomarker, in discriminating between irritable bowel syndrome (IBS) and IBD, as well as recognized role in monitoring disease activity and response to therapy, perhaps the biggest setback of calprotectin use in IBD is lack of specificity. Therefore, an additional biomarker in IBD is warranted. B-cell activating factor (BAFF), a member of the tumor necrosis factor (TNF) superfamily, recently emerged as a viable candidate for this role. So far, overproduction of BAFF has been observed in various autoimmune diseases, most notably in systemic lupus erythematosus, where BAFF-inhibitor belimumab was approved for treatment. As BAFF levels were also shown to correlate with indices of IBD, in this review we aimed to summarize the current evidence with respect to the role of BAFF in diagnosis and assessing the activity of IBD, as well as putative therapeutic implications that may arise from exploring of this relation.
Collapse
|
23
|
Zhang Y, Tian J, Xiao F, Zheng L, Zhu X, Wu L, Zhao C, Wang S, Rui K, Zou H, Lu L. B cell-activating factor and its targeted therapy in autoimmune diseases. Cytokine Growth Factor Rev 2021; 64:57-70. [DOI: 10.1016/j.cytogfr.2021.11.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 11/29/2021] [Accepted: 11/30/2021] [Indexed: 12/18/2022]
|
24
|
BAFF, involved in B cell activation through the NF-κB pathway, is related to disease activity and bone destruction in rheumatoid arthritis. Acta Pharmacol Sin 2021; 42:1665-1675. [PMID: 33483588 PMCID: PMC8463593 DOI: 10.1038/s41401-020-00582-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Accepted: 11/16/2020] [Indexed: 02/02/2023]
Abstract
B cell activating factor of TNF family (BAFF) is a member of TNF ligand superfamily and plays a key role in B cell homeostasis, proliferation, maturation, and survival. In this study, we detected BAFF level, the expressions of BAFF receptors and important molecules in NF-κB pathway in rheumatoid arthritis (RA) patients and analyzed the correlation between BAFF level and clinical variables, laboratory parameters or X-ray scores in order to elucidate the roles of BAFF in RA. A total of 50 RA patients and 50 healthy controls (HCs) were enrolled. We showed that the serum BAFF level in RA patients was significantly higher than that of HCs, and the percentages of B cell subsets (CD19+ B cells, CD19+CD27+ B cells, CD19+CD20+CD27+ B cells, and CD19+CD20-CD27+ B cells) in the serum of RA patients were significantly increased compared with those of HCs. The percentages of CD19+BAFFR+ B cells, CD19+ BCMA+ B cells, and CD19+ TACI+ B cells in RA patients were significantly increased compared with those in HCs. The expression of important molecules in the NF-κB pathway (MKK3, MKK6, p-P38, p-P65, TRAF2, and p52) was significantly higher in RA patients than in HCs, but p100 level in RA patients was lower than that in HCs. The serum BAFF level was positively correlated with C-reactive protein, rheumatoid factor, disease activity score (in 28 joints), swollen joint counts, tender joint counts, and X-ray scores. When normal B cells were treated with BAFF in vitro, the percentages of the B cell subset and the expression of BAFF receptors were significantly upregulated. BAFF also promoted the expression of MKK3, MKK6, p-P38, p-P65, TRAF2, and p52. In conclusion, this study demonstrates that BAFF level is correlated with the disease activity and bone destruction of RA. BAFF is involved in the differentiation, proliferation, and activation of B cells in RA through NF-κB signaling pathway, suggesting that BAFF might be an ideal therapeutic target for RA.
Collapse
|
25
|
Mahévas M, Azzaoui I, Crickx E, Canoui-Poitrine F, Gobert D, Languille L, Limal N, Guillaud C, Croisille L, Jeljeli M, Batteux F, Baloul S, Fain O, Pirenne F, Weill JC, Reynaud CA, Godeau B, Michel M. Efficacy, safety and immunological profile of combining rituximab with belimumab for adults with persistent or chronic immune thrombocytopenia: results from a prospective phase 2b trial. Haematologica 2021; 106:2449-2457. [PMID: 32817288 PMCID: PMC8409028 DOI: 10.3324/haematol.2020.259481] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Indexed: 12/24/2022] Open
Abstract
B-cell activating factor may be involved in the failure of B-cell depleting therapy with rituximab in immune thrombocytopenia (ITP) by promoting the emergence of splenic long-lived plasma cells. From results obtained in mouse models, we hypothesized that combining rituximab with sequential injections of belimumab could increase the rate of response at one year in patients with persistent or chronic ITP by preventing the emergence of these long-lived plasma cells. The study was a single-center, single arm, prospective phase 2b trial (RITUX-PLUS, NCT03154385) investigating the safety and efficacy of rituximab given at a fixed dose of 1,000 mg, two weeks apart, combined with five infusions of belimumab, 10 mg/kg at week 0 (W0)+2 days, W2+2 days, W4, W8 and W12 for adults with primary persistent or chronic ITP. The primary endpoint was the total number of patients achieving an overall response (complete response + response) at W52 according to a standard definition. In total, 15 non-splenectomized adults, nine (60%) with persistent IPT and six (40%) with chronic ITP, were included. No severe adverse event, infection, or severe hypogammaglobulinemia was observed. Thirteen patients achieved an initial overall response. At W52, 12 (80%) patients achieved an overall response, including ten (66.7%) with complete response. When compared with a cohort of patients receiving rituximab alone, the kinetics of B-cell repopulation appeared similar, but the number of circulating T follicular helper cells was significantly decreased with belimumab combination therapy. Combining rituximab and belimumab seems a promising strategy in ITP, with high efficacy and acceptable safety.
Collapse
Affiliation(s)
- Matthieu Mahévas
- Centre Hospitalier Universitaire Henri-Mondor, Université Paris Est Creteil, Creteil, France
| | | | - Etienne Crickx
- Centre Hospitalier Universitaire Henri-Mondor, Université Paris Est Creteil, Creteil, France
| | - Florence Canoui-Poitrine
- University Paris-Est, Paris Est Creteil University, CEpiA, Henri-Mondor Hospital, Creteil, France
| | | | - Laetitia Languille
- Centre Hospitalier Universitaire Henri-Mondor, Université Paris Est Creteil, Creteil, France
| | - Nicolas Limal
- Centre Hospitalier Universitaire Henri-Mondor, Université Paris Est Creteil, Creteil, France
| | - Constance Guillaud
- Centre Hospitalier Universitaire Henri-Mondor, Université Paris Est Creteil, Creteil, France
| | - Laure Croisille
- Service d'immunologie plaquettaire, Hopital Henri Mondor, Creteil, France
| | - Mohamed Jeljeli
- Service d'Immunologie biologique, Hopital Cochin, Institut Cochin, Paris, France
| | - Fréderic Batteux
- Service d'Immunologie biologique, Hopital Cochin, Institut Cochin, Paris, France
| | - Samia Baloul
- University Paris-Est, Paris Est Creteil University, CEpiA, Henri-Mondor Hospital, Creteil, France
| | - Olivier Fain
- Sorbonne Université, Service de Medecine Interne, Hopital Saint-Antoine, Paris, France
| | | | - Jean-Claude Weill
- Institut Necker Enfants Malades, Université Paris Descartes, Paris, France
| | | | - Bertrand Godeau
- Centre Hospitalier Universitaire Henri-Mondor, Université Paris Est Creteil, Creteil, France
| | - Marc Michel
- Centre Hospitalier Universitaire Henri-Mondor, Université Paris Est Creteil, Creteil, France
| |
Collapse
|
26
|
Neys SFH, Rip J, Hendriks RW, Corneth OBJ. Bruton's Tyrosine Kinase Inhibition as an Emerging Therapy in Systemic Autoimmune Disease. Drugs 2021; 81:1605-1626. [PMID: 34609725 PMCID: PMC8491186 DOI: 10.1007/s40265-021-01592-0] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/16/2021] [Indexed: 12/14/2022]
Abstract
Systemic autoimmune disorders are complex heterogeneous chronic diseases involving many different immune cells. A significant proportion of patients respond poorly to therapy. In addition, the high burden of adverse effects caused by "classical" anti-rheumatic or immune modulatory drugs provides a need to develop more specific therapies that are better tolerated. Bruton's tyrosine kinase (BTK) is a crucial signaling protein that directly links B-cell receptor (BCR) signals to B-cell activation, proliferation, and survival. BTK is not only expressed in B cells but also in myeloid cells, and is involved in many different signaling pathways that drive autoimmunity. This makes BTK an interesting therapeutic target in the treatment of autoimmune diseases. The past decade has seen the emergence of first-line BTK small-molecule inhibitors with great efficacy in the treatment of B-cell malignancies, but with unfavorable safety profiles for use in autoimmunity due to off-target effects. The development of second-generation BTK inhibitors with superior BTK specificity has facilitated the investigation of their efficacy in clinical trials with autoimmune patients. In this review, we discuss the role of BTK in key signaling pathways involved in autoimmunity and provide an overview of the different inhibitors that are currently being investigated in clinical trials of systemic autoimmune diseases, including rheumatoid arthritis and systemic lupus erythematosus, as well as available results from completed trials.
Collapse
Affiliation(s)
- Stefan F H Neys
- Department of Pulmonary Medicine, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - Jasper Rip
- Department of Pulmonary Medicine, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
- Department of Immunology, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - Rudi W Hendriks
- Department of Pulmonary Medicine, Erasmus MC, University Medical Center, Rotterdam, The Netherlands.
| | - Odilia B J Corneth
- Department of Pulmonary Medicine, Erasmus MC, University Medical Center, Rotterdam, The Netherlands.
| |
Collapse
|
27
|
Iwata S, Tanaka Y. Therapeutic perspectives on the metabolism of lymphocytes in patients with rheumatoid arthritis and systemic lupus erythematosus. Expert Rev Clin Immunol 2021; 17:1121-1130. [PMID: 34351835 DOI: 10.1080/1744666x.2021.1964957] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
INTRODUCTION The activation of autoreactive T- and B-cells and production of autoantibodies by B cells are involved in the pathogenesis of autoimmune diseases such as systemic lupus erythematosus (SLE) and rheumatoid arthritis (RA). Recently, the concept of 'immunometabolism' has attracted significant attention. Immune cells produce large amounts of energy in the form of ATP and biosynthesize biological components such as nucleic acids and lipids via metabolic reprogramming to activate, differentiate, and exert their functions. AREAS COVERED While the mechanisms underlying the metabolism of CD4+ T cells in SLE have been extensively studied, the metabolic changes underlying B cell activation, differentiation, and function remain unclear. Drugs targeting mTOR and AMPK, such as sirolimus, rapamycin, and metformin, have shown some efficacy and tolerability in clinical trials on patients with SLE, but have not led to breakthroughs. In this review, we summarize the current knowledge on the immunometabolic mechanisms involved in SLE and RA and discuss the potential novel therapeutic drugs. EXPERT OPINION The intensity of activation of different immune cells and their metabolic kinetics vary in different autoimmune diseases; thus, understanding the disease- and cell-specific metabolic mechanisms may help in the development of clinically effective immunometabolism-targeting drugs.
Collapse
Affiliation(s)
- Shigeru Iwata
- The First Department of Internal Medicine, Assistant Professor, University of Occupational and Environmental Health, Japan, School of Medicine, Kitakyushu, Japan
| | - Yoshiya Tanaka
- The First Department of Internal Medicine, Professor and Chairman, Deputy Director, University of Occupational and Environmental Health, Japan, the University Hospital, School of Medicine, Kitakyushu, Japan
| |
Collapse
|
28
|
Chung SW. Vasculitis: From Target Molecules to Novel Therapeutic Approaches. Biomedicines 2021; 9:757. [PMID: 34209028 PMCID: PMC8301353 DOI: 10.3390/biomedicines9070757] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 06/17/2021] [Accepted: 06/25/2021] [Indexed: 12/18/2022] Open
Abstract
Systemic vasculitis is a group of diverse diseases characterized by immune-mediated inflammation of blood vessels. Current treatments for vasculitis, such as glucocorticoids and alkylating agents, are associated with significant side effects. In addition, the management of both small and large vessel vasculitis is challenging due to a lack of robust markers of disease activity. Recent research has advanced our understanding of the pathogenesis of both small and large vessel vasculitis, and this has led to the development of novel biologic therapies capable of targeting key cytokine and cellular effectors of the inflammatory cascade. It is anticipated that these novel treatments will lead to more effective and less toxic treatment regimens for patients with systemic vasculitis.
Collapse
Affiliation(s)
- Sang-Wan Chung
- Division of Rheumatology, Department of Internal Medicine, School of Medicine, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Korea
| |
Collapse
|
29
|
Ogwang R, Muhanguzi D, Mwikali K, Anguzu R, Kubofcik J, Nutman TB, Taylor M, Newton CR, Vincent A, Conroy AL, Marsh K, Idro R. Systemic and cerebrospinal fluid immune and complement activation in Ugandan children and adolescents with long-standing nodding syndrome: A case-control study. Epilepsia Open 2021; 6:297-309. [PMID: 34033255 PMCID: PMC8166803 DOI: 10.1002/epi4.12463] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 12/03/2020] [Accepted: 12/17/2020] [Indexed: 12/12/2022] Open
Abstract
OBJECTIVE Nodding syndrome is a poorly understood epileptic encephalopathy characterized by a unique seizure type-head nodding-and associated with Onchocerca volvulus infection. We hypothesized that altered immune activation in the cerebrospinal fluid (CSF) and plasma of children with nodding syndrome may yield insights into the pathophysiology and progression of this seizure disorder. METHOD We conducted a case-control study of 154 children (8 years or older) with long-standing nodding syndrome and 154 healthy age-matched community controls in 3 districts of northern Uganda affected by nodding syndrome. Control CSF samples were obtained from Ugandan children in remission from hematological malignancy during routine follow-up. Markers of immune activation and inflammation (cytokines and chemokines) and complement activation (C5a) were measured in plasma and CSF using ELISA or Multiplex Luminex assays. O volvulus infection was assessed by serology for anti-OV-16 IgG levels. RESULTS The mean (SD) age of the population was 15.1 (SD: 1.9) years, and the mean duration of nodding syndrome from diagnosis to enrollment was 8.3 (SD: 2.7) years. The majority with nodding syndrome had been exposed to O volvulus (147/154 (95.4%)) compared with community children (86/154 (55.8%)), with an OR of 17.04 (95% CI: 7.33, 45.58), P < .001. C5a was elevated in CSF of children with nodding syndrome compared to controls (P < .0001). The levels of other CSF markers tested were comparable between cases and controls after adjusting for multiple comparisons. Children with nodding syndrome had lower plasma levels of IL-10, APRIL, CCL5 (RANTES), CCL2, CXCL13, and MMP-9 compared with community controls (P < .05 for all; multiple comparisons). Plasma CRP was elevated in children with nodding syndrome compared to community children and correlated with disease severity. SIGNIFICANCE Nodding syndrome is associated with exposure to O. volvulus. Compared to controls, children with long-standing symptoms of nodding syndrome show evidence of complement activation in CSF and altered immune activation in plasma.
Collapse
Affiliation(s)
- Rodney Ogwang
- Makerere University College of Health Sciences, Kampala, Uganda.,Centre of Tropical Neuroscience (CTN), Kitgum Site, Uganda.,KEMRI-Wellcome Trust Research Programme, Centre for Geographic Medicine Coast, Kilifi, Kenya
| | - Dennis Muhanguzi
- College of Veterinary Medicine Animal Resources and Biosecurity, Makerere University, Kampala, Uganda
| | - Kioko Mwikali
- KEMRI-Wellcome Trust Research Programme, Centre for Geographic Medicine Coast, Kilifi, Kenya
| | - Ronald Anguzu
- Centre of Tropical Neuroscience (CTN), Kitgum Site, Uganda.,Division of Epidemiology, Institute of Health and Equity, Medical College of Wisconsin, Wisconsin, WI, USA
| | - Joe Kubofcik
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, Bethesda, MD, USA
| | - Thomas B Nutman
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, Bethesda, MD, USA
| | - Mark Taylor
- Liverpool School of Tropical Medicine, Liverpool, UK
| | - Charles R Newton
- KEMRI-Wellcome Trust Research Programme, Centre for Geographic Medicine Coast, Kilifi, Kenya.,Department of Psychiatry, University of Oxford, Oxford, UK
| | - Angela Vincent
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Andrea L Conroy
- Indiana University School of Medicine, Ryan White Center for Pediatric Infectious Disease & Global Health, Indianapolis, IN, USA
| | - Kevin Marsh
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Richard Idro
- Makerere University College of Health Sciences, Kampala, Uganda.,Centre of Tropical Neuroscience (CTN), Kitgum Site, Uganda.,Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| |
Collapse
|
30
|
Nguyen TG. The therapeutic implications of activated immune responses via the enigmatic immunoglobulin D. Int Rev Immunol 2021; 41:107-122. [PMID: 33410368 DOI: 10.1080/08830185.2020.1861265] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Immunoglobulin D (IgD) is an enigmatic antibody and the least appreciated member of the immunoglobulin (Ig) family. Since its discovery over half a century ago, the essence of its function in the immune system has been somewhat enigmatic and less well-defined than other antibody classes. Membrane-bound IgD (mIgD) is mostly recognized as B-cell receptor (BCR) while secreted IgD (sIgD) has been recently implicated in 'arming' basophils and mast cells in mucosal innate immunity. Activations of immune responses via mIgD-BCR or sIgD by specific antigens or anti-IgD antibody thereby produce a broad and complex mix of cellular, antibody and cytokine responses from both the innate and adaptive immune systems. Such broadly activated immune responses via IgD were initially deemed to potentiate and exacerbate the onset of autoimmune and allergic conditions. Paradoxically, treatments with anti-IgD antibody suppressed and ameliorated autoimmune conditions and allergic inflammations in mouse models without compromising the host's general immune defence, demonstrating a unique and novel therapeutic application for anti-IgD antibody treatment. Herein, this review endeavored to collate and summarize the evidence of the unique characteristics and features of activated immune responses via mIgD-BCR and sIgD that revealed an unappreciated immune-regulatory function of IgD in the immune system via an amplifying loop of anti-inflammatory Th2 and tolerogenic responses, and highlighted a novel therapeutic paradigm in harnessing these immune responses to treat human autoimmune and allergic conditions.
Collapse
|
31
|
Orefice V, Ceccarelli F, Barbati C, Lucchetti R, Olivieri G, Cipriano E, Natalucci F, Perricone C, Spinelli FR, Alessandri C, Valesini G, Conti F. Caffeine intake influences disease activity and clinical phenotype in systemic lupus erythematosus patients. Lupus 2020; 29:1377-1384. [PMID: 32703116 DOI: 10.1177/0961203320941920] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
OBJECTIVE Caffeine, one of the most widely consumed products in the world, seems to interact with multiple components of the immune system by acting as a non-specific phosphodiesterase inhibitor. In vitro dose-dependent treatment with caffeine down-regulates mRNA levels of key inflammation-related genes in peripheral blood mononuclear cells. So far, no robust data are available about the possible contribution of caffeine in systemic lupus erythematosus (SLE). The aim of this study was to evaluate the impact of caffeine consumption on SLE-related disease phenotype and activity, in terms of clinimetric assessment and cytokine serum levels. METHODS We performed a cross-sectional study, enrolling consecutive patients and reporting their clinical and laboratory data. Disease activity was assessed by SLE Disease Activity Index 2000 (SLEDAI-2K). Caffeine intake was evaluated by a 7-day food frequency questionnaire, including all the main sources of caffeine. As previously reported, patients were divided into four groups according to the daily caffeine intake: <29.1 mg/day (group 1), 29.2-153.7 mg/day (group 2), 153.8-376.5 mg/day (group 3) and >376.6 mg/day (group 4). At the end of questionnaire filling, blood samples were collected from each patient to assess cytokine levels. These were assessed by using a panel by Bio-Plex assays to measure the levels of IL-6, IL-10, IL-17, IL-27, IFNγ, IFNα and BLyS. RESULTS We enrolled 89 consecutive SLE patients. We observed a negative correlation between caffeine consumption and disease activity, measured with SLEDAI-2K. A significantly higher prevalence of lupus nephritis, neuropsychiatric involvement, haematological manifestations, hypocomplementaemia and anti-dsDNA positivity was observed in patients with a low intake of caffeine. Furthermore, patients with a low intake of caffeine were more frequently treated with glucocorticoids. Regarding cytokine analysis, a negative correlation between daily caffeine consumption and serum level of IFNγ was found (p = 0.03, r = -0.2); furthermore, patients with a high intake of caffeine showed lower serum levels of IFNα (p = 0.02), IL-17 (p = 0.01) and IL-6 (p = 0.003). CONCLUSIONS In this report we demonstrated the impact of caffeine on SLE disease activity status, as confirmed by the inverse correlation between its intake and both SLEDAI-2K values and cytokine levels. Moreover, patients with a low caffeine consumption seem to have a more severe disease phenotype.
Collapse
Affiliation(s)
- Valeria Orefice
- Lupus Clinic, Department of Clinical, Internal, Anesthesiological and Cardiovascular Sciences, 9311Sapienza University of Rome, Rome, Italy
| | - Fulvia Ceccarelli
- Lupus Clinic, Department of Clinical, Internal, Anesthesiological and Cardiovascular Sciences, 9311Sapienza University of Rome, Rome, Italy
| | - Cristiana Barbati
- Lupus Clinic, Department of Clinical, Internal, Anesthesiological and Cardiovascular Sciences, 9311Sapienza University of Rome, Rome, Italy
| | - Ramona Lucchetti
- Lupus Clinic, Department of Clinical, Internal, Anesthesiological and Cardiovascular Sciences, 9311Sapienza University of Rome, Rome, Italy
| | - Giulio Olivieri
- Lupus Clinic, Department of Clinical, Internal, Anesthesiological and Cardiovascular Sciences, 9311Sapienza University of Rome, Rome, Italy
| | - Enrica Cipriano
- Lupus Clinic, Department of Clinical, Internal, Anesthesiological and Cardiovascular Sciences, 9311Sapienza University of Rome, Rome, Italy
| | - Francesco Natalucci
- Lupus Clinic, Department of Clinical, Internal, Anesthesiological and Cardiovascular Sciences, 9311Sapienza University of Rome, Rome, Italy
| | - Carlo Perricone
- Department of Internal Medicine, University of Perugia, Perugia, Italy
| | - Francesca Romana Spinelli
- Lupus Clinic, Department of Clinical, Internal, Anesthesiological and Cardiovascular Sciences, 9311Sapienza University of Rome, Rome, Italy
| | - Cristiano Alessandri
- Lupus Clinic, Department of Clinical, Internal, Anesthesiological and Cardiovascular Sciences, 9311Sapienza University of Rome, Rome, Italy
| | - Guido Valesini
- Lupus Clinic, Department of Clinical, Internal, Anesthesiological and Cardiovascular Sciences, 9311Sapienza University of Rome, Rome, Italy
| | - Fabrizio Conti
- Lupus Clinic, Department of Clinical, Internal, Anesthesiological and Cardiovascular Sciences, 9311Sapienza University of Rome, Rome, Italy
| |
Collapse
|
32
|
Cellular immune dysregulation in the pathogenesis of immune thrombocytopenia. Blood Coagul Fibrinolysis 2020; 31:113-120. [PMID: 31977328 DOI: 10.1097/mbc.0000000000000891] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
: Immune thrombocytopenia (ITP) is an acquired autoimmune hemorrhagic disease characterized by immune-mediated increased platelet destruction and decreased platelet production, resulting from immune intolerance to autoantigen. The pathogenesis of ITP remains unclear, although dysfunction of T and B lymphocytes has been shown to be involved in the pathogenesis of ITP. More recently, it is found that dendritic cells, natural killer, and myeloid-derived suppressor cells also play an important role in ITP. Elucidating its pathogenesis is expected to provide novel channels for the targeted therapy of ITP. This article will review the role of different immune cells in ITP.
Collapse
|
33
|
Homeostasis and regulation of autoreactive B cells. Cell Mol Immunol 2020; 17:561-569. [PMID: 32382130 DOI: 10.1038/s41423-020-0445-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Accepted: 04/10/2020] [Indexed: 12/15/2022] Open
Abstract
In contrast to the previous belief that autoreactive B cells are eliminated from the normal repertoire of B cells, many autoreactive B cells actually escape clonal deletion and develop into mature B cells. These autoreactive B cells in healthy individuals perform some beneficial functions in the host and are homeostatically regulated by regulatory T and B cells or other mechanisms to prevent autoimmune diseases. Autoreactive B-1 cells constitutively produce polyreactive natural antibodies for tissue homeostasis. Recently, autoreactive follicular B cells were reported to participate actively in the germinal center reaction. Furthermore, the selection and usefulness of autoreactive marginal zone (MZ) B cells found in autoimmune diseases are not well understood, although the repertoire of MZ B-cell receptors (BCRs) is presumed to be biased to detect bacterial antigens. In this review, we discuss the autoreactive B-cell populations among all three major B-cell subsets and their regulation in immune responses and diseases.
Collapse
|
34
|
Degner KR, Wilson NA, Reese SR, Parajuli S, Aziz F, Garg N, Mohamed M, Singh T, Mandelbrot DA, Panzer SE, Redfield RR, Van Hyfte K, Zhong W, Hidalgo LG, Djamali A. Short-term Immunopathological Changes Associated with Pulse Steroids/IVIG/Rituximab Therapy in Late Kidney Allograft Antibody Mediated Rejection. KIDNEY360 2020; 1:389-398. [PMID: 34476406 PMCID: PMC8409258 DOI: 10.34067/kid.0001082019] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 03/11/2020] [Indexed: 01/07/2023]
Abstract
BACKGROUND B-cell depletion is a common treatment of antibody-mediated rejection (ABMR). We sought to determine the specific immunopathologic effects of this therapeutic approach in kidney transplantation. METHODS This was a prospective observational study of kidney transplant recipients diagnosed with late ABMR (>3 months after transplant). Patients received treatment with pulse steroids, IVIG, and rituximab. Donor specific HLA antibodies (DSA), kidney allograft pathology, renal function, immune cell phenotypes, and 47 circulating cytokines were assessed at baseline and at three months. RESULTS We enrolled 23 patients in this study between April 2015 and March 2019. The majority of patients were male (74%) and Caucasian (78%) with an average age of 45.6±13.8 years. ABMR was diagnosed at 6.8±5.9 years (4 months-25 years) post-transplant. Treatment was associated with a significant decline in circulating HLA class I DSA (P=0.003) and class II DSA (P=0.002) and peritubular capillaritis (ptc, P=0.04) compared to baseline. Serum creatinine, BUN, eGFR, and proteinuria (UPC) remained stable. Circulating B-cells were depleted to barely detectable levels (P≤0.001), whereas BAFF (P=0.001), APRIL (P<0.001), and IL-10 (P=0.02), levels increased significantly post-treatment. Notably, there was a significant rise in circulating CD4+ (P=0.02) and CD8+ T-cells (P=0.003). We also noted a significant correlation between circulating cytotoxic CD8+ T-cells and BAFF (P=0.05), regulatory T-cells and IL10 (P=0.002), and HLA class I DSA (P=0.005). CONCLUSIONS Short-term pulse steroids/IVIG/rituximab therapy was associated with inhibition of ABMR (DSA and ptc), stabilization of kidney function, and increased regulatory B-cell and T-cell survival cytokines. Additional studies are needed to understand the implications of B cell-depletion on the crosstalk between T-cells, B-cells, and humoral components that regulate ABMR.
Collapse
Affiliation(s)
- Kenna R. Degner
- Department of Surgery, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Nancy A. Wilson
- Department of Surgery, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Shannon R. Reese
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Sandesh Parajuli
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Fahad Aziz
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Neetika Garg
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Maha Mohamed
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Tripti Singh
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Didier A. Mandelbrot
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Sarah E. Panzer
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Robert R. Redfield
- Department of Surgery, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Kristin Van Hyfte
- The Office of Clinical Trials, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Weixiong Zhong
- Department of Pathology and Laboratory Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Luis G. Hidalgo
- Department of Surgery, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Arjang Djamali
- Department of Surgery, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| |
Collapse
|
35
|
Zhang CX, Wang HY, Yin L, Mao YY, Zhou W. Immunometabolism in the pathogenesis of systemic lupus erythematosus. J Transl Autoimmun 2020; 3:100046. [PMID: 32743527 PMCID: PMC7388408 DOI: 10.1016/j.jtauto.2020.100046] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 03/07/2020] [Accepted: 03/08/2020] [Indexed: 12/25/2022] Open
Abstract
Systemic lupus erythematosus (SLE) is a typical autoimmune disease characterized by chronic inflammation and pathogenic auto-antibodies. Apart from B cells, dysregulation of other immune cells also plays an essential role in the pathogenesis and development of the disease including CD4+T cells, dendritic cells, macrophages and neutrophils. Since metabolic programs control immune cell fate and function, they are critical checkpoints in an effective immune response and are involved in the etiology of autoimmune disease. In addition, mitochondria and oxidative stress are both involved in cellular metabolism and is also essential in immune response. In this review, apart from the disturbed immune system, we will discuss mitochondrial dysfunction, oxidative stress, abnormal metabolism (including glucose, lipid and amino acid metabolism) of immune cells as well as epigenetic control of metabolism reprogramming to elucidate the underlying pathogenic mechanisms of systemic lupus erythematosus. Mitochondria plays a vital role in cellular metabolism and is involved in immune response. There are alterations in glucose, lipid and amino acid metabolism of various immune cells in SLE patients. Epigenetic status is influenced by the presence of metabolic intermediates and certain autoimmunity-related genes are hypomethylated in CD4+T cells, CD19+ B cells as well as CD14+ monocytes of SLE.
Collapse
Affiliation(s)
- Chen-Xing Zhang
- Department of Nephrology, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, 200127, Shanghai, China
| | - Hui-Yu Wang
- Institute of Physiological Chemistry and Pathobiochemistry, University of Muenster, 48149, Muenster, Germany
| | - Lei Yin
- Department of Nephrology, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, 200127, Shanghai, China
| | - You-Ying Mao
- Department of Nephrology, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, 200127, Shanghai, China
| | - Wei Zhou
- Department of Nephrology, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, 200127, Shanghai, China
| |
Collapse
|
36
|
Positive and negative cooperativity of TNF and Interferon-γ in regulating synovial fibroblast function and B cell survival in fibroblast/B cell co-cultures. Sci Rep 2020; 10:780. [PMID: 31964950 PMCID: PMC6972945 DOI: 10.1038/s41598-020-57772-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Accepted: 01/07/2020] [Indexed: 12/21/2022] Open
Abstract
Synovial fibroblasts (SF) were reported to produce B cell activating factor (BAFF) in response to stimulation with interferon-γ (IFN-γ) or tumor necrosis factor (TNF). However, the influence of these pro-inflammatory cytokines on other receptors/ligands of the TNF superfamily or associated cytokine receptors in SF has not been investigated yet. Here we show the differential regulation of BAFF (CD257), Fn14 (CD266), TACI (CD267), BAFF-R (CD268), BCMA (CD269), CD40 ligand (CD40L, CD154), IFN-γR (CD119), Leptin receptor (ObR, CD295), VCAM-1 (CD106) and membrane TGF-β in isolated SF and the impact of IFN-γ/TNF co-incubation on proliferation, IL-6 and IL-8 production. In addition, the impact of differentially stimulated SF on B cell survival in co-cultures was assessed. Surface cytokines and cytokine receptors were detected by flow cytometry. Soluble cytokine receptors and cytokines were quantified by ELISA. Proliferation was assessed by cell titer blue. Murine B cell survival in fibroblast/ B cell co-cultures was determined by annexin V/propidium iodide staining and flow cytometry. IFN-γ together with TNF synergistically and significantly increased the cell surface levels of BAFF, Fn14, TACI, BAFF-R, BCMA, CD40L, ObR and IFN-γR in rheumatoid arthritis SF after 72 h incubation. Soluble BAFF was only induced by IFN-γ and inhibited by TNF. Addition of TWEAK had no influence on proliferation or IL-8 production but decreased TNF-induced IL-6 production, whereas APRIL, BAFF and leptin did not modulate TNF or TNF/IFN-γ-induced proliferation or cytokine production. Proliferation was increased by TNF and further enhanced by the addition of IFN-γ. In co-culture experiments, SF stimulated with TNF/IFN but not TNF or IFN-γ alone increased shedding of VCAM-1 and expression of membrane TGFβ, which was associated with reduced survival of murine B cells. IFN-γ and TNF regulate the expression of TNF family member cytokines and associated receptors. Ligation of IFN-γR and Fn14 under pro-inflammatory conditions modulated IL-6/IL-8 production and proliferation. In B cell/SF co-cultures, the combination of TNF/IFN reduced B cell survival possibly via enhanced VCAM-1 shedding and/or increased TGF-β production. IFN-γ is necessary for the observed effects on B cell survival and SF cytokine production and emphasizes its anti-inflammatory role in rheumatoid arthritis.
Collapse
|
37
|
Ma K, Du W, Wang X, Yuan S, Cai X, Liu D, Li J, Lu L. Multiple Functions of B Cells in the Pathogenesis of Systemic Lupus Erythematosus. Int J Mol Sci 2019; 20:E6021. [PMID: 31795353 PMCID: PMC6929160 DOI: 10.3390/ijms20236021] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Revised: 11/25/2019] [Accepted: 11/28/2019] [Indexed: 12/15/2022] Open
Abstract
Systemic lupus erythematosus (SLE) is an autoimmune disease characterized by excessive autoantibody production and multi-organ involvement. Although the etiology of SLE still remains unclear, recent studies have characterized several pathogenic B cell subsets and regulatory B cell subsets involved in the pathogenesis of SLE. Among pathogenic B cell subsets, age-associated B cells (ABCs) are a newly identified subset of autoreactive B cells with T-bet-dependent transcriptional programs and unique functional features in SLE. Accumulation of T-bet+ CD11c+ ABCs has been observed in SLE patients and lupus mouse models. In addition, innate-like B cells with the autoreactive B cell receptor (BCR) expression and long-lived plasma cells with persistent autoantibody production contribute to the development of SLE. Moreover, several regulatory B cell subsets with immune suppressive functions have been identified, while the impaired inhibitory effects of regulatory B cells have been indicated in SLE. Thus, further elucidation on the functional features of B cell subsets will provide new insights in understanding lupus pathogenesis and lead to novel therapeutic interventions in the treatment of SLE.
Collapse
Affiliation(s)
- Kongyang Ma
- Department of Rheumatology and Immunology, Second Clinical Medical College of Jinan University, Shenzhen People’s Hospital, Shenzhen 518000, China; (K.M.); (D.L.)
- Department of Pathology and Shenzhen Institute of Research and Innovation, The University of Hong Kong, Hong Kong 999077, China; (W.D.); (X.W.)
| | - Wenhan Du
- Department of Pathology and Shenzhen Institute of Research and Innovation, The University of Hong Kong, Hong Kong 999077, China; (W.D.); (X.W.)
| | - Xiaohui Wang
- Department of Pathology and Shenzhen Institute of Research and Innovation, The University of Hong Kong, Hong Kong 999077, China; (W.D.); (X.W.)
| | - Shiwen Yuan
- Department of Rheumatology, Guangzhou First People’s Hospital, School of Medicine, South China University of Technology, Guangzhou, 510000, China; (S.Y.); (X.C.)
| | - Xiaoyan Cai
- Department of Rheumatology, Guangzhou First People’s Hospital, School of Medicine, South China University of Technology, Guangzhou, 510000, China; (S.Y.); (X.C.)
| | - Dongzhou Liu
- Department of Rheumatology and Immunology, Second Clinical Medical College of Jinan University, Shenzhen People’s Hospital, Shenzhen 518000, China; (K.M.); (D.L.)
| | - Jingyi Li
- Department of Rheumatology and Immunology, Southwest Hospital, The First Hospital Affiliated to The Army Medical University, Chongqing 400038, China
| | - Liwei Lu
- Department of Pathology and Shenzhen Institute of Research and Innovation, The University of Hong Kong, Hong Kong 999077, China; (W.D.); (X.W.)
| |
Collapse
|
38
|
Hobeika L, Ng L, Lee IJ. Moving Forward With Biologics in Lupus Nephritis. Adv Chronic Kidney Dis 2019; 26:338-350. [PMID: 31733718 DOI: 10.1053/j.ackd.2019.08.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2019] [Revised: 07/17/2019] [Accepted: 08/09/2019] [Indexed: 12/27/2022]
Abstract
The majority of patients with systemic lupus erythematosus develop lupus nephritis (LN) which significantly contributes to increased risks of hospitalizations, ESRD, and death. Unfortunately, treatments for LN have not changed over the past 15 years. Despite continued efforts to elucidate the pathogenesis of LN, no new drugs have yet replaced the standard-of-care regimens of cyclophosphamide or mycophenolate mofetil plus high-dose corticosteroids. The significant limitations of standard-of-care are low complete response rates, risk of flares, and ongoing inflammation in the kidney leading to progressive renal dysfunction. Repeat and prolonged treatments are often needed to control disease, leading to a high level of severe side effects. The development of targeted drugs with better efficacy and safety are desperately needed. The rationale for targeting key immunologic pathways in LN continues to be strongly supported by basic and translational research and has generated the hope and excitement of testing these therapies in human LN. This review provides an overview of biologics studied to date in clinical trials of LN, discusses the potential reasons for their failure, and addresses the challenges moving forward.
Collapse
|
39
|
Tedesco M, Gallieni M, Pellegata F, Cozzolino M, Alberici F. Update on ANCA-associated vasculitis: from biomarkers to therapy. J Nephrol 2019; 32:871-882. [DOI: 10.1007/s40620-019-00628-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Accepted: 07/02/2019] [Indexed: 12/17/2022]
|
40
|
Kato R, Sumitomo S, Tsuchida Y, Tsuchiya H, Nakachi S, Sakurai K, Hanata N, Nagafuchi Y, Kubo K, Tateishi S, Kanda H, Okamura T, Yamamoto K, Fujio K. CD4 +CD25 +LAG3 + T Cells With a Feature of Th17 Cells Associated With Systemic Lupus Erythematosus Disease Activity. Front Immunol 2019; 10:1619. [PMID: 31354747 PMCID: PMC6640175 DOI: 10.3389/fimmu.2019.01619] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2019] [Accepted: 06/28/2019] [Indexed: 12/14/2022] Open
Abstract
Systemic lupus erythematosus (SLE) is an autoimmune disease that involves multiple immune cell subsets. We analyzed immune cell subsets in human peripheral blood mononuclear cells (PBMC) in order to identify the cells that are significantly associated with SLE disease activity and treatment. The frequencies of various subsets of CD4+ T cells, B cells, monocytes and NK cells in PBMC were assessed in 30 healthy controls (HC), 30 rheumatoid arthritis (RA) patients and 26 SLE patients using flow cytometry. The correlations between subset frequencies in SLE and clinical traits including Systemic Lupus Erythematosus Disease Activity Index (SLEDAI) were examined. Changes in subset frequencies after the treatment in SLE patients were investigated. We focused on CD25+LAG3+ T cells and investigated their characteristics, including cytokine secretion, mRNA expression and suppression capacity. We assessed correlations between CD25+LAG3+ T cells and SLEDAI by Spearman's rank correlation coefficient. CD25+LAG3+ T cells were significantly increased in SLE whereas there were few in RA and HC groups. CD25+LAG3+ T cell frequencies were significantly correlated with SLEDAI and were increased in patients with a high SLEDAI score (> 10). CD25+LAG3+ T cells produced both IL-17 and FOXP3, expressed mRNA of both FOXP3 and RORC and lacked suppressive capacity. CD25+LAG3+ T cells were associated with disease activity of SLE. CD25+LAG3+ T cells had features of both CD25+FOXP3+ regulatory T cells (CD25+ Treg) and Th17. CD25+LAG3+ T cells could be associated with the inflammatory pathophysiology of SLE.
Collapse
Affiliation(s)
- Rika Kato
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Shuji Sumitomo
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yumi Tsuchida
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Haruka Tsuchiya
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Shinichiro Nakachi
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Keiichi Sakurai
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Norio Hanata
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yasuo Nagafuchi
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Kanae Kubo
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Shoko Tateishi
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.,Department of Immunotherapy Management, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Hiroko Kanda
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.,Department of Immunotherapy Management, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Tomohisa Okamura
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.,Max Planck-The University of Tokyo Center for Integrative Inflammology, The University of Tokyo, Tokyo, Japan
| | - Kazuhiko Yamamoto
- Center for Integrative Medical Sciences, The Institute of Physical and Chemical Research, Yokohama, Japan
| | - Keishi Fujio
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
41
|
Farrah TE, Basu N, Dweck M, Calcagno C, Fayad ZA, Dhaun N. Advances in Therapies and Imaging for Systemic Vasculitis. Arterioscler Thromb Vasc Biol 2019; 39:1520-1541. [PMID: 31189432 DOI: 10.1161/atvbaha.118.310957] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Vasculitis is a systemic disease characterized by immune-mediated injury of blood vessels. Current treatments for vasculitis, such as glucocorticoids and alkylating agents, are associated with significant side effects. Furthermore, the management of both small and large vessel vasculitis is challenging because of a lack of robust markers of disease activity. Recent research has advanced our understanding of the pathogenesis of both small and large vessel vasculitis, and this has led to the development of novel biologic therapies capable of targeting key cytokine and cellular effectors of the inflammatory cascade. In parallel, a diverse range of imaging modalities with the potential to monitor vessel inflammation are emerging. Continued expansion of combined structural and molecular imaging using positron emission tomography with computed tomography or magnetic resonance imaging may soon provide reliable longitudinal tracking of vascular inflammation. In addition, the emergence of radiotracers able to assess macrophage activation and immune checkpoint activity represents an exciting new frontier in imaging vascular inflammation. In the near future, these advances will allow more precise imaging of disease activity enabling clinicians to offer more targeted and individualized patient management.
Collapse
Affiliation(s)
- Tariq E Farrah
- From the University/British Heart Foundation Centre of Research Excellence, Centre of Cardiovascular Science, The Queen's Medical Research Institute, University of Edinburgh, Scotland (T.E.F., M.D., N.D.)
| | - Neil Basu
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Scotland (N.B.)
| | - Marc Dweck
- From the University/British Heart Foundation Centre of Research Excellence, Centre of Cardiovascular Science, The Queen's Medical Research Institute, University of Edinburgh, Scotland (T.E.F., M.D., N.D.)
| | - Claudia Calcagno
- Translational and Molecular Imaging Institute, Icahn School of Medicine at Mount Sinai, New York (C.C., Z.A.F.)
| | - Zahi A Fayad
- Translational and Molecular Imaging Institute, Icahn School of Medicine at Mount Sinai, New York (C.C., Z.A.F.)
| | - Neeraj Dhaun
- From the University/British Heart Foundation Centre of Research Excellence, Centre of Cardiovascular Science, The Queen's Medical Research Institute, University of Edinburgh, Scotland (T.E.F., M.D., N.D.)
| |
Collapse
|
42
|
Shu JL, Zhang XZ, Han L, Zhang F, Wu YJ, Tang XY, Wang C, Tai Y, Wang QT, Chen JY, Chang Y, Wu HX, Zhang LL, Wei W. Paeoniflorin-6'-O-benzene sulfonate alleviates collagen-induced arthritis in mice by downregulating BAFF-TRAF2-NF-κB signaling: comparison with biological agents. Acta Pharmacol Sin 2019; 40:801-813. [PMID: 30446734 DOI: 10.1038/s41401-018-0169-5] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Accepted: 09/06/2018] [Indexed: 11/09/2022]
Abstract
Paeoniflorin-6'-O-benzene sulfonate (CP-25) is a new ester derivative of paeoniflorin with improved lipid solubility and oral bioavailability, as well as better anti-inflammatory activity than its parent compound. In this study we explored whether CP-25 exerted therapeutic effects in collagen-induced arthritis (CIA) mice through regulating B-cell activating factor (BAFF)-BAFF receptors-mediated signaling pathways. CIA mice were given CP-25 or injected with biological agents rituximab or etanercept for 40 days. In CIA mice, we found that T cells and B cells exhibited abnormal proliferation; the percentages of CD19+ total B cells, CD19+CD27+-activated B cells, CD19+BAFFR+ and CD19+TACI+ cells were significantly increased in PBMCs and spleen lymphocytes. CP-25 suppressed the indicators of arthritis, alleviated histopathology, accompanied by reduced BAFF and BAFF receptors expressions, inhibited serum immunoglobulin levels, decreased the B-cell subsets percentages, and prevented the expressions of key molecules in NF-κB signaling. Furthermore, we showed that treatment with CP-25 reduced CD19+TRAF2+ cell expressions stimulated by BAFF and decreased TRAF2 overexpression in HEK293 cells in vitro. Thus, CP-25 restored the abnormal T cells proliferation and B-cell percentages to the normal levels, and normalized the elevated levels of IgA, IgG2a and key proteins in NF-κB signaling. In comparison, rituximab and etanercept displayed stronger anti-inflammatory activities than CP-25; they suppressed the elevated inflammatory indexes to below the normal levels in CIA mice. In summary, our results provide evidence that CP-25 alleviates CIA and regulates the functions of B cells through BAFF-TRAF2-NF-κB signaling. CP-25 would be a soft immunomodulatory drug with anti-inflammatory effect.
Collapse
|
43
|
Regola F, Piantoni S, Lowin T, Archetti S, Reggia R, Kumar R, Franceschini F, Airò P, Tincani A, Andreoli L, Pongratz G. Association Between Changes in BLyS Levels and the Composition of B and T Cell Compartments in Patients With Refractory Systemic Lupus Erythematosus Treated With Belimumab. Front Pharmacol 2019; 10:433. [PMID: 31105569 PMCID: PMC6494924 DOI: 10.3389/fphar.2019.00433] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Accepted: 04/04/2019] [Indexed: 12/31/2022] Open
Abstract
Introduction: Belimumab is a monoclonal antibody against soluble BLyS used for treatment of refractory Systemic Lupus Erythematosus (SLE). Although B cells are the main target of this therapy, a BLyS-dependent T cell activation pathway has also been demonstrated. The aim of the study is to analyze B and T cells phenotype modifications in a cohort of SLE patients treated with belimumab in correlation with serum BLyS levels. Materials and Methods: Fourteen SLE patients were enrolled in the study. Lymphocyte immunophenotyping by flow cytometry and determination of serum BLyS levels by high sensitivity ELISA were performed before the first infusion of belimumab, after 6 and 12 months of treatment. Sex and age-matched healthy controls were enrolled for the comparisons. Results: Baseline number of total B cells, especially switched memory B cells, were lower in SLE patients compared to control subjects. After 6 months of treatment, the total number of B cells, particularly, naive and transitional B cells, was significantly reduced in correlation with the reduction of BLyS levels. No significant association was found between baseline counts of B cells and reduction of SLEDAI-2K over time. In terms of response prediction, a significant association between SLEDAI-2K improvement at 12 months and the decrease of total number of B cells within the first 6 months of therapy was observed. Concerning the T cell compartment, the baseline percentage number of CD8+ effector memory was associated with SLEDAI-2K at baseline and with its improvement after 12 months of therapy. Furthermore, T cell lymphopenia and low number of circulating recent thymic emigrants were also observed compared to control subjects measured at baseline. Discussion: The effects of belimumab on B cell subpopulations could be explained by the direct blockage of soluble BLyS, while the mild effects on T cells might be explained indirectly by the reduction of disease activity by means of therapy. B cell immunophenotyping during belimumab might be useful for monitoring the response to treatment.
Collapse
Affiliation(s)
- Francesca Regola
- Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy.,Rheumatology and Clinical Immunology Unit, ASST-Spedali Civili of Brescia, Brescia, Italy
| | - Silvia Piantoni
- Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy.,Rheumatology and Clinical Immunology Unit, ASST-Spedali Civili of Brescia, Brescia, Italy
| | - Torsten Lowin
- Department of Rheumatology and Hiller Research Centre for Rheumatology, University Hospital Düsseldorf, Düsseldorf, Germany
| | - Silvia Archetti
- Rheumatology and Clinical Immunology Unit, ASST-Spedali Civili of Brescia, Brescia, Italy
| | - Rossella Reggia
- Rheumatology and Clinical Immunology Unit, ASST-Spedali Civili of Brescia, Brescia, Italy
| | - Rajesh Kumar
- Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Franco Franceschini
- Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy.,Rheumatology and Clinical Immunology Unit, ASST-Spedali Civili of Brescia, Brescia, Italy
| | - Paolo Airò
- Rheumatology and Clinical Immunology Unit, ASST-Spedali Civili of Brescia, Brescia, Italy
| | - Angela Tincani
- Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy.,Rheumatology and Clinical Immunology Unit, ASST-Spedali Civili of Brescia, Brescia, Italy
| | - Laura Andreoli
- Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy.,Rheumatology and Clinical Immunology Unit, ASST-Spedali Civili of Brescia, Brescia, Italy
| | - Georg Pongratz
- Department of Rheumatology and Hiller Research Centre for Rheumatology, University Hospital Düsseldorf, Düsseldorf, Germany
| |
Collapse
|
44
|
Hossny E, El-Ghoneimy D, Soliman DA, Ashour A. Diagnostic value of serum high-mobility group box-1 in pediatric systemic lupus erythematosus. Int J Rheum Dis 2019; 22:1402-1409. [PMID: 30938057 DOI: 10.1111/1756-185x.13556] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Revised: 12/22/2018] [Accepted: 02/25/2019] [Indexed: 12/20/2022]
Abstract
BACKGROUND High-mobility group box-1 (HMGB1) acts as a damage-associated molecular pattern or as an alarmin and it stimulates inflammatory and immunological activities. AIM We sought to investigate serum HMGB1 protein expression in patients with pediatric systemic lupus erythematosus (pSLE) in relation to the disease characteristics and activity. PATIENTS AND METHODS This is a controlled cross-sectional study which comprised 50 children and adolescents with Systemic lupus erythematosus (SLE) and 50 age- and sex-matched healthy subjects who served as a control group. Study measurements included clinical assessment, laboratory workup for SLE (complete blood count, erythrocyte sedimentation rate, serum creatinine, creatinine clearance and 24-hour urinary protein, C3 and anti-double-stranded DNA, lupus anticoagulant and anticardiolipin antibodies) and measurement of serum HMGB1 by enzyme-linked immunosorbent assay in patients and controls. RESULTS Serum HMGB1 expression was significantly higher in the pSLE patients than the control group (P < 0.001). Patients with lupus nephritis (LN) had significantly higher serum HMGB1 as compared to those with normal kidneys (P < 0.04). Serum HMGB1 in LN patients correlated positively to the SLE Disease Activity Index (P < 0.0001), and 24 hours urinary proteins and negatively to creatinine clearance (P < 0.001). At a cut-off point of ≥40 µg/L, serum HMGB1 showed good diagnostic value for pSLE with sensitivity and specificity of 98% and 95%, respectively. CONCLUSION Serum HMGB1 seems to be a reliable biomarker for diagnosis of pSLE and monitoring disease status, especially in LN. HMBG1 might prove to be a potential therapeutic target in LN.
Collapse
Affiliation(s)
- Elham Hossny
- Pediatric Allergy and Immunology Unit, Children's Hospital, Ain Shams University, Cairo, Egypt
| | - Dalia El-Ghoneimy
- Pediatric Allergy and Immunology Unit, Children's Hospital, Ain Shams University, Cairo, Egypt
| | - Dina A Soliman
- Department of Clinical Pathology, Ain Shams University, Cairo, Egypt
| | - Ayman Ashour
- Ministry of Public Health Hospitals, Cairo, Egypt
| |
Collapse
|
45
|
Hu S, Wang R, Zhang M, Liu K, Tao J, Tai Y, Zhou W, Wang Q, Wei W. BAFF promotes T cell activation through the BAFF-BAFF-R-PI3K-Akt signaling pathway. Biomed Pharmacother 2019; 114:108796. [PMID: 30921706 DOI: 10.1016/j.biopha.2019.108796] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Revised: 03/02/2019] [Accepted: 03/17/2019] [Indexed: 01/30/2023] Open
Abstract
B-cell activating factor from the tumor necrosis factor family (BAFF) has revealed its critical role in B cell proliferation and survival, as well as the pathogenesis of T-cell mediated autoimmune disease. However, the effect and molecular mechanisms of BAFF on T cell physiological function have not been fully elucidated. In this study it was seen that BAFF can promote the vitality of purified T cells, increase the proportion of CD3+CD4+, CD4+CD25+, CD4+CD154+, and CD4+CD69+ subgroups and reduce the proportion of CD4+CD62L+ subgroups. Negating BAFF activity with Atacicept (TACI-Fc) reverses vitality and activation of T cells. Furthermore, immunofluorescence detection revealed that BAFF promotes the expression of BAFF receptor (BAFF-R) and transmembrane activator and CAML interactor (TACI) in T cells. Flow cytometry displayed that BAFF/BAFF-R activates the PI3K-Akt signaling pathway while the application of PI3K inhibitor (wortmannin) illuminated that BAFF induces T cell vitality and activation through the PI3K-Akt signaling pathway. We conclude that BAFF is involved in not only the physiology of B cells, but also that of T cells. BAFF affects physiological T-cell activation through BAFF-R-mediated activation of the PI3K-Akt signaling pathway which mirrors one of the pathological mechanisms of T cell-mediated autoimmune diseases.
Collapse
Affiliation(s)
- Shanshan Hu
- Institute of Clinical Pharmacology, Anhui Medical University, Hefei, China; Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Hefei, China; Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei, China
| | - Rui Wang
- Institute of Clinical Pharmacology, Anhui Medical University, Hefei, China; Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Hefei, China; Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei, China
| | - Mei Zhang
- Institute of Clinical Pharmacology, Anhui Medical University, Hefei, China; Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Hefei, China; Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei, China
| | - Kangkang Liu
- Institute of Clinical Pharmacology, Anhui Medical University, Hefei, China; Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Hefei, China; Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei, China
| | - Juan Tao
- Institute of Clinical Pharmacology, Anhui Medical University, Hefei, China; Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Hefei, China; Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei, China
| | - Yu Tai
- Institute of Clinical Pharmacology, Anhui Medical University, Hefei, China; Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Hefei, China; Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei, China
| | - Weijie Zhou
- Institute of Clinical Pharmacology, Anhui Medical University, Hefei, China; Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Hefei, China; Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei, China
| | - Qingtong Wang
- Institute of Clinical Pharmacology, Anhui Medical University, Hefei, China; Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Hefei, China; Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei, China.
| | - Wei Wei
- Institute of Clinical Pharmacology, Anhui Medical University, Hefei, China; Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Hefei, China; Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei, China.
| |
Collapse
|
46
|
Postactivated B cells in systemic lupus erythematosus: update on translational aspects and therapeutic considerations. Curr Opin Rheumatol 2019; 31:175-184. [DOI: 10.1097/bor.0000000000000576] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
47
|
Nayar S, Campos J, Smith CG, Iannizzotto V, Gardner DH, Colafrancesco S, Pipi E, Kollert F, Hunter KJ, Brewer C, Buckley CD, Bowman SJ, Priori R, Valesini G, Juarez M, Fahy WA, Fisher BA, Payne A, Allen RA, Barone F. Phosphatidylinositol 3-kinase delta pathway: a novel therapeutic target for Sjögren's syndrome. Ann Rheum Dis 2019; 78:249-260. [PMID: 30472652 PMCID: PMC6352416 DOI: 10.1136/annrheumdis-2017-212619] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Revised: 09/28/2018] [Accepted: 10/18/2018] [Indexed: 01/08/2023]
Abstract
BACKGROUND The phosphatidylinositol 3-kinase delta isoform (PI3Kδ) belongs to an intracellular lipid kinase family that regulate lymphocyte metabolism, survival, proliferation, apoptosis and migration and has been successfully targeted in B-cell malignancies. Primary Sjögren's syndrome (pSS) is a chronic immune-mediated inflammatory disease characterised by exocrine gland lymphocytic infiltration and B-cell hyperactivation which results in systemic manifestations, autoantibody production and loss of glandular function. Given the central role of B cells in pSS pathogenesis, we investigated PI3Kδ pathway activation in pSS and the functional consequences of blocking PI3Kδ in a murine model of focal sialoadenitis that mimics some features of pSS. METHODS AND RESULTS Target validation assays showed significant expression of phosphorylated ribosomal protein S6 (pS6), a downstream mediator of the phosphatidylinositol 3-kinase delta (PI3Kδ) pathway, within pSS salivary glands. pS6 distribution was found to co-localise with T/B cell markers within pSS aggregates and the CD138+ plasma cells infiltrating the glands. In vivo blockade of PI3Kδ activity with seletalisib, a PI3Kδ-selective inhibitor, in a murine model of focal sialoadenitis decreased accumulation of lymphocytes and plasma cells within the glands of treated mice in the prophylactic and therapeutic regimes. Additionally, production of lymphoid chemokines and cytokines associated with ectopic lymphoneogenesis and, remarkably, saliva flow and autoantibody production, were significantly affected by treatment with seletalisib. CONCLUSION These data demonstrate activation of PI3Kδ pathway within the glands of patients with pSS and its contribution to disease pathogenesis in a model of disease, supporting the exploration of the therapeutic potential of PI3Kδ pathway inhibition in this condition.
Collapse
Affiliation(s)
- Saba Nayar
- Centre for Translational Inflammation Research, Institute of Inflammation and Ageing, College of Medical & Dental Sciences, University of Birmingham, Research Laboratories, Queen Elizabeth Hospital, Birmingham, UK
| | - Joana Campos
- Centre for Translational Inflammation Research, Institute of Inflammation and Ageing, College of Medical & Dental Sciences, University of Birmingham, Research Laboratories, Queen Elizabeth Hospital, Birmingham, UK
| | - Charlotte G Smith
- Centre for Translational Inflammation Research, Institute of Inflammation and Ageing, College of Medical & Dental Sciences, University of Birmingham, Research Laboratories, Queen Elizabeth Hospital, Birmingham, UK
| | - Valentina Iannizzotto
- Centre for Translational Inflammation Research, Institute of Inflammation and Ageing, College of Medical & Dental Sciences, University of Birmingham, Research Laboratories, Queen Elizabeth Hospital, Birmingham, UK
| | - David H Gardner
- Centre for Translational Inflammation Research, Institute of Inflammation and Ageing, College of Medical & Dental Sciences, University of Birmingham, Research Laboratories, Queen Elizabeth Hospital, Birmingham, UK
| | - Serena Colafrancesco
- Centre for Translational Inflammation Research, Institute of Inflammation and Ageing, College of Medical & Dental Sciences, University of Birmingham, Research Laboratories, Queen Elizabeth Hospital, Birmingham, UK
- Dipartimento di Medicina Interna e Specialita' Mediche, Sapienza, University of Rome, Rome, Italy
| | - Elena Pipi
- Centre for Translational Inflammation Research, Institute of Inflammation and Ageing, College of Medical & Dental Sciences, University of Birmingham, Research Laboratories, Queen Elizabeth Hospital, Birmingham, UK
| | - Florian Kollert
- Centre for Translational Inflammation Research, Institute of Inflammation and Ageing, College of Medical & Dental Sciences, University of Birmingham, Research Laboratories, Queen Elizabeth Hospital, Birmingham, UK
- Rheumatology Department, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
- Department of Rheumatology, Immunology and Allergology, University Hospital, University of Bern, Bern, Switzerland
| | - Kelly J Hunter
- Immunology, HBRC, University of Birmingham, Birmingham, UK
| | | | - Christopher Dominic Buckley
- NIHR Birmingham Biomedical Research Centre, University Hospitals Birmingham NHS Foundation Trust and University of Birmingham, Sandwell and West Birmingham NHS Foundation Trust & Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
| | - Simon J Bowman
- Centre for Translational Inflammation Research, Institute of Inflammation and Ageing, College of Medical & Dental Sciences, University of Birmingham, Research Laboratories, Queen Elizabeth Hospital, Birmingham, UK
| | - Roberta Priori
- Dipartimento di Medicina Interna e Specialita' Mediche, Sapienza, University of Rome, Rome, Italy
| | - Guido Valesini
- Dipartimento di Medicina Interna e Specialita' Mediche, Sapienza, University of Rome, Rome, Italy
| | | | | | - Benjamin A Fisher
- NIHR Birmingham Biomedical Research Centre, University Hospitals Birmingham NHS Foundation Trust and University of Birmingham, Sandwell and West Birmingham NHS Foundation Trust & Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
| | | | | | - Francesca Barone
- Centre for Translational Inflammation Research, Institute of Inflammation and Ageing, College of Medical & Dental Sciences, University of Birmingham, Research Laboratories, Queen Elizabeth Hospital, Birmingham, UK
- Rheumatology Department, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| |
Collapse
|
48
|
Park JS, Kim SM, Hwang SH, Choi SY, Kwon JY, Kwok SK, Cho ML, Park SH. Combinatory treatment using tacrolimus and a STAT3 inhibitor regulate Treg cells and plasma cells. Int J Immunopathol Pharmacol 2018; 32:2058738418778724. [PMID: 29873267 PMCID: PMC5992791 DOI: 10.1177/2058738418778724] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Systemic lupus erythematosus (SLE; lupus) is a prototypical autoimmune disease
characterized by circulating autoantibodies to nuclear antigens and immune
complex deposition, resulting in damage to target organs. To investigate the
effects of tacrolimus (TAC) on effector T cells and B cells, we examined its
involvement in the development of effector T cells, germinal center (GC) B
cells, and plasma cells in an in vitro system using wild-type (WT) and
lupus-prone mice. The population of T helper (Th) 1, Th2, and Th17 cells
interleukin (IL)-17-producing T (Th17) cells and the production of interferon-γ
and interleukin-17A IL-17A were suppressed by TAC. TAC also reduced the
population of regulatory T (Treg) cells; however, a combination treatment with
the signal transducer and activator of transcription 3 (STAT3) inhibitor STA-21
promoted the population of Treg cells. TAC also suppressed the populations of GC
B cells and plasma cells synergistically with STA-21. These findings suggest
that the application of TAC with a STAT3 signal inhibitor may provide benefits
in SLE treatment.
Collapse
Affiliation(s)
- Jin-Sil Park
- 1 The Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Sung-Min Kim
- 1 The Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Sun-Hee Hwang
- 1 The Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Si-Young Choi
- 1 The Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Ji Ye Kwon
- 1 The Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Seung-Ki Kwok
- 1 The Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, South Korea.,2 Division of Rheumatology, Department of Internal Medicine, School of Medicine, The Catholic University of Korea, Seoul St. Mary's Hospital, Seoul, South Korea
| | - Mi-La Cho
- 1 The Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Sung-Hwan Park
- 1 The Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, South Korea.,2 Division of Rheumatology, Department of Internal Medicine, School of Medicine, The Catholic University of Korea, Seoul St. Mary's Hospital, Seoul, South Korea
| |
Collapse
|
49
|
Zhang SH, Zhao Y, Xie QB, Jiang Y, Wu YK, Yan B. Aberrant activation of the type I interferon system may contribute to the pathogenesis of anti-melanoma differentiation-associated gene 5 dermatomyositis. Br J Dermatol 2018; 180:1090-1098. [PMID: 29947075 DOI: 10.1111/bjd.16917] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/04/2018] [Indexed: 02/05/2023]
Abstract
BACKGROUND Anti-melanoma differentiation-associated gene 5 (MDA5) dermatomyositis (DM) is a distinctive subtype of DM that carries a significant risk of interstitial lung disease (ILD). The mechanisms remain elusive. OBJECTIVES To explore the role of the type I interferon (IFN) system in the pathogenesis of anti-MDA5 DM. METHODS Twenty patients with anti-MDA5 DM were studied and compared with patients with anti-aminoacyl-tRNA synthetase (ARS) DM (n = 10) and autoantibody-negative patients with DM (n = 20). The levels of inflammatory cytokines, B-cell-activating factor (BAFF) and Krebs von den Lungen (KL)-6 in blood were tested by enzyme-linked immunosorbent assay and multiplex assays. Expressions of transcripts for IFN-associated sensors and type I IFN-inducible genes in peripheral blood mononuclear cells (PBMCs) were detected by real-time polymerase chain reaction. Expressions of the signal transducer and activator of transcription (STAT)1, interferon-stimulated gene (ISG)15 and MxA proteins in skin lesions were analysed by immunohistochemistry. RESULTS Plasma IFN-α levels were significantly increased in patients with anti-MDA5 DM. PBMCs from patients with anti-MDA5 DM showed significant upregulation of the TLR3, TLR7, IFIH1 and DDX58 genes, as well as serial IFN-inducible genes. Skin biopsies from patients with anti-MDA5 DM were characterized by strong expression of the STAT1, ISG15 and MxA proteins. In the patients with anti-MDA5 DM and ILD with high IFN-α production, there was a positive quantitative correlation between IFN-α and BAFF (rs = 0·63, P = 0·044). In addition, the higher levels of BAFF paralleled the higher concentrations of KL-6 (rs = 0·86, P = 0·0012). CONCLUSIONS Our data confirm the aberrant activation of the type I IFN system in anti-MDA5 DM. Overproduction of IFN-α linked with BAFF may be implicated in the development of ILD.
Collapse
Affiliation(s)
- S H Zhang
- Department of Rheumatology, West China Hospital, Sichuan University, No. 37 Guoxue Lane, Chengdu, 610041, China
| | - Y Zhao
- Department of Rheumatology, West China Hospital, Sichuan University, No. 37 Guoxue Lane, Chengdu, 610041, China
| | - Q B Xie
- Department of Rheumatology, West China Hospital, Sichuan University, No. 37 Guoxue Lane, Chengdu, 610041, China
| | - Y Jiang
- Department of Pathology, West China Hospital, Sichuan University, No. 37 Guoxue Lane, Chengdu, 610041, China
| | - Y K Wu
- Department of Laboratory Medicine, West China Hospital, Sichuan University, No. 37 Guoxue Lane, Chengdu, 610041, China
| | - B Yan
- Department of Rheumatology, West China Hospital, Sichuan University, No. 37 Guoxue Lane, Chengdu, 610041, China
| |
Collapse
|
50
|
Huang W, Quach TD, Dascalu C, Liu Z, Leung T, Byrne-Steele M, Pan W, Yang Q, Han J, Lesser M, Rothstein TL, Furie R, Mackay M, Aranow C, Davidson A. Belimumab promotes negative selection of activated autoreactive B cells in systemic lupus erythematosus patients. JCI Insight 2018; 3:122525. [PMID: 30185675 DOI: 10.1172/jci.insight.122525] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Accepted: 07/24/2018] [Indexed: 12/16/2022] Open
Abstract
Belimumab has therapeutic benefit in active systemic lupus erythematosus (SLE), especially in patients with high-titer anti-dsDNA antibodies. We asked whether the profound B cell loss in belimumab-treated SLE patients is accompanied by shifts in the immunoglobulin repertoire. We enrolled 15 patients who had been continuously treated with belimumab for more than 7 years, 17 matched controls, and 5 patients who were studied before and after drug initiation. VH genes of sort-purified mature B cells and plasmablasts were subjected to next-generation sequencing. We found that B cell-activating factor (BAFF) regulates the transitional B cell checkpoint, with conservation of transitional 1 (T1) cells and approximately 90% loss of T3 and naive B cells after chronic belimumab treatment. Class-switched memory B cells, B1 B cells, and plasmablasts were also substantially depleted. Next-generation sequencing revealed no redistribution of VH, DH, or JH family usage and no effect of belimumab on representation of the autoreactive VH4-34 gene or CDR3 composition in unmutated IgM sequences, suggesting a minimal effect on selection of the naive B cell repertoire. Interestingly, a significantly greater loss of VH4-34 was observed among mutated IgM and plasmablast sequences in chronic belimumab-treated subjects than in controls, suggesting that belimumab promotes negative selection of activated autoreactive B cells.
Collapse
Affiliation(s)
- Weiqing Huang
- Center for Autoimmunity and Musculoskeletal and Hematologic Diseases, and
| | - Tam D Quach
- Center for Autoimmunity and Musculoskeletal and Hematologic Diseases, and
| | - Cosmin Dascalu
- Center for Autoimmunity and Musculoskeletal and Hematologic Diseases, and
| | - Zheng Liu
- Center for Autoimmunity and Musculoskeletal and Hematologic Diseases, and
| | - Tungming Leung
- Biostatistics Unit, Feinstein Institute for Medical Research, Manhasset, New York, New York, USA
| | | | - Wenjing Pan
- HudsonAlpha Institute for Biotechnology, Huntsville, Alabama, USA
| | - Qunying Yang
- HudsonAlpha Institute for Biotechnology, Huntsville, Alabama, USA
| | - Jian Han
- HudsonAlpha Institute for Biotechnology, Huntsville, Alabama, USA
| | - Martin Lesser
- Biostatistics Unit, Feinstein Institute for Medical Research, Manhasset, New York, New York, USA
| | - Thomas L Rothstein
- Western Michigan University Homer Stryker M.D. School of Medicine, Kalamazoo, Michigan, USA
| | - Richard Furie
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Northwell Health, Great Neck, New York, USA
| | - Meggan Mackay
- Center for Autoimmunity and Musculoskeletal and Hematologic Diseases, and.,Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Northwell Health, Great Neck, New York, USA
| | - Cynthia Aranow
- Center for Autoimmunity and Musculoskeletal and Hematologic Diseases, and.,Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Northwell Health, Great Neck, New York, USA
| | - Anne Davidson
- Center for Autoimmunity and Musculoskeletal and Hematologic Diseases, and.,Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Northwell Health, Great Neck, New York, USA
| |
Collapse
|