1
|
Le Maître M, Guerrier T, Sanges S, Chepy A, Collet A, Launay D. Beyond circulating B cells: Characteristics and role of tissue-infiltrating B cells in systemic sclerosis. Autoimmun Rev 2025; 24:103782. [PMID: 40010623 DOI: 10.1016/j.autrev.2025.103782] [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: 01/10/2025] [Revised: 02/21/2025] [Accepted: 02/22/2025] [Indexed: 02/28/2025]
Abstract
B cells play a key role in the pathophysiology of systemic sclerosis (SSc). While they are less characterized than their circulating counterparts, tissue-infiltrating B cells may have a more direct pathological role in tissues. In this review, we decipher the multiple evidence of B cells infiltration in the skin and lungs of SSc patients and animal models of SSc but also of other chronic fibrotic diseases with similar pathological mechanisms such as chronic graft versus host disease, idiopathic pulmonary fibrosis or morphea. We also recapitulate the current knowledge about mechanisms of B cells infiltration and their functions in tissues. Finally, we discuss B cell targeted therapies, and their specific impact on infiltrated B cells. Understanding the local consequences of infiltrating B cells is an important step for a better management of patients and the improvement of therapies in SSc.
Collapse
Affiliation(s)
- Mathilde Le Maître
- Univ. Lille, Inserm, CHU Lille, U1286 - INFINITE - Institute for Translational Research in Inflammation, Lille, France.
| | - Thomas Guerrier
- Univ. Lille, Inserm, CHU Lille, U1286 - INFINITE - Institute for Translational Research in Inflammation, Lille, France
| | - Sébastien Sanges
- Univ. Lille, Inserm, CHU Lille, U1286 - INFINITE - Institute for Translational Research in Inflammation, Lille, France; CHU Lille, Département de Médecine Interne et Immunologie Clinique, F-59000 Lille, France; Centre National de Référence Maladies Auto-immunes Systémiques Rares du Nord, Nord-Ouest, Méditerranée et Guadeloupe (CeRAINOM), F-59000 Lille, France; Health Care Provider of the European Reference Network on Rare Connective Tissue and Musculoskeletal Diseases (ReCONNET), France
| | - Aurélien Chepy
- Univ. Lille, Inserm, CHU Lille, U1286 - INFINITE - Institute for Translational Research in Inflammation, Lille, France; CHU Lille, Département de Médecine Interne et Immunologie Clinique, F-59000 Lille, France; Centre National de Référence Maladies Auto-immunes Systémiques Rares du Nord, Nord-Ouest, Méditerranée et Guadeloupe (CeRAINOM), F-59000 Lille, France; Health Care Provider of the European Reference Network on Rare Connective Tissue and Musculoskeletal Diseases (ReCONNET), France
| | - Aurore Collet
- Univ. Lille, Inserm, CHU Lille, U1286 - INFINITE - Institute for Translational Research in Inflammation, Lille, France; CHU Lille, Institut d'Immunologie, Pôle de Biologie Pathologie Génétique, Lille, France
| | - David Launay
- Univ. Lille, Inserm, CHU Lille, U1286 - INFINITE - Institute for Translational Research in Inflammation, Lille, France; CHU Lille, Département de Médecine Interne et Immunologie Clinique, F-59000 Lille, France; Centre National de Référence Maladies Auto-immunes Systémiques Rares du Nord, Nord-Ouest, Méditerranée et Guadeloupe (CeRAINOM), F-59000 Lille, France; Health Care Provider of the European Reference Network on Rare Connective Tissue and Musculoskeletal Diseases (ReCONNET), France
| |
Collapse
|
2
|
Jimenez SA, Mendoza FA, Piera-Velazquez S. A review of recent studies on the pathogenesis of Systemic Sclerosis: focus on fibrosis pathways. Front Immunol 2025; 16:1551911. [PMID: 40308583 PMCID: PMC12040652 DOI: 10.3389/fimmu.2025.1551911] [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: 12/26/2024] [Accepted: 03/07/2025] [Indexed: 05/02/2025] Open
Abstract
Systemic Sclerosis (SSc) is a systemic autoimmune disease of unknown etiology characterized by the development of frequently progressive cutaneous and internal organ fibrosis accompanied by severe vascular alterations. The pathogenesis of SSc is highly complex and, despite extensive investigation, has not been fully elucidated. Numerous studies have suggested that unknown etiologic factors cause multiple alterations in genetically receptive hosts, leading to SSc development and progression. These events may be functionally and pathologically interconnected and include: 1) Structural and functional microvascular and endothelial cell abnormalities; 2) Severe oxidative stress and high reactive oxygen species (3); Frequently progressive cutaneous and visceral fibrosis; 4) Transdifferentiation of various cell types into activated myofibroblasts, the cells ultimately responsible for the fibrotic process; 5) Establishment of a chronic inflammatory process in various affected tissues; 6) Release of cytokines, chemokines, and growth factors from the inflammatory cells; 7) Abnormalities in humoral and cellular immunity with the production of specific autoantibodies; and 8) Epigenetic alterations including changes in multiple non-coding RNAs. These events manifest with different levels of intensity in the affected organs and display remarkable individual variability, resulting in a wide heterogeneity in the extent and severity of clinical manifestations. Here, we will review some of the recent studies related to SSc pathogenesis.
Collapse
Affiliation(s)
- Sergio A. Jimenez
- Jefferson Institute of Molecular Medicine and Scleroderma Center, Thomas Jefferson University, Philadelphia, PA, United States
| | - Fabian A. Mendoza
- Jefferson Institute of Molecular Medicine and Scleroderma Center, Thomas Jefferson University, Philadelphia, PA, United States
- Division of Rheumatology, Department of Medicine, Thomas Jefferson University, Philadelphia, PA, United States
| | - Sonsoles Piera-Velazquez
- Jefferson Institute of Molecular Medicine and Scleroderma Center, Thomas Jefferson University, Philadelphia, PA, United States
| |
Collapse
|
3
|
De Luca G, De Lorenzis E, Campochiaro C, Cacciapaglia F, Del Papa N, Zanatta E, Airò P, Lazzaroni MG, Giuggioli D, De Santis M, Alonzi G, Stano S, Binda M, Moccaldi B, Tonutti A, Cavalli S, Batani V, Natalello G, Iannone F, D’Agostino MA, Dagna L, Matucci-Cerinic M, Bosello SL. Rituximab retention rate in systemic sclerosis: a long term real-life multicentre study. Rheumatology (Oxford) 2025; 64:1284-1291. [PMID: 38745439 PMCID: PMC11879284 DOI: 10.1093/rheumatology/keae280] [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: 03/04/2024] [Revised: 04/24/2024] [Accepted: 05/09/2024] [Indexed: 05/16/2024] Open
Abstract
OBJECTIVES To report real-life data on rituximab retention rate as an indicator of safety and efficacy in a multicentric national cohort of systemic sclerosis patients. METHODS SSc patients treated with rituximab and followed for at least 36 months were included, clinically characterized and longitudinally monitored. A competing risk analysis with sub-hazard ratio (sHR) definition was performed to explore the clinical variables linked to specific cause of rituximab discontinuation. RESULTS One-hundred and fifty-two SSc-patients [mean age 47.3 (12.3) years; females 79.6%; diffuse disease 77.6%; anti-topoisomerase-I positivity 63.2%] were evaluated over a median (interquartile range) time of 3.3 (1.7-5.0) years. The primary indications for rituximab were interstitial lung disease (38.8%), worsening skin fibrosis (36.8%) and arthritis (13.8%); 138 patients (90.8%) received more than one rituximab course. The 5-year rituximab retention rate was 59.9% (44.6-64.7%). Clinical response was the most common reason for rituximab discontinuation [5.7; 95% CI: (3.7-8.4) per 100 patient-years] and was associated with a shorter disease duration (sHR 0.8; 95% CI: 0.7, 0.9), anti-topoisomerase-I negativity (sHR 0.4; 95% CI: 0.2, 0.9), previous digital ulcers (sHR 2.6; 95% CI: 1.1, 6.2) and no history of arthritis (sHR 0.3; 95% CI: 0.1, 0.8). Treatment failure was the second cause of rituximab discontinuation [3.7 (95% CI: 2.2, 6.0) per 100 patient-years] and was associated with anti-centromere antibody positivity (sHR 2.8; 95% CI: 1.1, 7.4) and anti-topoisomerase-I negativity (sHR 0.2; 95% CI: 0.1, 0.6). Adverse events (AEs) were the less common cause of discontinuation [3.1 (95% CI: 1.7, 5.2) per 100 patient-years], associated with limited cutaneous subset (sHR 3.4; 95% CI: 1.2, 9.7) and previous mycophenolate mofetil treatment (sHR 4.5; 95% CI: 1.2, 16.3). CONCLUSION Rituximab is a safe and effective treatment in SSc: clinical response emerged as the primary reason for rituximab discontinuation, and AEs had a limited impact on treatment persistence. The identification of specific disease features associated with a response to rituximab will be useful in the management of SSc-patients.
Collapse
Affiliation(s)
- Giacomo De Luca
- Scleroderma Unit, Unit of Immunology, Rheumatology, Allergy and Rare Diseases, IRCCS San Raffaele Hospital, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Enrico De Lorenzis
- Rheumatology Unit, Università Cattolica del Sacro Cuore, Fondazione Policlinico Universitario A. Gemelli—IRCCS, Rome, Italy
| | - Corrado Campochiaro
- Scleroderma Unit, Unit of Immunology, Rheumatology, Allergy and Rare Diseases, IRCCS San Raffaele Hospital, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | | | | | | | - Paolo Airò
- Scleroderma Unit, UOC Rheumatology and Clinical Immunology, ASST Spedali Civili, Brescia, Italy
| | - Maria Grazia Lazzaroni
- Scleroderma Unit, UOC Rheumatology and Clinical Immunology, ASST Spedali Civili, Brescia, Italy
| | - Dilia Giuggioli
- Rheumatology Unit, School of Medicine, University of Modena and Reggio Emilia, Modena
| | - Maria De Santis
- Rheumatology and Clinical Immunology, IRCCS Humanitas Research Hospital, Rozzano, Italy
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy
| | - Gabriella Alonzi
- Rheumatology Unit, Università Cattolica del Sacro Cuore, Fondazione Policlinico Universitario A. Gemelli—IRCCS, Rome, Italy
| | - Stefano Stano
- Rheumatology Unit, DiMePRe-J University of Bari, Bari, Italy
| | - Marco Binda
- Rheumatology Unit, Padova University Hospital, Padova, Italy
| | | | - Antonio Tonutti
- Rheumatology and Clinical Immunology, IRCCS Humanitas Research Hospital, Rozzano, Italy
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy
| | | | - Veronica Batani
- Scleroderma Unit, Unit of Immunology, Rheumatology, Allergy and Rare Diseases, IRCCS San Raffaele Hospital, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Gerlando Natalello
- Rheumatology Unit, Università Cattolica del Sacro Cuore, Fondazione Policlinico Universitario A. Gemelli—IRCCS, Rome, Italy
| | | | - Maria Antonietta D’Agostino
- Rheumatology Unit, Università Cattolica del Sacro Cuore, Fondazione Policlinico Universitario A. Gemelli—IRCCS, Rome, Italy
| | - Lorenzo Dagna
- Scleroderma Unit, Unit of Immunology, Rheumatology, Allergy and Rare Diseases, IRCCS San Raffaele Hospital, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Marco Matucci-Cerinic
- Scleroderma Unit, Unit of Immunology, Rheumatology, Allergy and Rare Diseases, IRCCS San Raffaele Hospital, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Silvia Laura Bosello
- Rheumatology Unit, Università Cattolica del Sacro Cuore, Fondazione Policlinico Universitario A. Gemelli—IRCCS, Rome, Italy
| |
Collapse
|
4
|
Diehl R, Hübner S, Lehr S, Rizzi M, Eyerich K, Nyström A. Skin Deep and Beyond: Unravelling B Cell Extracellular Matrix Interactions in Cutaneous Immunity and Disease. Exp Dermatol 2025; 34:e70068. [PMID: 40051023 PMCID: PMC11885703 DOI: 10.1111/exd.70068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 01/27/2025] [Accepted: 02/11/2025] [Indexed: 03/09/2025]
Abstract
The extracellular matrix (ECM) is a crucial component in multicellular organisms, serving as both a structural scaffold and active signalling units. While the role of the ECM, namely, to maintain homeostasis and steer adaptive immunity, is well described in secondary lymphoid organs, it is underappreciated in the skin-despite remarkable molecular similarity. Here, we examine how the specialised organisation of the ECM influences B cell development and function in both skin and secondary lymphoid organs with a special focus on ECM-integrin signalling. We discuss the presence and function of B cells in healthy and diseased skin, including their role in wound healing, autoimmune responses and inflammatory conditions. Additionally, we explore the formation of tertiary lymphoid structures in chronic skin diseases as a window into studying B cell-ECM interactions. By integrating fundamental immunology with skin biology, we aim to identify key knowledge gaps and explore potential clinical implications of B cell-ECM interactions in dermatology and beyond.
Collapse
Affiliation(s)
- Rebecca Diehl
- Department of DermatologyUniversity Medical Center Freiburg, Faculty of Medicine, University of FreiburgFreiburgGermany
| | - Stefanie Hübner
- Department of DermatologyUniversity Medical Center Freiburg, Faculty of Medicine, University of FreiburgFreiburgGermany
| | - Saskia Lehr
- Department of DermatologyUniversity Medical Center Freiburg, Faculty of Medicine, University of FreiburgFreiburgGermany
| | - Marta Rizzi
- Center of Chronic Immunodeficiency CCIUniversity Clinics and Medical FacultyFreiburgGermany
- CIBSS ‐ Centre for Integrative Biological Signalling StudiesUniversity of FreiburgFreiburgGermany
- Division of Clinical and Experimental Immunology, Institute of Immunology, Center for Pathophysiology, Infectiology and ImmunologyMedical University of ViennaViennaAustria
- Department of Rheumatology and Clinical ImmunologyUniversity Medical Center Freiburg, Faculty of Medicine, University of FreiburgFreiburgGermany
| | - Kilian Eyerich
- Department of DermatologyUniversity Medical Center Freiburg, Faculty of Medicine, University of FreiburgFreiburgGermany
| | - Alexander Nyström
- Department of DermatologyUniversity Medical Center Freiburg, Faculty of Medicine, University of FreiburgFreiburgGermany
| |
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
|
Lim YW, Quinn R, Bharti K, Ferrer M, Zarkoob H, Song MJ. Development of immunocompetent full thickness skin tissue constructs to model skin fibrosis for high-throughput drug screening. Biofabrication 2024; 17:015033. [PMID: 39622178 PMCID: PMC11638742 DOI: 10.1088/1758-5090/ad998c] [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: 05/20/2024] [Revised: 10/30/2024] [Accepted: 12/02/2024] [Indexed: 12/14/2024]
Abstract
The lack of the immune component in most of the engineered skin models remains a challenge to study the interplay between different immune and non-immune cell types of the skin. Immunocompetent humanin vitroskin models offer potential advantages in recapitulatingin vivolike behavior which can serve to accelerate translational research and therapeutics development for skin diseases. Here we describe a three-dimensional human full-thickness skin (FTS) equivalent incorporating polarized M1 and M2 macrophages from human peripheral CD14+monocytes. This macrophage-incorporated FTS model demonstrates discernible immune responses with physiologically relevant cytokine production and macrophage plasticity under homeostatic and lipopolysaccharide stimulation conditions. M2-incorporated FTS recapitulates skin fibrosis phenotypes with transforming growth factor-β1 treatment as reflected by significant collagen deposition and myofibroblast expression, demonstrating a M2 potentiation effect. In conclusion, we successfully biofabricated an immunocompetent FTS with functional macrophages in a high-throughput (HT) amenable format. This model is the first step towards a HT-assay platform to develop new therapeutics for skin diseases.
Collapse
Affiliation(s)
- Yi Wei Lim
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD 20850, United States of America
| | - Russell Quinn
- National Eye Institute, National Institutes of Health, Bethesda, MD 20814, United States of America
| | - Kapil Bharti
- National Eye Institute, National Institutes of Health, Bethesda, MD 20814, United States of America
| | - Marc Ferrer
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD 20850, United States of America
| | - Hoda Zarkoob
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD 20850, United States of America
| | - Min Jae Song
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD 20850, United States of America
| |
Collapse
|
7
|
Barile R, Rotondo C, Rella V, Trotta A, Cantatore FP, Corrado A. Fibrosis mechanisms in systemic sclerosis and new potential therapies. Postgrad Med J 2024:qgae169. [PMID: 39656890 DOI: 10.1093/postmj/qgae169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Revised: 08/20/2024] [Accepted: 11/19/2024] [Indexed: 12/17/2024]
Abstract
Systemic sclerosis is a rare rheumatic disease characterized by immune cell activation, tissue fibrosis, and endothelial dysfunction. Extracellular matrix synthesis disorder causes widespread fibrosis, primarily in skin and internal organs. Various factors such as TGFβ, VEGF, Galectin-3, and signaling pathways like Wnt/β-catenin are involved in pathophysiological processes. Treatment lacks a unified approach but combines diverse modalities tailored to disease subtype and progression. Current therapeutic strategies include biologics, JAK inhibitors, and IL-6 pathway modulators. Monoclonal antibodies and hypomethylating agents demonstrate potential in fibrosis inhibition. This review focuses on emerging therapeutic evidence regarding drugs targeting collagen, cytokines, and cell surface molecules in systemic sclerosis, aiming to provide insight into potential innovative treatment strategies.
Collapse
Affiliation(s)
- Raffaele Barile
- Rheumatology Unit, Department of Medical and Surgical Sciences, University of Foggia, Luigi Pinto 1, 71121, Foggia, Italy
| | - Cinzia Rotondo
- Rheumatology Unit, Department of Medical and Surgical Sciences, University of Foggia, Luigi Pinto 1, 71121, Foggia, Italy
| | - Valeria Rella
- Rheumatology Unit, Department of Medical and Surgical Sciences, University of Foggia, Luigi Pinto 1, 71121, Foggia, Italy
| | - Antonello Trotta
- Rheumatology Unit, Department of Medical and Surgical Sciences, University of Foggia, Luigi Pinto 1, 71121, Foggia, Italy
| | - Francesco Paolo Cantatore
- Rheumatology Unit, Department of Medical and Surgical Sciences, University of Foggia, Luigi Pinto 1, 71121, Foggia, Italy
| | - Addolorata Corrado
- Rheumatology Unit, Department of Medical and Surgical Sciences, University of Foggia, Luigi Pinto 1, 71121, Foggia, Italy
| |
Collapse
|
8
|
Wang X, Guo J, Dai Q. Mesenchymal stem cell-derived extracellular vesicles in systemic sclerosis: role and therapeutic directions. Front Cell Dev Biol 2024; 12:1492821. [PMID: 39483335 PMCID: PMC11524835 DOI: 10.3389/fcell.2024.1492821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Accepted: 10/07/2024] [Indexed: 11/03/2024] Open
Abstract
Systemic sclerosis (SSc) is a complex autoimmune disease with clinical symptoms of vascular damage, immune disorders, and fibrosis, presenting significant treatment challenges and limited therapeutic options. Mesenchymal stem cell-derived extracellular vesicles (MSC-EVs) have been demonstrated in numerous studies as more effective than MSCs in treating autoimmune diseases. Recent studies demonstrate that MSC-EVs can significantly ameliorate the symptoms of SSc and mitigate pathological changes such as vascular injury, immune dysregulation, and fibrosis. These findings underscore the promising therapeutic potential of MSC-EVs in the treatment of SSc. MSC-EVs promote angiogenesis, modulate immune dysfunction, and combat fibrosis. This article summarizes the therapeutic applications and possible mechanisms of MSC-EVs for SSc, thereby offering a novel therapeutic direction for the treatment of SSc.
Collapse
Affiliation(s)
- Xuan Wang
- Department of Rheumatology and Immunology, The Second Affiliated Hospital of Wannan Medical College, Wuhu, China
| | - Jiaying Guo
- Department of Geriatric Medicine, The Second Affiliated Hospital of Wannan Medical College, Wuhu, China
| | - Qiangfu Dai
- Department of Rheumatology and Immunology, The Second Affiliated Hospital of Wannan Medical College, Wuhu, China
| |
Collapse
|
9
|
Le Maître M, Guerrier T, Collet A, Derhourhi M, Meneboo JP, Toussaint B, Bonnefond A, Villenet C, Sebda S, Bongiovanni A, Tardivel M, Simon M, Jendoubi M, Daunou B, Largy A, Figeac M, Dubucquoi S, Launay D. Characteristics and impact of infiltration of B-cells from systemic sclerosis patients in a 3D healthy skin model. Front Immunol 2024; 15:1373464. [PMID: 39185406 PMCID: PMC11341436 DOI: 10.3389/fimmu.2024.1373464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Accepted: 07/23/2024] [Indexed: 08/27/2024] Open
Abstract
Introduction In systemic sclerosis (SSc), B-cells are activated and present in the skin and lung of patients where they can interact with fibroblasts. The precise impact and mechanisms of the interaction of B-cells and fibroblasts at the tissular level are poorly studied. Objective We investigated the impact and mechanisms of B-cell/fibroblast interactions in cocultures between B-cells from patients with SSc and 3-dimensional reconstituted healthy skin model including fibroblasts, keratinocytes and extracellular matrix. Methods The quantification and description of the B-cell infiltration in 3D cocultures were performed using cells imagery strategy and cytometry. The effect of coculture on the transcriptome of B-cells and fibroblasts was studied with bulk and single-cell RNA sequencing approaches. The mechanisms of this interaction were studied by blocking key cytokines like IL-6 and TNF. Results We showed a significant infiltration of B-cells in the 3D healthy skin model. The amount but not the depth of infiltration was higher with B-cells from SSc patients and with activated B-cells. B-cell infiltrates were mainly composed of naïve and memory cells, whose frequencies differed depending on B-cells origin and activation state: infiltrated B-cells from patients with SSc showed an activated profile and an overexpression of immunoglobulin genes compared to circulating B-cells before infiltration. Our study has shown for the first time that activated B-cells modified the transcriptomic profile of both healthy and SSc fibroblasts, toward a pro-inflammatory (TNF and IL-17 signaling) and interferon profile, with a key role of the TNF pathway. Conclusion B-cells and 3D skin cocultures allowed the modelization of B-cells infiltration in tissues observed in SSc, uncovering an influence of the underlying disease and the activation state of B-cells. We showed a pro-inflammatory effect on skin fibroblasts and pro-activation effect on infiltrating B-cells during coculture. This reinforces the role of B-cells in SSc and provide potential targets for future therapeutic approach in this disease.
Collapse
Affiliation(s)
- Mathilde Le Maître
- Univ. Lille, Inserm, CHU Lille, U1286 - INFINITE - Institute for Translational Research in Inflammation, Lille, France
| | - Thomas Guerrier
- Univ. Lille, Inserm, CHU Lille, U1286 - INFINITE - Institute for Translational Research in Inflammation, Lille, France
| | - Aurore Collet
- Univ. Lille, Inserm, CHU Lille, U1286 - INFINITE - Institute for Translational Research in Inflammation, Lille, France
- CHU Lille, Institut d’Immunologie, Pôle de Biologie Pathologie Génétique, Lille, France
| | - Mehdi Derhourhi
- Inserm UMR1283, CNRS UMR8199, European Genomic Institute for Diabetes (EGID), Institut Pasteur de Lille, Lille University Hospital, Lille, France
- Université de Lille, Lille, France
| | - Jean-Pascal Meneboo
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, US 41 - UAR 2014 - PLBS, Lille, France
| | - Bénédicte Toussaint
- Inserm UMR1283, CNRS UMR8199, European Genomic Institute for Diabetes (EGID), Institut Pasteur de Lille, Lille University Hospital, Lille, France
- Université de Lille, Lille, France
| | - Amélie Bonnefond
- Inserm UMR1283, CNRS UMR8199, European Genomic Institute for Diabetes (EGID), Institut Pasteur de Lille, Lille University Hospital, Lille, France
- Université de Lille, Lille, France
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom
| | - Céline Villenet
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, US 41 - UAR 2014 - PLBS, Lille, France
| | - Shéhérazade Sebda
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, US 41 - UAR 2014 - PLBS, Lille, France
| | - Antonino Bongiovanni
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, US 41 - UAR 2014 - PLBS, Lille, France
| | - Meryem Tardivel
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, US 41 - UAR 2014 - PLBS, Lille, France
| | - Myriam Simon
- Service de Médecine Interne et d’Immunologie Clinique, Centre de Référence Des Maladies Auto-Immunes et Systémiques Rares du Nord et Nord-Ouest de France (CeRAINO), CHU Lille, Lille, France
| | - Manel Jendoubi
- Univ. Lille, Inserm, CHU Lille, U1286 - INFINITE - Institute for Translational Research in Inflammation, Lille, France
| | - Blanche Daunou
- Univ. Lille, Inserm, CHU Lille, U1286 - INFINITE - Institute for Translational Research in Inflammation, Lille, France
| | - Alexis Largy
- Univ. Lille, Inserm, CHU Lille, U1286 - INFINITE - Institute for Translational Research in Inflammation, Lille, France
| | - Martin Figeac
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, US 41 - UAR 2014 - PLBS, Lille, France
| | - Sylvain Dubucquoi
- Univ. Lille, Inserm, CHU Lille, U1286 - INFINITE - Institute for Translational Research in Inflammation, Lille, France
- CHU Lille, Institut d’Immunologie, Pôle de Biologie Pathologie Génétique, Lille, France
| | - David Launay
- Univ. Lille, Inserm, CHU Lille, U1286 - INFINITE - Institute for Translational Research in Inflammation, Lille, France
- Service de Médecine Interne et d’Immunologie Clinique, Centre de Référence Des Maladies Auto-Immunes et Systémiques Rares du Nord et Nord-Ouest de France (CeRAINO), CHU Lille, Lille, France
| |
Collapse
|
10
|
Raniszewska A, Kwiecień I, Rutkowska E, Bednarek J, Sokołowski R, Miklusz P, Rzepecki P, Jahnz-Różyk K. Imbalance of B-Cell Subpopulations in the Microenvironment of Sarcoidosis or Lung Cancer. Cells 2024; 13:1274. [PMID: 39120304 PMCID: PMC11311476 DOI: 10.3390/cells13151274] [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: 07/02/2024] [Revised: 07/24/2024] [Accepted: 07/26/2024] [Indexed: 08/10/2024] Open
Abstract
Although the role of T lymphocytes in sarcoidosis (SA) and lung cancer (LC) is quite well reported, the occurrence of B cells in disease microenvironments may suggest their potential role as natural modifiers of the immune response. The aim of this study was to investigate the B-cell profile and lymphocyte-related hematological parameters between patients with SA, LC and healthy controls (HCs). The cells were assessed by flow cytometry and a hematological analyzer in peripheral blood (PB) and material from lymph nodes (LNs) obtained by the EBUS/TBNA method. We showed that in SA patients, there were higher percentages of naïve B and CD21low B cells and a lower percentage of class-switched memory B cells than LC patients in LNs. We observed a higher median proportion of non-switched memory and transitional B cells in the PB of SA patients than in LC patients. We noticed the lowest median proportion of class-switched memory B cells in the PB from SA patients. LC patients had a higher percentage of RE-LYMP and AS-LYMP than SA patients. Our study presented a different profile of B-cell subpopulations in SA and LC patients, distinguishing dominant subpopulations, and showed the relocation from distant compartments of the circulation to the disease microenvironment, thus emphasizing their role.
Collapse
Affiliation(s)
- Agata Raniszewska
- Laboratory of Hematology and Flow Cytometry, Department of Internal Medicine and Hematology, Military Institute of Medicine-National Research Institute, 04-141 Warsaw, Poland; (I.K.); (E.R.)
| | - Iwona Kwiecień
- Laboratory of Hematology and Flow Cytometry, Department of Internal Medicine and Hematology, Military Institute of Medicine-National Research Institute, 04-141 Warsaw, Poland; (I.K.); (E.R.)
| | - Elżbieta Rutkowska
- Laboratory of Hematology and Flow Cytometry, Department of Internal Medicine and Hematology, Military Institute of Medicine-National Research Institute, 04-141 Warsaw, Poland; (I.K.); (E.R.)
| | - Joanna Bednarek
- Department of Internal Medicine, Pulmonology, Allergology and Clinical Immunology, Military Institute of Medicine, 04-141 Warsaw, Poland; (J.B.); (R.S.); (P.M.); (K.J.-R.)
| | - Rafał Sokołowski
- Department of Internal Medicine, Pulmonology, Allergology and Clinical Immunology, Military Institute of Medicine, 04-141 Warsaw, Poland; (J.B.); (R.S.); (P.M.); (K.J.-R.)
| | - Piotr Miklusz
- Department of Internal Medicine, Pulmonology, Allergology and Clinical Immunology, Military Institute of Medicine, 04-141 Warsaw, Poland; (J.B.); (R.S.); (P.M.); (K.J.-R.)
| | - Piotr Rzepecki
- Department of Internal Medicine and Hematology, Military Institute of Medicine-National Research Institute, 04-141 Warsaw, Poland;
| | - Karina Jahnz-Różyk
- Department of Internal Medicine, Pulmonology, Allergology and Clinical Immunology, Military Institute of Medicine, 04-141 Warsaw, Poland; (J.B.); (R.S.); (P.M.); (K.J.-R.)
| |
Collapse
|
11
|
Bangert C, Alkon N, Chennareddy S, Arnoldner T, Levine JP, Pilz M, Medjimorec MA, Ruggiero J, Cohenour ER, Jonak C, Damsky W, Griss J, Brunner PM. Dupilumab-associated head and neck dermatitis shows a pronounced type 22 immune signature mediated by oligoclonally expanded T cells. Nat Commun 2024; 15:2839. [PMID: 38565563 PMCID: PMC10987549 DOI: 10.1038/s41467-024-46540-0] [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: 07/03/2023] [Accepted: 03/01/2024] [Indexed: 04/04/2024] Open
Abstract
Dupilumab, an IL4R-blocking antibody, has shown clinical efficacy for atopic dermatitis (AD) treatment. In addition to conjunctivitis/blepharitis, the de novo appearance of head/neck dermatitis is now recognized as a distinct side effect, occurring in up to 10% of patients. Histopathological features distinct from AD suggest a drug effect, but exact underlying mechanisms remain unknown. We profiled punch biopsies from dupilumab-associated head and neck dermatitis (DAHND) by using single-cell RNA sequencing and compared data with untreated AD and healthy control skin. We show that dupilumab treatment was accompanied by normalization of IL-4/IL-13 downstream activity markers such as CCL13, CCL17, CCL18 and CCL26. By contrast, we found strong increases in type 22-associated markers (IL22, AHR) especially in oligoclonally expanded T cells, accompanied by enhanced keratinocyte activation and IL-22 receptor upregulation. Taken together, we demonstrate that dupilumab effectively dampens conventional type 2 inflammation in DAHND lesions, with concomitant hyperactivation of IL22-associated responses.
Collapse
Affiliation(s)
- Christine Bangert
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Natalia Alkon
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | | | - Tamara Arnoldner
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Jasmine P Levine
- Icahn School of Medicine at Mount Sinai, New York City, NY, USA
- New York Medical College, Valhalla, NY, USA
| | - Magdalena Pilz
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Marco A Medjimorec
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - John Ruggiero
- Icahn School of Medicine at Mount Sinai, New York City, NY, USA
| | - Emry R Cohenour
- Icahn School of Medicine at Mount Sinai, New York City, NY, USA
| | - Constanze Jonak
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - William Damsky
- Department of Dermatology, Yale School of Medicine, New Haven, CT, USA
| | - Johannes Griss
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | | |
Collapse
|
12
|
Wu J, Zhang X, Lin S, Wei Q, Lin Z, Jin O, Gu J. Alterations in peripheral T- and B-cell subsets in patients with systemic sclerosis. Int J Rheum Dis 2024; 27:e15145. [PMID: 38661314 DOI: 10.1111/1756-185x.15145] [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: 03/03/2024] [Revised: 03/22/2024] [Accepted: 03/27/2024] [Indexed: 04/26/2024]
Abstract
OBJECTIVES To determine the alteration of peripheral T and B cell subsets in patients with systemic sclerosis (SSc) and to evaluate their correlation with the progression of SSc. METHODS We recruited 47 SSc patients and 45 healthy controls (HCs) in this study. Demographic and clinical data were then collected. Flow cytometry was used to detect the proportions of 44 different T and B cell subsets in circulating blood. RESULTS The proportion of total B cells (p = .043) decreased in SSc patients, together with similar frequencies of total T cells, CD4+ T cells, and CD8+ T cells in both groups. Several subsets of T and B cells differed significantly between these two groups. Follicular helper T cells-1 (Tfh1) (p < .001), helper T cells-1 (Th1) (p = .001), regulatory T cells (Treg) (p = .004), effector memory CD8+ T cells (p = .041), and cytotoxic T cells-17 (Tc17) (p = .01) were decreased in SSc patients. Follicular helper T cells-2 (Tfh2) (p = .001) and, helper T cells-2 (Th2) (p = .001) levels increased in the SSc group. Regulatory B cells (Breg) (p = .015) were lower in the SSc group, together with marginal zone (MZ) B cells (p < .001), memory B cells (p = .001), and non-switched B cells (p = .005). The modified Rodnan skin score (mRSS) correlated with helper T cells-17 (Th17) (r = -.410, p = .004), Tfh1 (r = -.321, p = .028), peripheral helper T cells (Tph) (r = -.364, p = .012) and plasma cells (r = -.312, p = .033). CONCLUSIONS The alterations in T and B cells implied immune dysfunction, which may play an essential role in systemic sclerosis.
Collapse
Affiliation(s)
- Jialing Wu
- Department of Rheumatology and Immunology, Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Xi Zhang
- Department of Rheumatology and Immunology, Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Shen Lin
- Department of Rheumatology and Immunology, Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Qiujing Wei
- Department of Rheumatology and Immunology, Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Zhiming Lin
- Department of Rheumatology and Immunology, Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Ou Jin
- Department of Rheumatology and Immunology, Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Jieruo Gu
- Department of Rheumatology and Immunology, Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
| |
Collapse
|
13
|
Lee EG, Oh JE. From neglect to spotlight: the underappreciated role of B cells in cutaneous inflammatory diseases. Front Immunol 2024; 15:1328785. [PMID: 38426103 PMCID: PMC10902158 DOI: 10.3389/fimmu.2024.1328785] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Accepted: 01/30/2024] [Indexed: 03/02/2024] Open
Abstract
The skin, covering our entire body as its largest organ, manifests enormous complexities and a profound interplay of systemic and local responses. In this heterogeneous domain, B cells were considered strangers. Yet, recent studies have highlighted their existence in the skin and their distinct role in modulating cutaneous immunity across various immune contexts. Accumulating evidence is progressively shedding light on the significance of B cells in maintaining skin health and in skin disorders. Herein, we integrate current insights on the systemic and local contributions of B cells in three prevalent inflammatory skin conditions: Pemphigus Vulgaris (PV), Systemic Lupus Erythematosus (SLE), and Atopic Dermatitis (AD), underscoring the previously underappreciated importance of B cells within skin immunity. Moreover, we address the potential adverse effects of current treatments used for skin diseases, emphasizing their unintentional consequences on B cells. These comprehensive approaches may pave the way for innovative therapeutic strategies that effectively address the intricate nature of skin disorders.
Collapse
Affiliation(s)
- Eun-Gang Lee
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Ji Eun Oh
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
- BioMedical Research Center, KAIST, Daejeon, Republic of Korea
| |
Collapse
|
14
|
Yoshifuji H, Yomono K, Yamano Y, Kondoh Y, Yasuoka H. Role of rituximab in the treatment of systemic sclerosis: A literature review. Mod Rheumatol 2023; 33:1068-1077. [PMID: 37053127 DOI: 10.1093/mr/road040] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 03/30/2023] [Accepted: 04/10/2023] [Indexed: 04/14/2023]
Abstract
This literature review aimed to evaluate the effectiveness of rituximab (RTX) in patients with systemic sclerosis (SSc). PubMed was searched for articles, published through 31 March 2022, on any controlled studies using RTX in the treatment of SSc. Of 85 identified articles, 9 were selected by title/abstract screening and full text examination. All nine articles reported outcomes of forced vital capacity (%FVC), and seven reported those of modified Rodnan skin scores (mRSS). The results showed that among the seven controlled studies evaluating skin lesions in patients with SSc, four showed a significant improvement of mRSS by RTX when compared with a control group, whereas three showed no significant effect. Among the nine controlled studies evaluating lung lesions, five showed a significant improvement of %FVC compared with a control group, whereas four showed no significant effect. In conclusion, RTX may be effective in the treatment of skin and lung lesions in patients with SSc. The profiles of SSc patients for whom RTX was indicated were unclear, although patients with diffuse cutaneous SSc and those positive for anti-topoisomerase I antibody were considered potential targets. Additional studies are needed to assess the long-term effectiveness of RTX in the treatment of patients with SSc.
Collapse
Affiliation(s)
- Hajime Yoshifuji
- Department of Rheumatology and Clinical Immunology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Keina Yomono
- Department of Allergy and Rheumatology, Nippon Medical School, Graduate School of Medicine, Tokyo, Japan
| | - Yasuhiko Yamano
- Department of Respiratory Medicine and Allergy, Tosei General Hospital, Aichi, Japan
| | - Yasuhiro Kondoh
- Department of Respiratory Medicine and Allergy, Tosei General Hospital, Aichi, Japan
| | - Hidekata Yasuoka
- Division of Rheumatology, Department of Internal Medicine, Fujita Health University School of Medicine, Aichi, Japan
| |
Collapse
|
15
|
Wang Q, Li CL, Wu L, Hu JY, Yu Q, Zhang SX, He PF. Distinct molecular subtypes of systemic sclerosis and gene signature with diagnostic capability. Front Immunol 2023; 14:1257802. [PMID: 37849750 PMCID: PMC10577296 DOI: 10.3389/fimmu.2023.1257802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Accepted: 09/19/2023] [Indexed: 10/19/2023] Open
Abstract
Background As Systemic Sclerosis (SSc) is a connective tissue ailment that impacts various bodily systems. The study aims to clarify the molecular subtypes of SSc, with the ultimate objective of establishing a diagnostic model that can inform clinical treatment decisions. Methods Five microarray datasets of SSc were retrieved from the GEO database. To eliminate batch effects, the combat algorithm was applied. Immune cell infiltration was evaluated using the xCell algorithm. The ConsensusClusterPlus algorithm was utilized to identify SSc subtypes. Limma was used to determine differential expression genes (DEGs). GSEA was used to determine pathway enrichment. A support vector machine (SVM), Random Forest(RF), Boruta and LASSO algorithm have been used to select the feature gene. Diagnostic models were developed using SVM, RF, and Logistic Regression (LR). A ROC curve was used to evaluate the performance of the model. The compound-gene relationship was obtained from the Comparative Toxicogenomics Database (CTD). Results The identification of three immune subtypes in SSc samples was based on the expression profiles of immune cells. The utilization of 19 key intersectional DEGs among subtypes facilitated the classification of SSc patients into three robust subtypes (gene_ClusterA-C). Gene_ClusterA exhibited significant enrichment of B cells, while gene_ClusterC showed significant enrichment of monocytes. Moderate activation of various immune cells was observed in gene_ClusterB. We identified 8 feature genes. The SVM model demonstrating superior diagnostic performance. Furthermore, correlation analysis revealed a robust association between the feature genes and immune cells. Eight pertinent compounds, namely methotrexate, resveratrol, paclitaxel, trichloroethylene, formaldehyde, silicon dioxide, benzene, and tetrachloroethylene, were identified from the CTD. Conclusion The present study has effectively devised an innovative molecular subtyping methodology for patients with SSc and a diagnostic model based on machine learning to aid in clinical treatment. The study has identified potential molecular targets for therapy, thereby offering novel perspectives for the treatment and investigation of SSc.
Collapse
Affiliation(s)
- Qi Wang
- School of Basic Medical Sciences, Shanxi Medical University, Taiyuan, China
- Shanxi Key Laboratory of Big Data for Clinical Decision Research, Taiyuan, China
| | - Chen-Long Li
- School of Basic Medical Sciences, Shanxi Medical University, Taiyuan, China
- Shanxi Key Laboratory of Big Data for Clinical Decision Research, Taiyuan, China
| | - Li Wu
- School of Basic Medical Sciences, Shanxi Medical University, Taiyuan, China
- Department of Anesthesiology , Shanxi Provincial People’s Hospital (Fifth Hospital) of Shanxi Medical University, Taiyuan, China
| | - Jing-Yi Hu
- School of Management, Shanxi Medical University, Taiyuan, China
| | - Qi Yu
- Shanxi Key Laboratory of Big Data for Clinical Decision Research, Taiyuan, China
- School of Management, Shanxi Medical University, Taiyuan, China
| | - Sheng-Xiao Zhang
- Department of Rheumatology, Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Pei-Feng He
- Shanxi Key Laboratory of Big Data for Clinical Decision Research, Taiyuan, China
- School of Management, Shanxi Medical University, Taiyuan, China
| |
Collapse
|
16
|
Steinmetz TD, Verstappen GM, Suurmond J, Kroese FGM. Targeting plasma cells in systemic autoimmune rheumatic diseases - Promises and pitfalls. Immunol Lett 2023; 260:44-57. [PMID: 37315847 DOI: 10.1016/j.imlet.2023.06.005] [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: 02/03/2023] [Revised: 05/12/2023] [Accepted: 06/10/2023] [Indexed: 06/16/2023]
Abstract
Plasma cells are the antibody secretors of the immune system. Continuous antibody secretion over years can provide long-term immune protection but could also be held responsible for long-lasting autoimmunity in case of self-reactive plasma cells. Systemic autoimmune rheumatic diseases (ARD) affect multiple organ systems and are associated with a plethora of different autoantibodies. Two prototypic systemic ARDs are systemic lupus erythematosus (SLE) and Sjögren's disease (SjD). Both diseases are characterized by B-cell hyperactivity and the production of autoantibodies against nuclear antigens. Analogues to other immune cells, different subsets of plasma cells have been described. Plasma cell subsets are often defined dependent on their current state of maturation, that also depend on the precursor B-cell subset from which they derived. But, a universal definition of plasma cell subsets is not available so far. Furthermore, the ability for long-term survival and effector functions may differ, potentially in a disease-specific manner. Characterization of plasma cell subsets and their specificity in individual patients can help to choose a suitable targeting approach for either a broad or more selective plasma cell depletion. Targeting plasma cells in systemic ARDs is currently challenging because of side effects or varying depletion efficacies in the tissue. Recent developments, however, like antigen-specific targeting and CAR-T-cell therapy might open up major benefits for patients beyond current treatment options.
Collapse
Affiliation(s)
- Tobit D Steinmetz
- University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.
| | - Gwenny M Verstappen
- University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Jolien Suurmond
- Department of Rheumatology, Leiden University Medical Center, Leiden, The Netherlands
| | - Frans G M Kroese
- University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| |
Collapse
|
17
|
Mitsui Y, Yamabe F, Hori S, Uetani M, Kobayashi H, Nagao K, Nakajima K. Molecular Mechanisms and Risk Factors Related to the Pathogenesis of Peyronie's Disease. Int J Mol Sci 2023; 24:10133. [PMID: 37373277 PMCID: PMC10299070 DOI: 10.3390/ijms241210133] [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: 04/29/2023] [Revised: 05/25/2023] [Accepted: 06/03/2023] [Indexed: 06/29/2023] Open
Abstract
Peyronie's disease (PD) is a benign condition caused by plaque formation on the tunica albuginea of the penis. It is associated with penile pain, curvature, and shortening, and contributes to erectile dysfunction, which worsens patient quality of life. In recent years, research into understanding of the detailed mechanisms and risk factors involved in the development of PD has been increasing. In this review, the pathological mechanisms and several closely related signaling pathways, including TGF-β, WNT/β-catenin, Hedgehog, YAP/TAZ, MAPK, ROCK, and PI3K/AKT, are described. Findings regarding cross-talk among these pathways are then discussed to elucidate the complicated cascade behind tunica albuginea fibrosis. Finally, various risk factors including the genes involved in the development of PD are presented and their association with the disease summarized. The purpose of this review is to provide a better understanding regarding the involvement of risk factors in the molecular mechanisms associated with PD pathogenesis, as well as to provide insight into disease prevention and novel therapeutic interventions.
Collapse
Affiliation(s)
- Yozo Mitsui
- Department of Urology, Toho University Faculty of Medicine, Tokyo 143-8540, Japan; (F.Y.); (S.H.); (M.U.); (H.K.); (K.N.); (K.N.)
| | | | | | | | | | | | | |
Collapse
|
18
|
Ayoglu B, Donato M, Furst DE, Crofford LJ, Goldmuntz E, Keyes-Elstein L, James J, Macwana S, Mayes MD, McSweeney P, Nash RA, Sullivan KM, Welch B, Pinckney A, Mao R, Chung L, Khatri P, Utz PJ. Characterising the autoantibody repertoire in systemic sclerosis following myeloablative haematopoietic stem cell transplantation. Ann Rheum Dis 2023; 82:670-680. [PMID: 36653124 PMCID: PMC10176357 DOI: 10.1136/ard-2021-221926] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Accepted: 12/30/2022] [Indexed: 01/20/2023]
Abstract
OBJECTIVES Results from the SCOT (Scleroderma: Cyclophosphamide Or Transplantation) clinical trial demonstrated significant benefits of haematopoietic stem cell transplant (HSCT) versus cyclophosphamide (CTX) in patients with systemic sclerosis. The objective of this study was to test the hypothesis that transplantation stabilises the autoantibody repertoire in patients with favourable clinical outcomes. METHODS We used a bead-based array containing 221 protein antigens to profile serum IgG autoantibodies in participants of the SCOT trial. RESULTS Comparison of autoantibody profiles at month 26 (n=23 HSCT; n=22 CTX) revealed antibodies against two viral antigens and six self-proteins (SSB/La, CX3CL1, glycyl-tRNA synthetase (EJ), parietal cell antigen, bactericidal permeability-increasing protein and epidermal growth factor receptor (EGFR)) that were significantly different between treatment groups. Linear mixed model analysis identified temporal increases in antibody levels for hepatitis B surface antigen, CCL3 and EGFR in HSCT-treated patients. Eight of 32 HSCT-treated participants and one of 31 CTX-treated participants had temporally varying serum antibody profiles for one or more of 14 antigens. Baseline autoantibody levels against 20 unique antigens, including 9 secreted proteins (interleukins, IL-18, IL-22, IL-23 and IL-27), interferon-α2A, stem cell factor, transforming growth factor-β, macrophage colony-stimulating factor and macrophage migration inhibitory factor were significantly higher in patients who survived event-free to month 54. CONCLUSIONS Our results suggest that HSCT favourably alters the autoantibody repertoire, which remains virtually unchanged in CTX-treated patients. Although antibodies recognising secreted proteins are generally thought to be pathogenic, our results suggest a subset could potentially modulate HSCT in scleroderma.
Collapse
Affiliation(s)
- Burcu Ayoglu
- Department of Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - Michele Donato
- Department of Medicine, Stanford University School of Medicine, Stanford, California, USA
- Stanford Institute for Immunity Transplantation and Infection, Stanford, California, USA
| | - Daniel E Furst
- Department of Medicine, University of California, Los Angeles, California, USA
| | - Leslie J Crofford
- Division of Rheumatology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Ellen Goldmuntz
- Division of Allergy, Immunology, and Transplantation, National Institute of Allergy and Infectious Diseases, Bethesda, Maryland, USA
| | | | - Judith James
- Arthritis & Clinical Immunology, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, USA
- Departments of Medicine and Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - Susan Macwana
- Arthritis & Clinical Immunology, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, USA
| | - Maureen D Mayes
- Department of Rheumatology, The University of Texas Health Science Center, Houston, Texas, USA
| | | | | | - Keith M Sullivan
- Department of Medicine, Duke University Medical Center, Durham, North Carolina, USA
| | - Beverly Welch
- Division of Allergy, Immunology, and Transplantation, National Institute of Allergy and Infectious Diseases, Bethesda, Maryland, USA
| | | | - Rong Mao
- Department of Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - Lorinda Chung
- Departments of Medicine & Dermatology, Stanford University, Stanford, California, USA
- Palo Alto VA Health Care System, Palo Alto, California, USA
| | - Purvesh Khatri
- Department of Medicine, Stanford University School of Medicine, Stanford, California, USA
- Stanford Institute for Immunity Transplantation and Infection, Stanford, California, USA
| | - Paul J Utz
- Department of Medicine, Stanford University School of Medicine, Stanford, California, USA
- Stanford Institute for Immunity Transplantation and Infection, Stanford, California, USA
| |
Collapse
|
19
|
Stenger S, Grasshoff H, Hundt JE, Lange T. Potential effects of shift work on skin autoimmune diseases. Front Immunol 2023; 13:1000951. [PMID: 36865523 PMCID: PMC9972893 DOI: 10.3389/fimmu.2022.1000951] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Accepted: 11/29/2022] [Indexed: 02/16/2023] Open
Abstract
Shift work is associated with systemic chronic inflammation, impaired host and tumor defense and dysregulated immune responses to harmless antigens such as allergens or auto-antigens. Thus, shift workers are at higher risk to develop a systemic autoimmune disease and circadian disruption with sleep impairment seem to be the key underlying mechanisms. Presumably, disturbances of the sleep-wake cycle also drive skin-specific autoimmune diseases, but epidemiological and experimental evidence so far is scarce. This review summarizes the effects of shift work, circadian misalignment, poor sleep, and the effect of potential hormonal mediators such as stress mediators or melatonin on skin barrier functions and on innate and adaptive skin immunity. Human studies as well as animal models were considered. We will also address advantages and potential pitfalls in animal models of shift work, and possible confounders that could drive skin autoimmune diseases in shift workers such as adverse lifestyle habits and psychosocial influences. Finally, we will outline feasible countermeasures that may reduce the risk of systemic and skin autoimmunity in shift workers, as well as treatment options and highlight outstanding questions that should be addressed in future studies.
Collapse
Affiliation(s)
- Sarah Stenger
- Lübeck Institute of Experimental Dermatology, University of Lübeck, Lübeck, Germany
| | - Hanna Grasshoff
- Department of Rheumatology and Clinical Immunology, University of Lübeck, Lübeck, Germany
| | - Jennifer Elisabeth Hundt
- Lübeck Institute of Experimental Dermatology, University of Lübeck, Lübeck, Germany
- Center for Research on Inflammation of the Skin, University of Lübeck, Lübeck, Germany
| | - Tanja Lange
- Department of Rheumatology and Clinical Immunology, University of Lübeck, Lübeck, Germany
- Center for Research on Inflammation of the Skin, University of Lübeck, Lübeck, Germany
- Center of Brain, Behavior and Metabolism (CBBM), University of Lübeck, Lübeck, Germany
| |
Collapse
|
20
|
Fang H, Xue K, Cao T, Li Q, Dang E, Liu Y, Zhang J, Qiao P, Chen J, Ma J, Shen S, Pang B, Bai Y, Qiao H, Shao S, Wang G. CXCL12/CXCR4 Axis Drives the Chemotaxis and Differentiation of B Cells in Bullous Pemphigoid. J Invest Dermatol 2023; 143:197-208.e6. [PMID: 36075452 DOI: 10.1016/j.jid.2022.08.041] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 07/20/2022] [Accepted: 08/01/2022] [Indexed: 01/27/2023]
Abstract
Bullous pemphigoid (BP) is an autoimmune bullous skin disease characterized by autoantibodies against the hemidesmosomal proteins in the skin and mucous membranes. The efficiency of B-cell‒targeting biologics in BP indicates the important role of B cells in its pathogenesis. However, abnormal B-cell migration and differentiation in BP require further elucidation. We showed that the number of antibody-secreting cells increased in the circulation and skin lesions of patients with BP and was correlated with disease severity. Bulk RNA sequencing of the peripheral B cells identified 171 upregulated and 408 downregulated genes in patients with BP compared with those in healthy controls, among which CXCR4 was significantly upregulated. Notably, CXCR4+ B cells were enriched in BP skin lesions and exhibited antibody-secreting cell characteristics. Correspondingly, an elevated level of CXCL12, the CXCR4 ligand, was detected in the blister fluid and serum of patients with BP, mediating the chemotaxis and accumulation of CXCR4+ B cells to BP skin lesions. Moreover, CXCL12 activated the transcription factor c-Myc, thus promoting B-cell differentiation into antibody-secreting cells and facilitating autoantibody production, which was blocked by CXCR4 inhibitor in vitro. Collectively, our study reveals that the CXCL12/CXCR4 axis plays a pathogenic role in modulating B-cell trafficking and differentiation, thus targeting CXCR4 represents a potential strategy for treating BP and other autoimmune diseases.
Collapse
Affiliation(s)
- Hui Fang
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Ke Xue
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Tianyu Cao
- Department of Dermatology, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Qingyang Li
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Erle Dang
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Yanghe Liu
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Jieyu Zhang
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Pei Qiao
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Jiaoling Chen
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Jingyi Ma
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Shengxian Shen
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Bingyu Pang
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Yaxing Bai
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Hongjiang Qiao
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Shuai Shao
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Gang Wang
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, China.
| |
Collapse
|
21
|
Zhang Y, Zhu M, Xie L, Zhang H, Deng T. Identification and validation of key immune-related genes with promising diagnostic and predictive value in systemic sclerosis. Life Sci 2022; 312:121238. [PMID: 36460097 DOI: 10.1016/j.lfs.2022.121238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 11/18/2022] [Accepted: 11/22/2022] [Indexed: 11/30/2022]
Abstract
AIMS To screen and confirm key immune-related genes (IRGs) with diagnostic and predictive value in systemic sclerosis (SSc) and provide potential therapeutic targets for patients with SSc. MATERIALS AND METHODS In Gene Expression Omnibus database (GEO), four datasets of gene expression profiling related to SSc were downloaded and used for the analysis in this study. After differential analysis of SSc cases and controls in GSE130955, the differentially expressed genes (DEGs) were overlapped with IRGs to obtain the immune-related differentially expressed genes (IR-DEGs) in SSc. In addition, functional annotation and pathway enrichment of IR-DEGs were conducted. The protein-protein interaction network (PPI) was constructed to identify key IR-DEGs. Using GSE58095, GSE181549 and GSE130953, the diagnostic and predictive abilities for the key IR-DEGs in SSc were validated. Finally, the screened key genes were confirmed in skin derived bleomycin (BLM)-induced SSc mice by Real-time PCR. KEY FINDINGS NGFR, TNFSF13B, FCER1G, GIMAP5, TYROBP and CSF1R may have important or very high diagnostic value for SSc. TYROBP and TNFSF13B had moderate and mild predictive value respectively in SSc patients after treatment. Real-time PCR assay further confirmed that the expressions of Ngfr, Tyrobp, Csf1r, Fcer1g and Gimap5 were significantly higher in skin of BLM-induced SSc mice than that in controls. SIGNIFICANCE The key IR-DEGs, including NGFR, TNFSF13B, TYROBP, CSF1R, FCER1G and GIMAP5, may become auxiliary diagnostic indicators and potential biomarkers for SSc. Moreover, TNFSF13B and TYROBP could have good prospects as predictive indicators in SSc patients that accepted cyclophosphamide or transplantation therapy.
Collapse
Affiliation(s)
- Yajie Zhang
- National Clinical Research Center for Metabolic Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China; Key Laboratory of Diabetes Immunology, Ministry of Education, Metabolic Syndrome Research Center, The Second Xiangya Hospital of Central South University, Changsha, China.
| | - Mingxin Zhu
- National Clinical Research Center for Metabolic Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China; Key Laboratory of Diabetes Immunology, Ministry of Education, Metabolic Syndrome Research Center, The Second Xiangya Hospital of Central South University, Changsha, China.
| | - Limin Xie
- National Clinical Research Center for Metabolic Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China; Key Laboratory of Diabetes Immunology, Ministry of Education, Metabolic Syndrome Research Center, The Second Xiangya Hospital of Central South University, Changsha, China.
| | - Haowei Zhang
- The First Affiliated Hospital, Department of Orthopedics, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Tuo Deng
- National Clinical Research Center for Metabolic Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China; Key Laboratory of Diabetes Immunology, Ministry of Education, Metabolic Syndrome Research Center, The Second Xiangya Hospital of Central South University, Changsha, China; Clinical Immunology Center, The Second Xiangya Hospital of Central South University, Changsha, China.
| |
Collapse
|
22
|
Kubik P, Jankau J, Rauso R, Galadari H, Protasoni M, Gruszczyński W, Grzanka D, Smolińska M, Antosik P, Piesiaków ML, Kodłubańska L, Zagajewska A, Łukasik B, Stabile G, Zerbinati N. HA PEGylated Filler in Association with an Infrared Energy Device for the Treatment of Facial Skin Aging: 150 Day Follow-Up Data Report. Pharmaceuticals (Basel) 2022; 15:1355. [PMID: 36355527 PMCID: PMC9697715 DOI: 10.3390/ph15111355] [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] [Received: 09/18/2022] [Revised: 10/13/2022] [Accepted: 10/26/2022] [Indexed: 08/27/2023] Open
Abstract
BACKGROUND The face is the area most exposed to the normal course of skin aging, both intrinsically and extrinsically. The aim of the study was to evaluate the cellular and clinical response of a therapeutic protocol aimed at countering facial skin aging. MATERIALS AND METHODS Twenty female patients with facial skin laxity and photodamage underwent combined therapy including mesotherapy using non-cross-linked hyaluronic acid with calcium hydroxyapatite and an infrared energy-based device treatment with subsequent implementation of PEG-cross-linked hyaluronic acid soft tissue fillers. To evaluate the benefits, patients underwent histological, immunological, and biomechanical evaluations before the treatment and at 21 and 150 days after the treatment. RESULTS The histological results at 21 days and 150 days after the procedure showed an increase in the number of fibroblasts and angiogenesis. As for the immunological aspect, it was shown that the treatment has an immunomodulating action, avoiding the activation of CD4 and CD8 cells. Biomechanical data showed that, at 150 days after treatment, the average changes in skin elasticity increased by 72% and the skin hydration increased by 49%. CONCLUSIONS A combination of an infrared energy-based device treatment with both non-cross-linked hyaluronic acid and novel PEG-cross-linked hyaluronic acid leads to numerous positive cutaneous changes after histological, immunological, and biomechanical evaluations.
Collapse
Affiliation(s)
- Paweł Kubik
- Centrum Medyczne dr Kubik, Skwer Kościuszki 15/17, 81-370 Gdynia, Poland
| | - Jerzy Jankau
- Department of Plastic Surgery, Medical University of Gdansk, 80-210 Gdańsk, Poland
| | - Raffaele Rauso
- Maxillofacial Surgery Unit, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy
| | - Hassan Galadari
- Department of Medicine, College of Medicine and Health Sciences UAE, University Al Ain, Al-Ain 15551, United Arab Emirates
| | - Marina Protasoni
- Department of Medicine and Surgery, University of Insubria, 21100 Varese, Italy
| | | | - Dariusz Grzanka
- Department of Clinical Patomorphology, Nicolaus Copericus University, 85-094 Bydgoszcz, Poland
| | - Marta Smolińska
- Department of Clinical Patomorphology, Nicolaus Copericus University, 85-094 Bydgoszcz, Poland
| | - Paulina Antosik
- Department of Clinical Patomorphology, Nicolaus Copericus University, 85-094 Bydgoszcz, Poland
| | | | - Lidia Kodłubańska
- Centrum Medyczne dr Kubik, Skwer Kościuszki 15/17, 81-370 Gdynia, Poland
| | - Anna Zagajewska
- Centrum Medyczne dr Kubik, Skwer Kościuszki 15/17, 81-370 Gdynia, Poland
| | | | - Giorgio Stabile
- Department of Clinical Dermartology, Università Vita-Salute San Raffaele, 20132 Milan, Italy
| | - Nicola Zerbinati
- Department of Medicine and Surgery, University of Insubria, 21100 Varese, Italy
| |
Collapse
|
23
|
Higashioka K, Migita R, Ota T, Uchino A, Niiro H. Successful Treatment of Systemic Sclerosis-related Pericarditis with Mycophenolate Mofetil and Low-dose Prednisolone. Intern Med 2022; 61:3125-3130. [PMID: 35283383 PMCID: PMC9646350 DOI: 10.2169/internalmedicine.8844-21] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
We herein report a case of systemic sclerosis (SSc)-related pericarditis successfully treated with mycophenolate mofetil (MMF) and low-dose prednisolone (PSL). The patient was a 72-year-old woman with anti-centromere antibody. Her clinical manifestations were Raynaud phenomenon, bilateral pleural effusion, pericardial effusion and skin tightness. Based on the findings of exudative pericardial effusion with the absence of pulmonary arterial hypertension from the results of the cardiac catheter and pericardiocentesis, she was diagnosed with SSc-related pericarditis and treated with PSL10 mg and MMF 1 g per day, leading to the complete resolution of pericarditis. These findings suggested that MMF and low-dose PSL were effective for SSc-related pericarditis.
Collapse
Affiliation(s)
| | - Rioko Migita
- Department of Rheumatology, Aso Iizuka Hospital, Japan
| | - Toshiyuki Ota
- Department of Rheumatology, Aso Iizuka Hospital, Japan
| | - Ayumi Uchino
- Department of Rheumatology, Aso Iizuka Hospital, Japan
| | - Hiroaki Niiro
- Department of Medical Education, Kyushu University Graduate School of Medical Sciences, Japan
| |
Collapse
|
24
|
Abstract
Barrier tissues are the primary site of infection for pathogens likely to cause future pandemics. Tissue-resident lymphocytes can rapidly detect pathogens upon infection of barrier tissues and are critical in preventing viral spread. However, most vaccines fail to induce tissue-resident lymphocytes and are instead reliant on circulating antibodies to mediate protective immunity. Circulating antibody titers wane over time following vaccination leaving individuals susceptible to breakthrough infections by variant viral strains that evade antibody neutralization. Memory B cells were recently found to establish tissue residence following infection of barrier tissues. Here, we summarize emerging evidence for the importance of tissue-resident memory B cells in the establishment of protective immunity against viral and bacterial challenge. We also discuss the role of tissue-resident memory B cells in regulating the progression of non-infectious diseases. Finally, we examine new approaches to develop vaccines capable of eliciting barrier immunity.
Collapse
Affiliation(s)
- Changfeng Chen
- Division of Allergy and Immunology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, United States
| | - Brian J Laidlaw
- Division of Allergy and Immunology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, United States.
| |
Collapse
|
25
|
The Role of T Cells in Systemic Sclerosis: An Update. IMMUNO 2022. [DOI: 10.3390/immuno2030034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Systemic sclerosis (SSc) is a chronic disease characterized by microvasculopathy, autoantibodies (autoAbs), and fibrosis. The pathogenesis of the disease is incompletely understood. Microvasculopathy and autoAbs appear very early in the disease process. AutoAbs, such as those directed against DNA topoisomerase I (Topo I), are disease specific and associated with disease manifestations, and indicate activation of the adaptive immune system. B cells are involved in fibrosis in SSc. T cells are also involved in disease pathogenesis. T cells show signs of antigen-induced activation; T cells of TH2 type are increased and produce profibrotic cytokines interleukin (IL)-4, IL-13, and IL-31; CD4+ cytotoxic T lymphocytes are increased in skin lesions, and cause fibrosis and endothelial cell apoptosis; circulating T follicular helper (TFH) cells are increased in SSc produce IL-21 and promote plasmablast antibody production. On the other hand, regulatory T cells are impaired in SSc. These findings provide strong circumstantial evidence for T cell implication in SSc pathogenesis and encourage new T cell-directed therapeutic strategies for the disease.
Collapse
|
26
|
Campochiaro C, Lazzaroni MG, Bruni C, Zanatta E, De Luca G, Matucci-Cerinic M. Open questions on the management of targeted therapies for the treatment of systemic sclerosis-interstitial lung disease: results of a EUSTAR survey based on a systemic literature review. Ther Adv Musculoskelet Dis 2022; 14:1759720X221116408. [PMID: 36051631 PMCID: PMC9425887 DOI: 10.1177/1759720x221116408] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 07/08/2022] [Indexed: 11/15/2022] Open
Abstract
Background The results of randomized controlled (RCT) and retrospective studies have expanded the armamentarium of drugs for systemic sclerosis (SSc) - interstitial lung disease (ILD) treatment. The correct positioning of these drugs is not yet clarified. Objectives Systemic literature review (SLR) on rituximab (RTX), tocilizumab (TCZ), nintedanib and abatacept (ABT) for the treatment of SSc-ILD. The results of the SLR were used to create a dedicated survey. Design The study was performed as a systematic review. Data sources and methods the SLR was performed using the following terms: "(systemic sclerosis OR scleroderma) AND (interstitial lung disease OR lung fibrosis OR pulmonary fibrosis) AND (rituximab OR tocilizumab OR abatacept OR nintedanib)". The results of the SLR were integrated in a survey including 8 domains. These were sent to all EUSTAR members and to the participants of the 2020 Scleroderma World Congress. Results 41 studies (34 on RTX, 5 on TCZ, 2 on ABT, and 1 on nintedanib) were identified. RCTs supported the use of TCZ and nintedanib, while retrospective studies supported the use of RTX for SSc-ILD. No clear data were obtained about ABT. The survey showed that RTX is the most available option (96%) whereas the most frequent reason for targeted therapy introduction is lung progression while on csDMARDs (86% RTX, 59% TCZ and 63% nintedanib). Combination therapy was the most frequently mentioned therapeutic scheme for nintedanib (75%) and RTX (63%). Physicians' perception of safety was similar for all drugs, while drug efficacy was the same for RTX and nintedanib, followed by TCZ (4.8 ± 2). The most frequently raised concerns pertained to efficacy, safety and combination regimens. Conclusion Our SLR supports the use of RTX, TCZ and nintedanib for SSc-ILD patients and underlines the need for more data about upfront combination versus monotherapy. It also highlighted the need to identify predictors supporting drug choice according to both pulmonary and extra-pulmonary manifestations.
Collapse
Affiliation(s)
- Corrado Campochiaro
- Unit of Immunology, Rheumatology, Allergy and Rare Disease, IRCCS San Raffaele Hospital, Vita-Salute San Raffaele University, Milan, Italy. Via Olgettina 60, 20132, Milan, Italy
| | - Maria Grazia Lazzaroni
- Rheumatology and Clinical Immunology Unit, ASST Spedali Civili of Brescia, Brescia, Italy
- Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Cosimo Bruni
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
- Department of Geriatric Medicine, Division of Rheumatology AOUC, Florence, Italy
- Department of Rheumatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Elisabetta Zanatta
- Rheumatology Unit, Department of Medicine, University of Padova, Padova, Italy
| | - Giacomo De Luca
- Unit of Immunology, Rheumatology, Allergy and Rare Disease, IRCCS San Raffaele Hospital, Vita-Salute San Raffaele University, Milan, Italy
| | - Marco Matucci-Cerinic
- Unit of Immunology, Rheumatology, Allergy and Rare Disease, IRCCS San Raffaele Hospital, Vita-Salute San Raffaele University, Milan, Italy
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
- Department of Geriatric Medicine, Division of Rheumatology AOUC, Florence, Italy
| |
Collapse
|
27
|
Kawashima-Vasconcelos MY, Santana-Gonçalves M, Zanin-Silva DC, Malmegrim KCR, Oliveira MC. Reconstitution of the immune system and clinical correlates after stem cell transplantation for systemic sclerosis. Front Immunol 2022; 13:941011. [PMID: 36032076 PMCID: PMC9403547 DOI: 10.3389/fimmu.2022.941011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 07/25/2022] [Indexed: 11/13/2022] Open
Abstract
Systemic sclerosis (SSc) is a chronic autoimmune disease that includes fibrosis, diffuse vasculopathy, inflammation, and autoimmunity. Autologous hematopoietic stem cell transplantation (auto-HSCT) is considered for patients with severe and progressive SSc. In recent decades, knowledge about patient management and clinical outcomes after auto-HSCT has significantly improved. Mechanistic studies have contributed to increasing the comprehension of how profound and long-lasting are the modifications to the immune system induced by transplantation. This review revisits the immune monitoring studies after auto-HSCT for SSc patients and how they relate to clinical outcomes. This understanding is essential to further improve clinical applications of auto-HSCT and enhance patient outcomes.
Collapse
Affiliation(s)
- Marianna Y. Kawashima-Vasconcelos
- Center for Cell-Based Therapy, Regional Hemotherapy Center of the Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
- Internal Medicine Graduate Program, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Maynara Santana-Gonçalves
- Center for Cell-Based Therapy, Regional Hemotherapy Center of the Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
- Oncology, Stem Cell and Cell-Therapy Graduate Program, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Djúlio C. Zanin-Silva
- Center for Cell-Based Therapy, Regional Hemotherapy Center of the Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
- Basic and Applied Immunology Graduate Program, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Kelen C. R. Malmegrim
- Center for Cell-Based Therapy, Regional Hemotherapy Center of the Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
- Department of Clinical, Toxicological and Bromatological Analysis, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Maria Carolina Oliveira
- Center for Cell-Based Therapy, Regional Hemotherapy Center of the Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
- Department of Internal Medicine, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| |
Collapse
|
28
|
Thoreau B, Chaigne B, Mouthon L. Role of B-Cell in the Pathogenesis of Systemic Sclerosis. Front Immunol 2022; 13:933468. [PMID: 35903091 PMCID: PMC9315392 DOI: 10.3389/fimmu.2022.933468] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Accepted: 06/06/2022] [Indexed: 11/29/2022] Open
Abstract
Systemic sclerosis (SSc) is a rare multisystem autoimmune disease, characterized by fibrosis, vasculopathy, and autoimmunity. Recent advances have highlighted the significant implications of B-cells in SSc. B-cells are present in affected organs, their subpopulations are disrupted, and they display an activated phenotype, and the regulatory capacities of B-cells are impaired, as illustrated by the decrease in the IL-10+ producing B-cell subpopulation or the inhibitory membrane co-receptor density. Recent multi-omics evidence highlights the role of B-cells mainly in the early stage of SSc and preferentially during severe organ involvement. This dysregulated homeostasis partly explains the synthesis of anti-endothelial cell autoantibodies (AECAs) or anti-fibroblast autoantibodies (AFAs), proinflammatory or profibrotic cytokines (interleukin-6 and transforming growth factor-β) produced by B and plasma cells. That is associated with cell-to-cell interactions with endothelial cells, fibroblasts, vascular smooth muscle cells, and other immune cells, altogether leading to cell activation and proliferation, cell resistance to apoptosis, the impairment of regulatory mechanisms, and causing fibrosis of several organs encountered in the SSc. Finally, alongside these exploratory data, treatments targeting B-cells, through their depletion by cytotoxicity (anti-CD20 monoclonal antibody), or the cytokines produced by the B-cell, or their costimulation molecules, seem interesting, probably in certain profiles of early patients with severe organic damage.
Collapse
Affiliation(s)
- Benjamin Thoreau
- Department of Internal Medicine, National Referral Center for Rare Systemic Autoimmune Diseases, Cochin Hospital, AP‐HP, CEDEX 14, Paris, France
- Université Paris Cité, Paris, France
- INSERM U1016, Cochin Institute, CNRS UMR 8104, Université Paris Cité, Paris, France
| | - Benjamin Chaigne
- Department of Internal Medicine, National Referral Center for Rare Systemic Autoimmune Diseases, Cochin Hospital, AP‐HP, CEDEX 14, Paris, France
- Université Paris Cité, Paris, France
- INSERM U1016, Cochin Institute, CNRS UMR 8104, Université Paris Cité, Paris, France
| | - Luc Mouthon
- Department of Internal Medicine, National Referral Center for Rare Systemic Autoimmune Diseases, Cochin Hospital, AP‐HP, CEDEX 14, Paris, France
- Université Paris Cité, Paris, France
- INSERM U1016, Cochin Institute, CNRS UMR 8104, Université Paris Cité, Paris, France
- *Correspondence: Luc Mouthon,
| |
Collapse
|
29
|
Melissaropoulos K, Iliopoulos G, Sakkas LI, Daoussis D. Pathogenetic Aspects of Systemic Sclerosis: A View Through the Prism of B Cells. Front Immunol 2022; 13:925741. [PMID: 35812378 PMCID: PMC9259786 DOI: 10.3389/fimmu.2022.925741] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 05/26/2022] [Indexed: 12/24/2022] Open
Abstract
Systemic sclerosis (SSc) is a rare fibrotic rheumatic disease, associated with psychological distress and increased morbidity and mortality due to skin involvement and internal organ damage. The current understanding of the complex pathogenesis is yet incomplete and disease therapeutic algorithms are far from optimal. Immunologic aberrations are considered key factors for the disease, along with vascular involvement and excess fibrosis. Adaptive immunity and its specialized responses are an attractive research target and both T and B cells have been extensively studied in recent years. In the present review, the focus is placed on B cells in SSc. B cell homeostasis is deranged and B cell subsets exhibit an activated phenotype and abnormal receptor signaling. Autoantibodies are a hallmark of the disease and the current perception of their diagnostic and pathogenetic role is analyzed. In addition, B cell cytokine release and its effect on immunity and fibrosis are examined, together with B cell tissue infiltration of the skin and lung. These data support the concept of targeting B cells as part of the therapeutic plan for SSc through well designed clinical trials.
Collapse
Affiliation(s)
| | - George Iliopoulos
- Department of Rheumatology , University of Patras Medical School, Patras University Hospital, Patras, Greece
| | - Lazaros I. Sakkas
- Department of Rheumatology and Clinical Immunology, Faculty of Medicine, School of Health Sciences, University of Thessaly, Larissa, Greece
| | - Dimitrios Daoussis
- Department of Rheumatology , University of Patras Medical School, Patras University Hospital, Patras, Greece
| |
Collapse
|
30
|
Kobayashi S, Nagafuchi Y, Shoda H, Fujio K. The Pathophysiological Roles of Regulatory T Cells in the Early Phase of Systemic Sclerosis. Front Immunol 2022; 13:900638. [PMID: 35686127 PMCID: PMC9172592 DOI: 10.3389/fimmu.2022.900638] [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: 03/21/2022] [Accepted: 04/26/2022] [Indexed: 11/13/2022] Open
Abstract
Systemic sclerosis (SSc) is an autoimmune disease that is characterized by vascular damage and fibrosis. Both clinical manifestations and immunological disturbances are diverse according to the disease duration. Particularly, changes in immunological processes are prominent in the early phase of SSc. The orchestration of several subsets of immune cells promotes autoimmune responses and inflammation, and eventually stimulates pro-fibrotic processes. Many reports have indicated that CD4+ T cells play pivotal roles in pathogenesis in the early phase of SSc. In particular, the pathogenic roles of regulatory T (Treg) cells have been investigated. Although the results were controversial, recent reports suggested an increase of Treg cells in the early phase of SSc patients. Treg cells secrete transforming growth factor-β (TGF-β), which promotes myofibroblast activation and fibrosis. In addition, the dysfunction of Treg cells in the early phase of SSc was reported, which results in the development of autoimmunity and inflammation. Notably, Treg cells have the plasticity to convert to T-helper17 (Th17) cells under pro-inflammatory conditions. Th17 cells secrete IL-17A, which could also promote myofibroblast transformation and fibrosis and contributes to vasculopathy, although the issue is still controversial. Our recent transcriptomic comparison between the early and late phases of SSc revealed a clear difference of gene expression patterns only in Treg cells. The gene signature of an activated Treg cell subpopulation was expanded in the early phase of SSc and the oxidative phosphorylation pathway was enhanced, which can promote Th17 differentiation. And this result was accompanied by the increase in Th17 cells frequency. Therefore, an imbalance between Treg and Th17 cells could also have an important role in the pathogenesis of the early phase of SSc. In this review, we outlined the roles of Treg cells in the early phase of SSc, summarizing the data of both human and mouse models. The contributions of Treg cells to autoimmunity, vasculopathy, and fibrosis were revealed, based on the dysfunction and imbalance of Treg cells. We also referred to the potential development in treatment strategies in SSc.
Collapse
Affiliation(s)
- Satomi Kobayashi
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Japan
- Department of Medicine and Rheumatology, Tokyo Metropolitan Geriatric Hospital, Itabashi-ku, Japan
| | - Yasuo Nagafuchi
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Japan
- Department of Functional Genomics and Immunological Diseases, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Japan
| | - Hirofumi Shoda
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Japan
| | - Keishi Fujio
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Japan
| |
Collapse
|
31
|
Dai B, Ding L, Zhao L, Zhu H, Luo H. Contributions of Immune Cells and Stromal Cells to the Pathogenesis of Systemic Sclerosis: Recent Insights. Front Pharmacol 2022; 13:826839. [PMID: 35185577 PMCID: PMC8852243 DOI: 10.3389/fphar.2022.826839] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 01/04/2022] [Indexed: 12/21/2022] Open
Abstract
Systemic sclerosis (SSc) is a multisystem rheumatic disease characterized by vascular dysfunction, autoimmune abnormalities, and progressive organ fibrosis. A series of studies in SSc patients and fibrotic models suggest that immune cells, fibroblasts, and endothelial cells participate in inflammation and aberrant tissue repair. Furthermore, the growing number of studies on single-cell RNA sequencing (scRNA-seq) technology in SSc elaborate on the transcriptomics and heterogeneities of these cell subsets significantly. In this review, we summarize the current knowledge regarding immune cells and stromal cells in SSc patients and discuss their potential roles in SSc pathogenesis, focusing on recent advances in the new subtypes by scRNA-seq.
Collapse
Affiliation(s)
- Bingying Dai
- Department of Rheumatology and Immunology, Xiangya Hospital, Central South University, Changsha, China
| | - Liqing Ding
- Department of Rheumatology and Immunology, Xiangya Hospital, Central South University, Changsha, China
| | - Lijuan Zhao
- Department of Rheumatology and Immunology, Xiangya Hospital, Central South University, Changsha, China
| | - Honglin Zhu
- Department of Rheumatology and Immunology, Xiangya Hospital, Central South University, Changsha, China
- Provincial Clinical Research Center for Rheumatic and Immunologic Diseases, Xiangya Hospital, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, China
- *Correspondence: Honglin Zhu, ; Hui Luo,
| | - Hui Luo
- Department of Rheumatology and Immunology, Xiangya Hospital, Central South University, Changsha, China
- Provincial Clinical Research Center for Rheumatic and Immunologic Diseases, Xiangya Hospital, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, China
- *Correspondence: Honglin Zhu, ; Hui Luo,
| |
Collapse
|
32
|
Papadimitriou TI, van Caam A, van der Kraan PM, Thurlings RM. Therapeutic Options for Systemic Sclerosis: Current and Future Perspectives in Tackling Immune-Mediated Fibrosis. Biomedicines 2022; 10:316. [PMID: 35203525 PMCID: PMC8869277 DOI: 10.3390/biomedicines10020316] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 01/21/2022] [Accepted: 01/26/2022] [Indexed: 02/01/2023] Open
Abstract
Systemic sclerosis (SSc) is a severe auto-immune, rheumatic disease, characterized by excessive fibrosis of the skin and visceral organs. SSc is accompanied by high morbidity and mortality rates, and unfortunately, few disease-modifying therapies are currently available. Inflammation, vasculopathy, and fibrosis are the key hallmarks of SSc pathology. In this narrative review, we examine the relationship between inflammation and fibrosis and provide an overview of the efficacy of current and novel treatment options in diminishing SSc-related fibrosis based on selected clinical trials. To do this, we first discuss inflammatory pathways of both the innate and acquired immune systems that are associated with SSc pathophysiology. Secondly, we review evidence supporting the use of first-line therapies in SSc patients. In addition, T cell-, B cell-, and cytokine-specific treatments that have been utilized in SSc are explored. Finally, the potential effectiveness of tyrosine kinase inhibitors and other novel therapeutic approaches in reducing fibrosis is highlighted.
Collapse
Affiliation(s)
- Theodoros-Ioannis Papadimitriou
- Department of Rheumatic Diseases, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands; (A.v.C.); (P.M.v.d.K.); (R.M.T.)
| | | | | | | |
Collapse
|
33
|
OUP accepted manuscript. Rheumatology (Oxford) 2022; 61:4364-4373. [DOI: 10.1093/rheumatology/keac023] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Revised: 12/21/2021] [Indexed: 11/13/2022] Open
|
34
|
Dang VD, Stefanski AL, Lino AC, Dörner T. B- and Plasma Cell Subsets in Autoimmune Diseases: Translational Perspectives. J Invest Dermatol 2021; 142:811-822. [PMID: 34955289 DOI: 10.1016/j.jid.2021.05.038] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 05/04/2021] [Accepted: 05/14/2021] [Indexed: 12/22/2022]
Abstract
B lymphocytes play a central role in immunity owing to their unique antibody-producing capacity that provides protection against certain infections and during vaccination. In autoimmune diseases, B cells can gain pathogenic relevance through autoantibody production, antigen presentation, and proinflammatory cytokine secretion. Recent data indicate that B and plasma cells can function as regulators through the production of immunoregulatory cytokines and/or employing checkpoint molecules. In this study, we review the key findings that define subsets of B and plasma cells with pathogenic and protective functions in autoimmunity. In addition to harsh B-cell depletion, we discuss the strategies that have the potential to reinstall the balance of pathogenic and protective B cells with the potential of more specific and personalized therapies.
Collapse
Affiliation(s)
- Van Duc Dang
- German Rheumatism Research Center (DRFZ) Berlin, a Leibniz Institute, Berlin, Germany; Department of Rheumatology and Clinical Immunology, Charite Universitatsmedizin Berlin, Berlin, Germany; Faculty of Biology, VNU University of Science, Vietnam National University, Hanoi, Vietnam
| | - Ana-Luisa Stefanski
- German Rheumatism Research Center (DRFZ) Berlin, a Leibniz Institute, Berlin, Germany; Department of Rheumatology and Clinical Immunology, Charite Universitatsmedizin Berlin, Berlin, Germany
| | - Andreia C Lino
- German Rheumatism Research Center (DRFZ) Berlin, a Leibniz Institute, Berlin, Germany; Department of Rheumatology and Clinical Immunology, Charite Universitatsmedizin Berlin, Berlin, Germany
| | - Thomas Dörner
- German Rheumatism Research Center (DRFZ) Berlin, a Leibniz Institute, Berlin, Germany; Department of Rheumatology and Clinical Immunology, Charite Universitatsmedizin Berlin, Berlin, Germany.
| |
Collapse
|
35
|
Rudnik M, Hukara A, Kocherova I, Jordan S, Schniering J, Milleret V, Ehrbar M, Klingel K, Feghali-Bostwick C, Distler O, Błyszczuk P, Kania G. Elevated Fibronectin Levels in Profibrotic CD14 + Monocytes and CD14 + Macrophages in Systemic Sclerosis. Front Immunol 2021; 12:642891. [PMID: 34504485 PMCID: PMC8421541 DOI: 10.3389/fimmu.2021.642891] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Accepted: 07/21/2021] [Indexed: 12/15/2022] Open
Abstract
Background Systemic sclerosis (SSc) is an autoimmune disease characterized by overproduction of extracellular matrix (ECM) and multiorgan fibrosis. Animal studies pointed to bone marrow-derived cells as a potential source of pathological ECM-producing cells in immunofibrotic disorders. So far, involvement of monocytes and macrophages in the fibrogenesis of SSc remains poorly understood. Methods and Results Immunohistochemistry analysis showed accumulation of CD14+ monocytes in the collagen-rich areas, as well as increased amount of alpha smooth muscle actin (αSMA)-positive fibroblasts, CD68+ and mannose-R+ macrophages in the heart and lungs of SSc patients. The full genome transcriptomics analyses of CD14+ blood monocytes revealed dysregulation in cytoskeleton rearrangement, ECM remodeling, including elevated FN1 (gene encoding fibronectin) expression and TGF-β signalling pathway in SSc patients. In addition, single cell RNA sequencing analysis of tissue-resident CD14+ pulmonary macrophages demonstrated activated profibrotic signature with the elevated FN1 expression in SSc patients with interstitial lung disease. Peripheral blood CD14+ monocytes obtained from either healthy subjects or SSc patients exposed to profibrotic treatment with profibrotic cytokines TGF-β, IL-4, IL-10, and IL-13 increased production of type I collagen, fibronectin, and αSMA. In addition, CD14+ monocytes co-cultured with dermal fibroblasts obtained from SSc patients or healthy individuals acquired a spindle shape and further enhanced production of profibrotic markers. Pharmacological blockade of the TGF-β signalling pathway with SD208 (TGF-β receptor type I inhibitor), SIS3 (Smad3 inhibitor) or (5Z)-7-oxozeaenol (TGF-β-activated kinase 1 inhibitor) ameliorated fibronectin levels and type I collagen secretion. Conclusions Our findings identified activated profibrotic signature with elevated production of profibrotic fibronectin in CD14+ monocytes and CD14+ pulmonary macrophages in SSc and highlighted the capability of CD14+ monocytes to acquire a profibrotic phenotype. Taking together, tissue-infiltrating CD14+ monocytes/macrophages can be considered as ECM producers in SSc pathogenesis.
Collapse
Affiliation(s)
- Michał Rudnik
- Department of Rheumatology, Center of Experimental Rheumatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Amela Hukara
- Department of Rheumatology, Center of Experimental Rheumatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Ievgeniia Kocherova
- Department of Rheumatology, Center of Experimental Rheumatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Suzana Jordan
- Department of Rheumatology, Center of Experimental Rheumatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Janine Schniering
- Department of Rheumatology, Center of Experimental Rheumatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Vincent Milleret
- Department of Obstetrics, University Hospital Zurich, Zurich, Switzerland
| | - Martin Ehrbar
- Department of Obstetrics, University Hospital Zurich, Zurich, Switzerland
| | - Karin Klingel
- Department of Molecular Pathology, University Hospital Tuebingen, Tuebingen, Germany
| | - Carol Feghali-Bostwick
- Division of Rheumatology, Medical University of South Carolina, Charleston, SC, United States
| | - Oliver Distler
- Department of Rheumatology, Center of Experimental Rheumatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Przemysław Błyszczuk
- Department of Rheumatology, Center of Experimental Rheumatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland.,Department of Clinical Immunology, Jagiellonian University Medical College, Krakow, Poland
| | - Gabriela Kania
- Department of Rheumatology, Center of Experimental Rheumatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| |
Collapse
|
36
|
Pauling JD, Christopher-Stine L. The aetiopathogenic significance, clinical relevance and therapeutic implications of vasculopathy in idiopathic inflammatory myopathy. Rheumatology (Oxford) 2021; 60:1593-1607. [PMID: 33458769 DOI: 10.1093/rheumatology/keaa816] [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] [Received: 07/08/2020] [Revised: 09/16/2020] [Accepted: 10/15/2020] [Indexed: 12/31/2022] Open
Abstract
It is 120 years since 'angiomyositis' was included alongside 'polymyositis' and 'dermatomyositis' in an attempt to propose a taxonomy that reflected the major clinical characteristics of idiopathic inflammatory myopathy (IIM). Endothelial injury, perivascular inflammation and capillary loss are important histological findings in affected tissues in IIM. Overt vascular clinical features including RP and abnormal nailfold capillaroscopy (NC) are also common in IIM. Despite the presence of endothelial injury, perivascular inflammation and capillary loss in affected tissues in IIM, and the presence of clinical features such as RP and NC abnormalities, the pathogenic and therapeutic implications of vasculopathy in IIM have been somewhat overlooked. RP and NC abnormalities are not always present, providing a valuable opportunity to explore aetiopathogenic factors driving vasculopathy within autoimmune rheumatic disease. The present review examines the aetiopathogenic, prognostic and therapeutic significance of vasculopathy in IIM. We describe the prevalence and clinical relevance of vasculopathy in IIM, and consider how vasculopathy may be better utilized to support improved IIM diagnosis and disease classification. Areas of unmet research need are highlighted where relevant.
Collapse
Affiliation(s)
- John D Pauling
- Royal National Hospital for Rheumatic Diseases (Part of the Royal United Hospitals NHS Foundation Trust), Bath, UK.,Department of Pharmacy and Pharmacology, University of Bath, Bath, UK
| | | |
Collapse
|
37
|
Orvain C, Boulch M, Bousso P, Allanore Y, Avouac J. Is there a place for CAR-T cells in the treatment of chronic autoimmune rheumatic diseases? Arthritis Rheumatol 2021; 73:1954-1965. [PMID: 34042325 DOI: 10.1002/art.41812] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Accepted: 05/11/2021] [Indexed: 11/09/2022]
Abstract
Chimeric-Antigen-Receptor T cell therapy or CAR-T cell is based on a specific targeting of tumor antigen leading to lysis and destruction of tumor cells development. CAR-T cells have demonstrated high potency for the management of B cell malignancies. This successful story was followed by the development of new CAR-T cell-derived constructions that have the ability to eradicate pathogenic B cells or restore tolerance. The objective of the herein manuscript is to review and discuss how the knowledge and technology generated by the use of CAR-T cells may be translated and integrated in the ongoing therapeutic strategies of autoimmune rheumatic diseases. To this end, we will introduce CAR-T cell technology, describe the meaningful achievements of CAR-T cells observed in onco-hematology and discuss preliminary data obtained with CAR-T cells and their derivative constructions in experimental models of autoimmune diseases. Then, we will focus on how CAR-T cell engineering is interfering with the pathogenesis of three chronic autoimmune rheumatic disorders - rheumatoid arthritis, systemic lupus erythematosus and systemic sclerosis - and discuss whether these constructs may permit to gain efficacy compared to current treatments and overcome their adverse events.
Collapse
Affiliation(s)
- Cindy Orvain
- INSERM U1016 and CNRS UMR8104, Institut Cochin, Paris, France
| | - Morgane Boulch
- Dynamics of Immune Responses Unit, Equipe Labellisée Ligue Contre le Cancer, Institut Pasteur, INSERM U1223, 75015, Paris, France
| | - Philippe Bousso
- Dynamics of Immune Responses Unit, Equipe Labellisée Ligue Contre le Cancer, Institut Pasteur, INSERM U1223, 75015, Paris, France
| | - Yannick Allanore
- INSERM U1016 and CNRS UMR8104, Institut Cochin, Paris, France.,Université de Paris, Université Paris Descartes, Paris, France.,Service de Rhumatologie, Hôpital Cochin, AP-HP.CUP, Paris, France
| | - Jérôme Avouac
- INSERM U1016 and CNRS UMR8104, Institut Cochin, Paris, France.,Université de Paris, Université Paris Descartes, Paris, France.,Service de Rhumatologie, Hôpital Cochin, AP-HP.CUP, Paris, France
| |
Collapse
|
38
|
De Luca G, Tomelleri A, Dagna L, Matucci-Cerinic M. The target on B cells in Systemic Sclerosis: a "midsummer dream" to extinguish inflammation and prevent early disease progression to fibrosis. Clin Rheumatol 2021; 40:2529-2533. [PMID: 34021841 DOI: 10.1007/s10067-021-05733-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Revised: 04/05/2021] [Accepted: 04/06/2021] [Indexed: 11/25/2022]
Affiliation(s)
- Giacomo De Luca
- Unit of Immunology, Rheumatology, Allergy and Rare diseases (UnIRAR), IRCCS San Raffaele Hospital, 20132, Milan, Italy
- Vita-Salute San Raffaele University, 20132, Milan, Italy
| | - Alessandro Tomelleri
- Unit of Immunology, Rheumatology, Allergy and Rare diseases (UnIRAR), IRCCS San Raffaele Hospital, 20132, Milan, Italy
- Vita-Salute San Raffaele University, 20132, Milan, Italy
| | - Lorenzo Dagna
- Unit of Immunology, Rheumatology, Allergy and Rare diseases (UnIRAR), IRCCS San Raffaele Hospital, 20132, Milan, Italy
- Vita-Salute San Raffaele University, 20132, Milan, Italy
| | - Marco Matucci-Cerinic
- Unit of Immunology, Rheumatology, Allergy and Rare diseases (UnIRAR), IRCCS San Raffaele Hospital, 20132, Milan, Italy.
- Department Experimental and Clinical Medicine and Division of Rheumatology AOUC, University of Florence, Via delle Oblate 4, 50139, Florence, Italy.
| |
Collapse
|
39
|
Rudnik M, Rolski F, Jordan S, Mertelj T, Stellato M, Distler O, Blyszczuk P, Kania G. Regulation of Monocyte Adhesion and Type I Interferon Signaling by CD52 in Patients With Systemic Sclerosis. Arthritis Rheumatol 2021; 73:1720-1730. [PMID: 33760395 DOI: 10.1002/art.41737] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Accepted: 03/11/2021] [Indexed: 12/21/2022]
Abstract
OBJECTIVE Systemic sclerosis (SSc) is characterized by dysregulation of type I interferon (IFN) signaling. CD52 is known for its immunosuppressive functions in T cells. This study was undertaken to investigate the role of CD52 in monocyte adhesion and type I IFN signaling in patients with SSc. METHODS Transcriptome profiles of circulating CD14+ monocytes from patients with limited cutaneous SSc (lcSSc), patients with diffuse cutaneous SSc (dcSSs), and healthy controls were analyzed by RNA sequencing. Levels of CD52, CD11b/integrin αΜ, and CD18/integrin β2 in whole blood were assessed by flow cytometry. CD52 expression was analyzed in relation to disease phenotype (early, lcSSc, dcSSc) and autoantibody profiles. The impact of overexpression, knockdown, and antibody blocking of CD52 was analyzed by gene and protein expression assays and functional assays. RESULTS Pathway enrichment analysis indicated an increase in adhesion- and type I IFN-related genes in monocytes from SSc patients. These cells displayed up-regulated expression of CD11b/CD18, reduced expression of CD52, and enhanced adhesion to intercellular adhesion molecule 1 and endothelial cells. Changes in CD52 expression were consistent with the SSc subtypes, as well as with immunosuppressive treatments, autoantibody profiles, and monocyte adhesion properties in patients with SSc. Overexpression of CD52 led to decreased levels of CD18 and monocyte adhesion, while knockdown of CD52 increased monocyte adhesion. Experiments with the humanized anti-CD52 monoclonal antibody alemtuzumab in blood samples from healthy controls increased monocyte adhesion and CD11b/CD18 expression, and enhanced type I IFN responses. Monocytic CD52 expression was up-regulated by interleukin-4 (IL-4)/IL-13 via the STAT6 pathway, and was down-regulated by lipopolysaccharide and IFNs α, β, and γ in a JAK1 and histone deacetylase IIa (HDAC IIa)-dependent manner. CONCLUSION Down-regulation of the antiadhesion CD52 antigen in CD14+ monocytes represents a novel mechanism in the pathogenesis of SSc. Targeting of the IFN-HDAC-CD52 axis in monocytes might represent a new therapeutic option for patients with early SSc.
Collapse
Affiliation(s)
- Michał Rudnik
- University Hospital Zurich and University of Zurich, Zurich, Switzerland
| | - Filip Rolski
- Jagiellonian University Medical College, Krakow, Poland
| | - Suzana Jordan
- University Hospital Zurich and University of Zurich, Zurich, Switzerland
| | - Tonja Mertelj
- University Hospital Zurich and University of Zurich, Zurich, Switzerland, and University Medical Centre Ljubljana, Ljubljana, Slovenia
| | - Mara Stellato
- University Hospital Zurich and University of Zurich, Zurich, Switzerland
| | - Oliver Distler
- University Hospital Zurich and University of Zurich, Zurich, Switzerland
| | - Przemysław Blyszczuk
- University Hospital Zurich and University of Zurich, Zurich, Switzerland, and Jagiellonian University Medical College, Krakow, Poland
| | - Gabriela Kania
- University Hospital Zurich and University of Zurich, Zurich, Switzerland
| |
Collapse
|
40
|
B cells in systemic sclerosis: from pathophysiology to treatment. Clin Rheumatol 2021; 40:2621-2631. [PMID: 33745085 DOI: 10.1007/s10067-021-05665-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 02/17/2021] [Accepted: 02/21/2021] [Indexed: 01/13/2023]
Abstract
Systemic sclerosis is a debilitating autoimmune disease with unknown pathogenesis. The clinical phenotype of fibrosis is preceded by vascular and immunologic aberrations. Adaptive immunity has been extensively studied in patients with the disease and B cells appear to be dysregulated. This is evident in peripheral blood B cell subsets, with activated effector B cells and impaired B regulatory function. In addition, B cells infiltrate target organs and tissues of patients with the disease, such as the skin and the lung, indicating a probable role in the pathogenesis. Impaired B cell homeostasis explains the rationale behind B cell therapeutic targeting. Indeed, several studies in recent years have shown that depletion of B cells appears to be a promising treatment alongside current established therapeutic choices, such as mycophenolate. In this review, B cell aberrations in animal models and human patients with systemic sclerosis will be presented. Moreover, we will also summarize current existing data regarding therapeutic targeting of the B cells in systemic sclerosis.
Collapse
|
41
|
Lerman I, Mitchell DC, Richardson CT. Human cutaneous B cells: what do we really know? ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:440. [PMID: 33842661 PMCID: PMC8033329 DOI: 10.21037/atm-20-5185] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
B cells play many critical roles in the systemic immune response, including antibody secretion, antigen presentation, T cell co-stimulation, and pro- and anti-inflammatory cytokine production. However, the contribution of B cells to the local immune response in many non-lymphoid tissues, such as the skin, is incompletely understood. Cutaneous B cells are scarce except in certain malignant and inflammatory conditions, and as such, have been poorly characterized until recently. Emerging evidence now suggests an important role for cutaneous B in both skin homeostasis and pathogenesis of skin disease. Herein, we discuss the potential mechanisms for cutaneous B cell recruitment, localized antibody production, and T cell interaction in human skin infections and primary skin malignancies (i.e., melanoma, squamous cell carcinoma). We further consider the likely contribution of cutaneous B cells to the pathogenesis of inflammatory skin diseases, including pemphigus vulgaris, lupus erythematosus, systemic sclerosis, hidradenitis suppurativa, and atopic dermatitis. Finally, we examine the feasibility of B cell targeted therapy in the dermatologic setting, emphasizing areas that are still open to investigation. Through this review, we hope to highlight what we really know about cutaneous B cells in human skin, which can sometimes be lost in reviews that more broadly incorporate extensive data from animal models.
Collapse
Affiliation(s)
- Irina Lerman
- University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Drew C Mitchell
- University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Christopher T Richardson
- Department of Dermatology, University of Rochester Medical Center, Rochester, NY, USA.,Division of Allergy, Immunology and Rheumatology, University of Rochester Medical Center, Rochester, NY, USA
| |
Collapse
|
42
|
Campochiaro C, De Luca G, Lazzaroni MG, Zanatta E, Bosello SL, De Santis M, Cariddi A, Bruni C, Selmi C, Gremese E, Matucci-Cerinic M, Doria A, Airò P, Dagna L. Safety and efficacy of rituximab biosimilar (CT-P10) in systemic sclerosis: an Italian multicentre study. Rheumatology (Oxford) 2021; 59:3731-3736. [PMID: 32413118 DOI: 10.1093/rheumatology/keaa136] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Accepted: 02/26/2020] [Indexed: 02/07/2023] Open
Abstract
OBJECTIVES Recent data have shown a significant efficacy of rituximab (RTX) in SSc. An RTX biosimilar (RTX-B) is a more affordable option. We assessed the safety and efficacy of an RTX-B (CT-P10) in SSc. METHODS SSc patients treated with RTX-B with at least 6 months of follow-up were retrospectively identified from six Italian referral centres. SSc patients naïve to RTX-B (RTX-Bn) or already treated with RTX originator and switched to an RTX-B (RTX-Bs) were evaluated. A comprehensive assessment of disease characteristics and organ involvement at baseline and after 6 months was obtained. RESULTS Thirty-three SSc patients were selected: 29 (87.9%) females, mean age 51.6 years (s.d. 14.2), mean disease duration 9.8 years (s.d. 8.1); 21 (64.5%) with dcSSc, 20 (60.1%) anti-topoisomerase I, 7 (21.2%) anti-RNA polymerase III and 6 (18.2%) anti-centromere positive. Seventeen (51.5%) were RTX-Bn and 16 were on RTX-Bs (48.5%). RTX was introduced because of skin progression in 18 patients (54.5%), interstitial lung disease (ILD) worsening in 11 (33.3%) and arthritis in 12 (36.4%). All patients were previously treated with immunosuppressants. At RTX-B introduction, 21 (63.6%) patients were on concomitant immunosuppressants: 15 (71.4%) on MMF and 6 (28.6%) on MTX. Twenty-three (69.7%) were on low-dose steroids. After 6 months, a significant reduction of the modified Rodnan skin score (mRSS), 28-joint DAS and CRP was observed (P = 0.002, 0.005 and 0.008, respectively); the mRSS significantly improved both in RTX-Bn (P < 0.024) and RTX-Bs patients (P < 0.031). No significant changes were observed for lung function tests, either in the entire cohort or in the subgroup of ILD patients. Only one RTX-Bs patient experienced transient neutropenia. CONCLUSION Our data suggest that RTX-B can represent a cheaper option in SSc patients, as it is effective in improving skin and joint involvement and in stabilizing lung function.
Collapse
Affiliation(s)
- Corrado Campochiaro
- Unit of Immunology, Rheumatology, Allergy and Rare Diseases, IRCCS San Raffaele Hospital, Vita-Salute San Raffaele University, Milan
| | - Giacomo De Luca
- Unit of Immunology, Rheumatology, Allergy and Rare Diseases, IRCCS San Raffaele Hospital, Vita-Salute San Raffaele University, Milan
| | | | - Elisabetta Zanatta
- Division of Rheumatology, Department of Medicine, DIMED, University of Padova, Padova
| | - Silvia Laura Bosello
- Division of Rheumatology, Fondazione Policlinico Universitario A. Gemelli-IRCCS, Rome
| | - Maria De Santis
- Division of Rheumatology and Clinical Immunology, Humanitas Clinical and Research Center-IRCCS, Rozzano, Milan
| | - Adriana Cariddi
- Unit of Immunology, Rheumatology, Allergy and Rare Diseases, IRCCS San Raffaele Hospital, Vita-Salute San Raffaele University, Milan
| | - Cosimo Bruni
- Division of Rheumatology, Azienda Ospedaliero Universitaria Careggi, University of Florence
| | - Carlo Selmi
- Division of Rheumatology and Clinical Immunology, Humanitas Clinical and Research Center-IRCCS, Rozzano, Milan.,Department of Biomedical Sciences, Humanitas University, Milan
| | - Elisa Gremese
- Division of Rheumatology, Fondazione Policlinico Universitario A. Gemelli-IRCCS, Rome.,Institute of Rheumatology, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Marco Matucci-Cerinic
- Division of Rheumatology, Azienda Ospedaliero Universitaria Careggi, University of Florence
| | - Andrea Doria
- Division of Rheumatology, Department of Medicine, DIMED, University of Padova, Padova
| | - Paolo Airò
- Rheumatology and Clinical Immunology, Spedali Civili and University of Brescia, Brescia
| | - Lorenzo Dagna
- Unit of Immunology, Rheumatology, Allergy and Rare Diseases, IRCCS San Raffaele Hospital, Vita-Salute San Raffaele University, Milan
| |
Collapse
|
43
|
Pauling JD, Skeoch S, Paik JJ. The clinicoserological spectrum of inflammatory myopathy in the context of systemic sclerosis and systemic lupus erythematosus. INDIAN JOURNAL OF RHEUMATOLOGY 2021; 15:81-90. [PMID: 33790525 DOI: 10.4103/injr.injr_136_20] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The autoimmune rheumatic diseases (ARDs) are characterised by a pathological triad composed of autoimmunity/inflammation, microangiopathy and aberrant tissue remodelling. Disease terms such as idiopathic inflammatory myopathy (IIM), scleroderma/systemic sclerosis (SSc), and systemic lupus erythematosus (SLE) are helpful clinically but disguise the considerable overlap that exists within these 'distinct' disorders. This is perhaps best demonstrated by inflammatory myopathy, which can be present in SSc or SLE, but can itself be absent in clinically amyopathic IIM. Archetypal clinical manifestations of ARD (such as Raynaud's phenomenon) are frequently present, albeit with varying prominence, within each of these diseases. This is certainly the case for inflammatory myositis, which has long been recognised as an important clinical feature of both SSc and SLE. Progress in elucidating the clinicoserological spectrum of autoimmune rheumatic diseases has identified autoantibody specificities that are strongly associated with 'overlap' disease and the presence of inflammatory myositis in SSc and SLE. In this review, we shall describe the prevalence, burden, prognostic value and management considerations of IIM in the context of both SSc and SLE. A major emphasis on the value of autoantibodies shall highlight the value of these tools in predicting the future occurrence of inflammatory myositis in both SSc and SLE. Where applicable, unmet research needs shall be highlighted. The review emphasises the importance of myopathy as a common feature across all the ARDs, and highlights specific antibody specificities that are strongly associated with myopathy in the context of SLE and SSc.
Collapse
Affiliation(s)
- John D Pauling
- Royal National Hospital for Rheumatic Diseases (part of the Royal United Hospitals NHS Foundation Trust), Bath, UK.,Department of Pharmacy and Pharmacology, University of Bath, Bath, UK
| | - Sarah Skeoch
- Royal National Hospital for Rheumatic Diseases (part of the Royal United Hospitals NHS Foundation Trust), Bath, UK.,Department of Pharmacy and Pharmacology, University of Bath, Bath, UK
| | - Julie J Paik
- Johns Hopkins Myositis Center. 5200 Eastern Avenue, MFL Building, Center Tower Suite 4500, Baltimore, MD USA
| |
Collapse
|
44
|
Skin-Associated B Cells in the Pathogenesis of Cutaneous Autoimmune Diseases-Implications for Therapeutic Approaches. Cells 2020; 9:cells9122627. [PMID: 33297481 PMCID: PMC7762338 DOI: 10.3390/cells9122627] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Revised: 12/01/2020] [Accepted: 12/04/2020] [Indexed: 12/11/2022] Open
Abstract
B lymphocytes are crucial mediators of systemic immune responses and are known to be substantial in the pathogenesis of autoimmune diseases with cutaneous manifestations. Amongst them are lupus erythematosus, dermatomyositis, systemic sclerosis and psoriasis, and particularly those driven by autoantibodies such as pemphigus and pemphigoid. However, the concept of autoreactive skin-associated B cells, which may reside in the skin and locally contribute to chronic inflammation, is gradually evolving. These cells are believed to differ from B cells of primary and secondary lymphoid organs and may provide additional features besides autoantibody production, including cytokine expression and crosstalk to autoreactive T cells in an antigen-presenting manner. In chronically inflamed skin, B cells may appear in tertiary lymphoid structures. Those abnormal lymph node-like structures comprise a network of immune and stromal cells possibly enriched by vascular structures and thus constitute an ideal niche for local autoimmune responses. In this review, we describe current considerations of different B cell subsets and their assumed role in skin autoimmunity. Moreover, we discuss traditional and B cell-associated approaches for the treatment of autoimmune skin diseases, including drugs targeting B cells (e.g., CD19- and CD20-antibodies), plasma cells (e.g., proteasome inhibitors, CXCR4 antagonists), activated pathways (such as BTK- and PI3K-inhibitors) and associated activator molecules (BLyS, APRIL).
Collapse
|
45
|
Higashioka K, Kikushige Y, Ayano M, Kimoto Y, Mitoma H, Kikukawa M, Akahoshi M, Arinobu Y, Horiuchi T, Akashi K, Niiro H. Generation of a novel CD30 + B cell subset producing GM-CSF and its possible link to the pathogenesis of systemic sclerosis. Clin Exp Immunol 2020; 201:233-243. [PMID: 32538493 PMCID: PMC7419935 DOI: 10.1111/cei.13477] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2020] [Revised: 05/26/2020] [Accepted: 06/06/2020] [Indexed: 12/12/2022] Open
Abstract
Systemic sclerosis (SSc) is a T helper type 2 (Th2)-associated autoimmune disease characterized by vasculopathy and fibrosis. Efficacy of B cell depletion therapy underscores antibody-independent functions of B cells in SSc. A recent study showed that the Th2 cytokine interleukin (IL)-4 induces granulocyte-macrophage colony-stimulating factor (GM-CSF)-producing effector B cells (GM-Beffs ) in humans. In this study, we sought to elucidate the generation mechanism of GM-Beffs and also determine a role of this subset in SSc. Among Th-associated cytokines, IL-4 most significantly facilitated the generation of GM-Beffs within memory B cells in healthy controls (HCs). In addition, the profibrotic cytokine transforming growth factor (TGF)-β further potentiated IL-4- and IL-13-induced GM-Beffs . Of note, tofacitinib, a Janus kinase (JAK) inhibitor, inhibited the expression of GM-CSF mRNA and protein in memory B cells induced by IL-4, but not by TGF-β. GM-Beffs were enriched within CD20+ CD30+ CD38-/low cells, a distinct population from plasmablasts, suggesting that GM-Beffs exert antibody-independent functions. GM-Beffs were also enriched in a CD30+ fraction of freshly isolated B cells. GM-Beffs generated under Th2 conditions facilitated the differentiation from CD14+ monocytes to DC-SIGN+ CD1a+ CD14- CD86+ cells, which significantly promoted the proliferation of naive T cells. CD30+ GM-Beffs were more pronounced in patients with SSc than in HCs. A subpopulation of SSc patients with the diffuse type and concomitant interstitial lung disease exhibited high numbers of GM-Beffs . Together, these findings suggest that human GM-Beffs are enriched in a CD30+ B cell subset and play a role in the pathogenesis of SSc.
Collapse
Affiliation(s)
- K. Higashioka
- Department of Medicine and Biosystemic ScienceGraduate School of Medical SciencesKyushu UniversityFukuokaJapan
| | - Y. Kikushige
- Department of Medicine and Biosystemic ScienceGraduate School of Medical SciencesKyushu UniversityFukuokaJapan
| | - M. Ayano
- Department of Medicine and Biosystemic ScienceGraduate School of Medical SciencesKyushu UniversityFukuokaJapan
| | - Y. Kimoto
- Department of Internal MedicineKyushu University Beppu HospitalTsurumiharaBeppuOitaJapan
| | - H. Mitoma
- Department of Medicine and Biosystemic ScienceGraduate School of Medical SciencesKyushu UniversityFukuokaJapan
| | - M. Kikukawa
- Department of Medical EducationFaculty of Medical SciencesKyushu UniversityFukuokaJapan
| | - M. Akahoshi
- Department of Medicine and Biosystemic ScienceGraduate School of Medical SciencesKyushu UniversityFukuokaJapan
| | - Y. Arinobu
- Department of Medicine and Biosystemic ScienceGraduate School of Medical SciencesKyushu UniversityFukuokaJapan
| | - T. Horiuchi
- Department of Internal MedicineKyushu University Beppu HospitalTsurumiharaBeppuOitaJapan
| | - K. Akashi
- Department of Medicine and Biosystemic ScienceGraduate School of Medical SciencesKyushu UniversityFukuokaJapan
| | - H. Niiro
- Department of Medical EducationFaculty of Medical SciencesKyushu UniversityFukuokaJapan
| |
Collapse
|
46
|
The Pathogenesis of Systemic Sclerosis: An Understanding Based on a Common Pathologic Cascade across Multiple Organs and Additional Organ-Specific Pathologies. J Clin Med 2020; 9:jcm9092687. [PMID: 32825112 PMCID: PMC7565034 DOI: 10.3390/jcm9092687] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 08/14/2020] [Accepted: 08/17/2020] [Indexed: 02/08/2023] Open
Abstract
Systemic sclerosis (SSc) is a multisystem autoimmune and vascular disease resulting in fibrosis of various organs with unknown etiology. Accumulating evidence suggests that a common pathologic cascade across multiple organs and additional organ-specific pathologies underpin SSc development. The common pathologic cascade starts with vascular injury due to autoimmune attacks and unknown environmental factors. After that, dysregulated angiogenesis and defective vasculogenesis promote vascular structural abnormalities, such as capillary loss and arteriolar stenosis, while aberrantly activated endothelial cells facilitate the infiltration of circulating immune cells into perivascular areas of various organs. Arteriolar stenosis directly causes pulmonary arterial hypertension, scleroderma renal crisis and digital ulcers. Chronic inflammation persistently activates interstitial fibroblasts, leading to the irreversible fibrosis of multiple organs. The common pathologic cascade interacts with a variety of modifying factors in each organ, such as keratinocytes and adipocytes in the skin, esophageal stratified squamous epithelia and myenteric nerve system in gastrointestinal tract, vasospasm of arterioles in the heart and kidney, and microaspiration of gastric content in the lung. To better understand SSc pathogenesis and develop new disease-modifying therapies, it is quite important to understand the complex pathogenesis of SSc from the two distinct perspectives, namely the common pathologic cascade and additional organ-specific pathologies.
Collapse
|
47
|
Worrell JC, O'Reilly S. Bi-directional communication: Conversations between fibroblasts and immune cells in systemic sclerosis. J Autoimmun 2020; 113:102526. [PMID: 32713676 DOI: 10.1016/j.jaut.2020.102526] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 07/13/2020] [Accepted: 07/14/2020] [Indexed: 02/09/2023]
Abstract
Systemic Sclerosis (SSc) is an autoimmune idiopathic connective tissue disease, characterized by aberrant fibro-proliferative and inflammatory responses, causing fibrosis of multiple organs. In recent years the interactions between innate and adaptive immune cells with resident fibroblasts have been uncovered. Cross-talk between immune and stromal cells mediates activation of stromal cells to myofibroblasts; key cells in the pathophysiology of fibrosis. These cells and their cytokines appear to mediate their effects in both a paracrine and autocrine fashion. This review examines the role of innate and adaptive immune cells in SSc, focusing on recent advances that have illuminated our understanding of ongoing bi-directional communication between immune and stromal cells. Finally, we appraise current and future therapies and how these may be useful in a disease that currently has no specific disease modifying treatment.
Collapse
Affiliation(s)
- Julie C Worrell
- Insititute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK
| | - Steven O'Reilly
- Durham University, Biosciences, Faculty of Science, Durham, UK. steven.o'
| |
Collapse
|
48
|
Chandrasekhar JL, Cox KM, Erickson LD. B Cell Responses in the Development of Mammalian Meat Allergy. Front Immunol 2020; 11:1532. [PMID: 32765532 PMCID: PMC7379154 DOI: 10.3389/fimmu.2020.01532] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Accepted: 06/10/2020] [Indexed: 12/11/2022] Open
Abstract
Studies of meat allergic patients have shown that eating meat poses a serious acute health risk that can induce severe cutaneous, gastrointestinal, and respiratory reactions. Allergic reactions in affected individuals following meat consumption are mediated predominantly by IgE antibodies specific for galactose-α-1,3-galactose (α-gal), a blood group antigen of non-primate mammals and therefore present in dietary meat. α-gal is also found within certain tick species and tick bites are strongly linked to meat allergy. Thus, it is thought that exposure to tick bites promotes cutaneous sensitization to tick antigens such as α-gal, leading to the development of IgE-mediated meat allergy. The underlying immune mechanisms by which skin exposure to ticks leads to the production of α-gal-specific IgE are poorly understood and are key to identifying novel treatments for this disease. In this review, we summarize the evidence of cutaneous exposure to tick bites and the development of mammalian meat allergy. We then provide recent insights into the role of B cells in IgE production in human patients with mammalian meat allergy and in a novel mouse model of meat allergy. Finally, we discuss existing data more generally focused on tick-mediated immunomodulation, and highlight possible mechanisms for how cutaneous exposure to tick bites might affect B cell responses in the skin and gut that contribute to loss of oral tolerance.
Collapse
Affiliation(s)
- Jessica L Chandrasekhar
- Beirne B. Carter Center for Immunology Research, University of Virginia School of Medicine, Charlottesville, VA, United States
| | - Kelly M Cox
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia School of Medicine, Charlottesville, VA, United States
| | - Loren D Erickson
- Beirne B. Carter Center for Immunology Research, University of Virginia School of Medicine, Charlottesville, VA, United States.,Department of Microbiology, Immunology, and Cancer Biology, University of Virginia School of Medicine, Charlottesville, VA, United States
| |
Collapse
|
49
|
CXCL4 triggers monocytes and macrophages to produce PDGF-BB, culminating in fibroblast activation: Implications for systemic sclerosis. J Autoimmun 2020; 111:102444. [DOI: 10.1016/j.jaut.2020.102444] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 03/28/2020] [Accepted: 03/29/2020] [Indexed: 12/26/2022]
|
50
|
A Niche for Plasma Cells: The Skin. J Invest Dermatol 2020; 139:2411-2414. [PMID: 31753124 DOI: 10.1016/j.jid.2019.06.133] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Revised: 06/18/2019] [Accepted: 06/18/2019] [Indexed: 01/02/2023]
Abstract
Antibodies are key components of the skin immune barrier, and antibodies directed toward skin structures can result in disease. Wilson et al. (2019) show that healthy skin is a niche for antibody secreting plasma cells and plasmablasts, and that inflammation and immunization increase their numbers. This work advances our understanding of skin associated B and plasma cells in health and disease.
Collapse
|