1
|
Liu M. Effector and regulatory B-cell imbalance in systemic sclerosis: cooperation or competition? Clin Rheumatol 2024; 43:2783-2789. [PMID: 39080112 PMCID: PMC11330388 DOI: 10.1007/s10067-024-07086-0] [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: 03/24/2024] [Revised: 07/19/2024] [Accepted: 07/23/2024] [Indexed: 08/18/2024]
Abstract
B cells play a central role in the pathogenesis of systemic sclerosis (SSc). Most B-cell studies have focused on their pathological role as antibody producers. However, in addition to immunoglobulin secretion, these cells have a wide range of functions in the immune response, including antigen presentation to T cells and cytokine production. Importantly, not all B-cell subsets promote the immune response. Regulatory B cells (Bregs) attenuate inflammation and contribute to the maintenance of immune tolerance. However, effector B cells (Beffs) positively modulate the immune response through the production of various cytokines. In SSc, Bregs are insufficient and/or dysfunctional. B-cell-targeting biologics have been trialled with promising results in the treatment of SSc. These therapies can affect Bregs or Beffs, which can potentially limit their long-term efficacy. Future strategies might involve the modulation of effector B cells in combination with the stimulation of regulatory subsets. Additionally, the monitoring of individual B-cell subsets in patients may lead to the discovery of novel biomarkers that could help predict disease relapse or progression. The purpose of this review is to summarize the relevant literatures and explain how Bregs and Beffs jointly participate in the pathogenesis of SSc.
Collapse
Affiliation(s)
- Mengguo Liu
- Department of Dermatology, Huashan Hospital, Fudan University, the 12Th Urumqi Road, Shanghai, 200040, China.
| |
Collapse
|
2
|
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
|
3
|
Streutker EM, Devamoglu U, Vonk MC, Verdurmen WPR, Le Gac S. Fibrosis-on-Chip: A Guide to Recapitulate the Essential Features of Fibrotic Disease. Adv Healthc Mater 2024; 13:e2303991. [PMID: 38536053 DOI: 10.1002/adhm.202303991] [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: 11/14/2023] [Revised: 03/15/2024] [Indexed: 05/05/2024]
Abstract
Fibrosis, which is primarily marked by excessive extracellular matrix (ECM) deposition, is a pathophysiological process associated with many disorders, which ultimately leads to organ dysfunction and poor patient outcomes. Despite the high prevalence of fibrosis, currently there exist few therapeutic options, and importantly, there is a paucity of in vitro models to accurately study fibrosis. This review discusses the multifaceted nature of fibrosis from the viewpoint of developing organ-on-chip (OoC) disease models, focusing on five key features: the ECM component, inflammation, mechanical cues, hypoxia, and vascularization. The potential of OoC technology is explored for better modeling these features in the context of studying fibrotic diseases and the interplay between various key features is emphasized. This paper reviews how organ-specific fibrotic diseases are modeled in OoC platforms, which elements are included in these existing models, and the avenues for novel research directions are highlighted. Finally, this review concludes with a perspective on how to address the current gap with respect to the inclusion of multiple features to yield more sophisticated and relevant models of fibrotic diseases in an OoC format.
Collapse
Affiliation(s)
- Emma M Streutker
- Department of Medical BioSciences, Radboud University Medical Center, Geert Grooteplein 28, Nijmegen, 6525 GA, The Netherlands
| | - Utku Devamoglu
- Applied Microfluidics for BioEngineering Research, MESA+ Institute for Nanotechnoloygy and TechMed Centre, Organ-on-Chip Centre, University of Twente, Drienerlolaan 5, Enschede, 7522 NB, The Netherlands
| | - Madelon C Vonk
- Department of Rheumatology, Radboud University Medical Center, Nijmegen, PO Box 9101, Nijmegen, 6500 HB, The Netherlands
| | - Wouter P R Verdurmen
- Department of Medical BioSciences, Radboud University Medical Center, Geert Grooteplein 28, Nijmegen, 6525 GA, The Netherlands
| | - Séverine Le Gac
- Applied Microfluidics for BioEngineering Research, MESA+ Institute for Nanotechnoloygy and TechMed Centre, Organ-on-Chip Centre, University of Twente, Drienerlolaan 5, Enschede, 7522 NB, The Netherlands
| |
Collapse
|
4
|
Murphy SL, Balzer NR, Ranheim T, Sagen EL, Huse C, Bjerkeli V, Michelsen AE, Finbråten AK, Heggelund L, Dyrhol-Riise AM, Tveita A, Holten AR, Trøseid M, Ueland T, Ulas T, Aukrust P, Barratt-Due A, Halvorsen B, Dahl TB. Extracellular matrix remodelling pathway in peripheral blood mononuclear cells from severe COVID-19 patients: an explorative study. Front Immunol 2024; 15:1379570. [PMID: 38957465 PMCID: PMC11217192 DOI: 10.3389/fimmu.2024.1379570] [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: 01/31/2024] [Accepted: 06/03/2024] [Indexed: 07/04/2024] Open
Abstract
There is a reciprocal relationship between extracellular matrix (ECM) remodelling and inflammation that could be operating in the progression of severe COVID-19. To explore the immune-driven ECM remodelling in COVID-19, we in this explorative study analysed these interactions in hospitalised COVID-19 patients. RNA sequencing and flow analysis were performed on peripheral blood mononuclear cells. Inflammatory mediators in plasma were measured by ELISA and MSD, and clinical information from hospitalised COVID-19 patients (N=15) at admission was included in the analysis. Further, we reanalysed two publicly available datasets: (1) lung tissue RNA-sequencing dataset (N=5) and (2) proteomics dataset from PBCM. ECM remodelling pathways were enriched in PBMC from COVID-19 patients compared to healthy controls. Patients treated at the intensive care unit (ICU) expressed distinct ECM remodelling gene profiles compared to patients in the hospital ward. Several markers were strongly correlated to immune cell subsets, and the dysregulation in the ICU patients was positively associated with plasma levels of inflammatory cytokines and negatively associated with B-cell activating factors. Finally, our analysis of publicly accessible datasets revealed (i) an augmented ECM remodelling signature in inflamed lung tissue compared to non-inflamed tissue and (ii) proteomics analysis of PBMC from severe COVID-19 patients demonstrated an up-regulation in an ECM remodelling pathway. Our results may suggest the presence of an interaction between ECM remodelling, inflammation, and immune cells, potentially initiating or perpetuating pulmonary pathology in severe COVID-19.
Collapse
Affiliation(s)
- Sarah Louise Murphy
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Nora Reka Balzer
- Genomics and Immunoregulation, Life & Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany
- Systems Medicine, German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- PRECISE Platform for Single Cell Genomics and Epigenomics, German Center for Neurodegenerative Diseases and the University of Bonn, Bonn, Germany
| | - Trine Ranheim
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Ellen Lund Sagen
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Camilla Huse
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
- Department of Medicine, Division of Cardiovascular Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, United States
| | - Vigdis Bjerkeli
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Annika E. Michelsen
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | | | - Lars Heggelund
- Department of Internal Medicine, Drammen Hospital, Vestre Viken Hospital Trust, Drammen, Norway
- Department of Clinical Science, Faculty of Medicine, University of Bergen, Bergen, Norway
| | - Anne Ma Dyrhol-Riise
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
- Department of Infectious Diseases, Oslo University Hospital Ullevål, Oslo, Norway
| | - Anders Tveita
- Department of Internal Medicine, Bærum Hospital, Vestre Viken Hospital Trust, Gjettum, Norway
- Division of Laboratory Medicine, Department of Immunology, Oslo University Hospital, Oslo, Norway
| | - Aleksander Rygh Holten
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
- Department of Acute Medicine, Oslo University Hospital, Oslo, Norway
| | - Marius Trøseid
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
- Section of Clinical Immunology and Infectious Diseases, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Thor Ueland
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
- Thrombosis Research Center (TREC), Division of Internal Medicine, University Hospital of North Norway, Tromsø, Norway
| | - Thomas Ulas
- Genomics and Immunoregulation, Life & Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany
- Systems Medicine, German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- PRECISE Platform for Single Cell Genomics and Epigenomics, German Center for Neurodegenerative Diseases and the University of Bonn, Bonn, Germany
| | - Pål Aukrust
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
- Section of Clinical Immunology and Infectious Diseases, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Andreas Barratt-Due
- Division of Laboratory Medicine, Department of Immunology, Oslo University Hospital, Oslo, Norway
- Department of Anesthesia and Intensive Care Medicine, Oslo University Hospital, Oslo, Norway
| | - Bente Halvorsen
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
- Department of Medicine, Division of Cardiovascular Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, United States
| | - Tuva Børresdatter Dahl
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway
| |
Collapse
|
5
|
Möckel T, Boegel S, Schwarting A. Transcriptome Analysis of BAFF/BAFF-R System in Murine Nephrotoxic Serum Nephritis. Int J Mol Sci 2024; 25:5415. [PMID: 38791453 PMCID: PMC11121395 DOI: 10.3390/ijms25105415] [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: 04/14/2024] [Revised: 05/02/2024] [Accepted: 05/07/2024] [Indexed: 05/26/2024] Open
Abstract
Chronic kidney disease (CKD) is an emerging cause for morbidity and mortality worldwide. Acute kidney injury (AKI) can transition to CKD and finally to end-stage renal disease (ESRD). Targeted treatment is still unavailable. NF-κB signaling is associated with CKD and activated by B cell activating factor (BAFF) via BAFF-R binding. In turn, renal tubular epithelial cells (TECs) are critical for the progression of fibrosis and producing BAFF. Therefore, the direct involvement of the BAFF/BAFF-R system to the pathogenesis of CKD is conceivable. We performed non-accelerated nephrotoxic serum nephritis (NTN) as the CKD model in BAFF KO (B6.129S2-Tnfsf13btm1Msc/J), BAFF-R KO (B6(Cg)-Tnfrsf13ctm1Mass/J) and wildtype (C57BL/6J) mice to analyze the BAFF/BAFF-R system in anti-glomerular basement membrane (GBM) disease using high throughput RNA sequencing. We found that BAFF signaling is directly involved in the upregulation of collagen III as BAFF ko mice showed a reduced expression. However, these effects were not mediated via BAFF-R. We identified several upregulated genes that could explain the effects of BAFF in chronic kidney injury such as Txnip, Gpx3, Igfbp7, Ccn2, Kap, Umod and Ren1. Thus, we conclude that targeted treatment with anti-BAFF drugs such as belimumab may reduce chronic kidney damage. Furthermore, upregulated genes may be useful prognostic CKD biomarkers.
Collapse
Affiliation(s)
- Tamara Möckel
- Division of Rheumatology and Clinical Immunology, Department of Internal Medicine I, University Medical Center of the Johannes Gutenberg University, 55131 Mainz, Germany; (T.M.); (S.B.)
| | - Sebastian Boegel
- Division of Rheumatology and Clinical Immunology, Department of Internal Medicine I, University Medical Center of the Johannes Gutenberg University, 55131 Mainz, Germany; (T.M.); (S.B.)
| | - Andreas Schwarting
- Division of Rheumatology and Clinical Immunology, Department of Internal Medicine I, University Medical Center of the Johannes Gutenberg University, 55131 Mainz, Germany; (T.M.); (S.B.)
- Center for Rheumatic Disease Rhineland-Palatinate GmbH, 55543 Bad Kreuznach, Germany
| |
Collapse
|
6
|
Shi Y, You H, Liu C, Qiu Y, Lv C, Zhu Y, Xu L, Wang F, Zhang M, Tan W. Elevated serum B-cell activator factor levels predict rapid progressive interstitial lung disease in anti-melanoma differentiation associated protein 5 antibody positive dermatomyositis. Orphanet J Rare Dis 2024; 19:170. [PMID: 38637830 PMCID: PMC11027411 DOI: 10.1186/s13023-024-03153-6] [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: 08/13/2023] [Accepted: 03/28/2024] [Indexed: 04/20/2024] Open
Abstract
BACKGROUND Rapid progressive interstitial lung disease (RP-ILD) is the leading cause of anti-melanoma differentiation associated protein 5 antibody positive dermatomyositis (anti-MDA5+DM) related death. Elevated serum B-cell activating factor (BAFF) levels have been implicated in connective tissue diseases associated ILD. Here, we evaluate whether BAFF could be a prognostic biomarker for predicting RP-ILD in anti-MDA5+DM patients. METHODS Serums were collected from 39 patients with anti-MDA5+DM (20 with RP-ILD and 19 with non-RP-ILD), 20 antisynthase syndrome (ASS) patients and 20 healthy controls (HC). BAFF concentration was measured by an enzyme-linked immunosorbent assay. RESULTS Serum BAFF level was higher in anti-MDA5+DM patients than those in ASS patients and HC (3882.32 ± 1880.09 vs. 2540.89 ± 1403.04 and 2486.28 ± 767.97 pg/mL, p = 0.0056 and 0.0038, respectively). Within anti-MDA5+DM groups, RP-ILD patients exhibited higher BAFF concentration than non-RP-ILD group (4549.78 ± 1839.97 vs. 3297.28 ± 1794.69 pg/mL, p = 0.04). The BAFF concentration was positively correlated with levels of C-reactive protein (CRP), dehydrogenase (LDH) and cytokeratin (CK) in anti-MDA5+DM patients (r = 0.350, p = 0.035; r = 0.393, p = 0.016; r = 0.518, p = 0.001; respectively). The best cut-off value of BAFF concentration was 2971.5 pg/mL by ROC curve (AUC area = 0.690, p = 0.045) and BAFF > 2971.5 pg/mL was an independent risk factor for RP-ILD using multivariate analysis (OR = 9.389, 95% CI = 1.609-54.769; p = 0.013). CONCLUSIONS Serum BAFF could be a useful prognostic biomarker for early detecting RP-ILD risk in anti-MDA5+DM patients.
Collapse
Affiliation(s)
- Yumeng Shi
- Department of Rheumatology, The First Affiliated Hospital of Nanjing Medical University, No. 300, Guangzhou Road, Gulou District, Nanjing, 210029, China
| | - Hanxiao You
- Department of Rheumatology, The First Affiliated Hospital of Nanjing Medical University, No. 300, Guangzhou Road, Gulou District, Nanjing, 210029, China
| | - Chang Liu
- Department of Rheumatology, The First Affiliated Hospital of Nanjing Medical University, No. 300, Guangzhou Road, Gulou District, Nanjing, 210029, China
| | - Yulu Qiu
- Department of Rheumatology, The First Affiliated Hospital of Nanjing Medical University, No. 300, Guangzhou Road, Gulou District, Nanjing, 210029, China
| | - Chengyin Lv
- Department of Rheumatology, The First Affiliated Hospital of Nanjing Medical University, No. 300, Guangzhou Road, Gulou District, Nanjing, 210029, China
| | - Yujing Zhu
- Department of Rheumatology, The First Affiliated Hospital of Nanjing Medical University, No. 300, Guangzhou Road, Gulou District, Nanjing, 210029, China
| | - Lingxiao Xu
- Department of Rheumatology, The First Affiliated Hospital of Nanjing Medical University, No. 300, Guangzhou Road, Gulou District, Nanjing, 210029, China
| | - Fang Wang
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Miaojia Zhang
- Department of Rheumatology, The First Affiliated Hospital of Nanjing Medical University, No. 300, Guangzhou Road, Gulou District, Nanjing, 210029, China.
| | - Wenfeng Tan
- Department of Rheumatology, The First Affiliated Hospital of Nanjing Medical University, No. 300, Guangzhou Road, Gulou District, Nanjing, 210029, China.
| |
Collapse
|
7
|
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
|
8
|
Liakouli V, Ciancio A, Del Galdo F, Giacomelli R, Ciccia F. Systemic sclerosis interstitial lung disease: unmet needs and potential solutions. Nat Rev Rheumatol 2024; 20:21-32. [PMID: 37923862 DOI: 10.1038/s41584-023-01044-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/06/2023] [Indexed: 11/06/2023]
Abstract
Systemic sclerosis (SSc), or scleroderma, is a rare, complex, systemic autoimmune disease of unknown aetiology, characterized by high morbidity and mortality often resulting from cardiopulmonary complications such as interstitial lung disease and pulmonary arterial hypertension. Despite substantial progress in unravelling the pathways involved in the pathogenesis of SSc and the increasing number of therapeutic targets tested in clinical trials, there is still no cure for this disease, although several proposed treatments might limit the involvement of specific organs, thereby slowing the natural history of the disease. A specific focus of recent research has been to address the plethora of unmet needs regarding the global management of SSc-related interstitial lung disease, including its pathogenesis, early diagnosis, risk stratification of patients, appropriate treatment regimens and monitoring of treatment response, as well as the definition of progression and predictors of progression and mortality. More refined stratification of patients on the basis of clinical features, molecular signatures, identification of subpopulations with distinct clinical trajectories and implementation of outcome measures for future clinical trials could also improve therapeutic management strategies, helping to avoid poor outcomes related to lung involvement.
Collapse
Affiliation(s)
- Vasiliki Liakouli
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy.
| | - Antonio Ciancio
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Francesco Del Galdo
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, UK
- Scleroderma Programme, NIHR Biomedical Research Centre, Leeds Teaching Hospital Trusts, Leeds, UK
| | - Roberto Giacomelli
- Rheumatology and Immunology Unit, Department of Medicine, University of Rome Campus Biomedico, Rome, Italy
| | - Francesco Ciccia
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
| |
Collapse
|
9
|
Ramos MJ, Lui AJ, Hollern DP. The Evolving Landscape of B Cells in Cancer Metastasis. Cancer Res 2023; 83:3835-3845. [PMID: 37815800 PMCID: PMC10914383 DOI: 10.1158/0008-5472.can-23-0620] [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: 02/23/2023] [Revised: 06/28/2023] [Accepted: 10/05/2023] [Indexed: 10/11/2023]
Abstract
Metastasis is the leading cause of cancer mortality. Functional and clinical studies have documented diverse B-cell and antibody responses in cancer metastasis. The presence of B cells in tumor microenvironments and metastatic sites has been associated with diverse effects that can promote or inhibit metastasis. Specifically, B cells can contribute to the spread of cancer cells by enhancing tumor cell motility, invasion, angiogenesis, lymphangiogenesis, and extracellular matrix remodeling. Moreover, they can promote metastatic colonization by triggering pathogenic immunoglobulin responses and recruiting immune suppressive cells. Contrastingly, B cells can also exhibit antimetastatic effects. For example, they aid in enhanced antigen presentation, which helps activate immune responses against cancer cells. In addition, B cells play a crucial role in preventing the dissemination of metastatic cells from the primary tumor and secrete antibodies that can aid in tumor recognition. Here, we review the complex roles of B cells in metastasis, delineating the heterogeneity of B-cell activity and subtypes by metastatic site, antibody class, antigen (if known), and molecular phenotype. These important attributes of B cells emphasize the need for a deeper understanding and characterization of B-cell phenotypes to define their effects in metastasis.
Collapse
Affiliation(s)
- Monika J. Ramos
- Salk Institute for Biological Sciences
- The University of California San Diego School of Biological Sciences
| | - Asona J. Lui
- Salk Institute for Biological Sciences
- Radiation Medicine and Applied Sciences, The University of California School of Medicine
| | - Daniel P. Hollern
- Salk Institute for Biological Sciences
- The University of California San Diego School of Biological Sciences
- Radiation Medicine and Applied Sciences, The University of California School of Medicine
- NOMIS Center for Immunobiology and Microbial Pathogenesis
| |
Collapse
|
10
|
Leask A, Naik A, Stratton RJ. Back to the future: targeting the extracellular matrix to treat systemic sclerosis. Nat Rev Rheumatol 2023; 19:713-723. [PMID: 37789119 DOI: 10.1038/s41584-023-01032-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/31/2023] [Indexed: 10/05/2023]
Abstract
Fibrosis is the excessive deposition of a stable extracellular matrix (ECM); fibrotic tissue is composed principally of highly crosslinked type I collagen and highly contractile myofibroblasts. Systemic sclerosis (SSc) is a multisystem autoimmune connective tissue disease characterized by skin and organ fibrosis. The fibrotic process has been recognized in SSc for >40 years, but drugs with demonstrable efficacy against SSc fibrosis in ameliorating the lung involvement have only recently been identified. Unfortunately, these treatments are ineffective at improving the skin score in patients with SSc. Previous clinical trials in SSc have largely focused on the cross-purposing of anti-inflammatory drugs and the use of immunosuppressive drugs from the transplantation field, which address inflammatory and/or autoimmune processes. Limited examination has taken place of specific anti-fibrotic agents developed through their ability to directly target the ECM in SSc by, for example, alleviating the persistent matrix stiffness and mechanotransduction that might be required for both the initiation and maintenance of fibrosis, including in SSc. However, because of the importance of the ECM in the SSc phenotype, attempts have now been made to identify drugs that specifically target the ECM, including some drugs that are currently under consideration for the treatment of cancer.
Collapse
Affiliation(s)
- Andrew Leask
- College of Dentistry, University of Saskatchewan, Saskatoon, Saskatchewan, Canada.
| | - Angha Naik
- College of Dentistry, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Richard J Stratton
- Centre for Rheumatology and Connective Tissue Diseases, UCL Division of Medicine, London, UK
| |
Collapse
|
11
|
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
|
12
|
Rezazadeh M, Jokar MH, Mehrnaz Aghili S, Mirfeizi Z, Mahmoudi M, Morovatdar N, Hashemzadeh K. Association between levels of serum and urinary B cell-activating factor and systemic lupus erythematosus disease activity. Arch Rheumatol 2023; 38:429-440. [PMID: 38046245 PMCID: PMC10689013 DOI: 10.46497/archrheumatol.2023.9549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 09/15/2022] [Indexed: 12/05/2023] Open
Abstract
Objectives This study investigated the correlation between serum and urinary B cell-activating factor (BAFF) levels and systemic lupus erythematosus (SLE) disease activity. Patients and methods This case-control study was conducted with 87 participants between December 2020 and September 2021. Sixty-two SLE patients who fulfilled the eligibility criteria were enrolled. SLE patients were categorized into active (n=34) and inactive (n=28) groups based on their Systemic Lupus Erythematosus Disease Activity Index 2000 (SLEDAI-2K) scores. The control group consisted of 25 healthy subjects. Serum and urine samples were collected for the measurement of BAFF levels. Finally, the relationship between these variables and SLE disease activity was investigated. Results The mean age of active (SLEDAI-2K >4) and inactive (SLEDAI-2K ≤4) SLE patients and healthy individuals were 32.8±7.8, 32.5±6.8, and 31.7±7.8 years, respectively (p=0.62). The median serum BAFF (s-BAFF) and urinary BAFF (u-BAFF) in active lupus patients (10.4 [2.3] ng/mL and 8.2 [3.7] ng/mL, respectively) were significantly higher than in inactive lupus patients (6 (7.1) ng/mL and 1.7 (4.7) ng/mL, respectively; p<0.001) and the control group (3 (3.7) ng/mL and 1.6 (2.2) ng/mL, respectively; p<0.001). However, s-BAFF (p=0.07) and u-BAFF (p=0.43) did not significantly differ between the inactive group and the control group. A significant positive correlation was observed between s-BAFF (r=0.41 and p=0.001) and u-BAFF (r=0.78 and p<0.001) levels and the SLEDAI-2K score. Conclusion There is a significant positive correlation between serum and urinary BAFF levels and SLE disease activity. Furthermore, significantly higher levels of s-BAFF and u-BAFF have been observed in patients with active lupus compared to inactive and healthy subjects, indicating a possible role for BAFF in the pathogenesis of SLE disease activity.
Collapse
Affiliation(s)
- Maryam Rezazadeh
- Mashhad University of Medical Sciences, Rheumatic Diseases Research Center, Mashhad, Iran
| | - Mohammad Hasan Jokar
- Mashhad University of Medical Sciences, Rheumatic Diseases Research Center, Mashhad, Iran
| | - Seyedeh Mehrnaz Aghili
- Division of Rheumatology, Allergy and Immunology, Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Zahra Mirfeizi
- Mashhad University of Medical Sciences, Rheumatic Diseases Research Center, Mashhad, Iran
| | - Mahmoud Mahmoudi
- Mashhad University of Medical Sciences, Bu-Ali Research Institute, Mashhad, Iran
| | - Negar Morovatdar
- Mashhad University of Medical Sciences, Clinical Research Unit, Mashhad, Iran
| | - Kamila Hashemzadeh
- Mashhad University of Medical Sciences, Rheumatic Diseases Research Center, Mashhad, Iran
| |
Collapse
|
13
|
Zhang Y, Yang Y, Gao X, Gao W, Zhang L. Research progress on mesenchymal stem cells and their exosomes in systemic sclerosis. Front Pharmacol 2023; 14:1263839. [PMID: 37693906 PMCID: PMC10485262 DOI: 10.3389/fphar.2023.1263839] [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: 07/20/2023] [Accepted: 08/15/2023] [Indexed: 09/12/2023] Open
Abstract
Systemic sclerosis (SSc) is a connective tissue disease with an unknown etiology. Clinically, it is characterized by localized or diffuse skin thickening and fibrosis. The pathogenesis of SSc includes microvascular injury, autoimmune-mediated inflammation, and fibroblast activation. These processes interact and contribute to the diverse clinicopathology and presentation of SSc. Given the limited effectiveness and substantial side effects of traditional treatments, the treatment strategy for SSc has several disadvantages. Mesenchymal stem cells (MSCs) are expected to serve as effective treatment options owing to their significant immunomodulatory, antifibrotic, and pro-angiogenic effects. Exosomes, secreted by MSCs via paracrine signaling, mirror the effect of MSCs as well as offer the benefit of targeted delivery, minimal immunogenicity, robust reparability, good safety and stability, and easy storage and transport. This enables them to circumvent the limitations of the MSCs. When using exosomes, it is crucial to consider preparation methods, quality standards, and suitable drug delivery systems, among other technical issues. Therefore, this review aims to summarize the latest research progress on MSCs and exosomes in SSc, offering novel ideas for treating SSc.
Collapse
Affiliation(s)
| | | | | | | | - Liyun Zhang
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
| |
Collapse
|
14
|
Steele H, Cheng J, Willicut A, Dell G, Breckenridge J, Culberson E, Ghastine A, Tardif V, Herro R. TNF superfamily control of tissue remodeling and fibrosis. Front Immunol 2023; 14:1219907. [PMID: 37465675 PMCID: PMC10351606 DOI: 10.3389/fimmu.2023.1219907] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 06/13/2023] [Indexed: 07/20/2023] Open
Abstract
Fibrosis is the result of extracellular matrix protein deposition and remains a leading cause of death in USA. Despite major advances in recent years, there remains an unmet need to develop therapeutic options that can effectively degrade or reverse fibrosis. The tumor necrosis super family (TNFSF) members, previously studied for their roles in inflammation and cell death, now represent attractive therapeutic targets for fibrotic diseases. In this review, we will summarize select TNFSF and their involvement in fibrosis of the lungs, the heart, the skin, the gastrointestinal tract, the kidney, and the liver. We will emphasize their direct activity on epithelial cells, fibroblasts, and smooth muscle cells. We will further report on major clinical trials targeting these ligands. Whether in isolation or in combination with other anti-TNFSF member or treatment, targeting this superfamily remains key to improve efficacy and selectivity of currently available therapies for fibrosis.
Collapse
Affiliation(s)
- Hope Steele
- Division of Immunobiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States
- University of Cincinnati, Cincinnati, OH, United States
| | - Jason Cheng
- Division of Immunobiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States
| | - Ashley Willicut
- University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Garrison Dell
- Division of Immunobiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States
- University of Cincinnati, Cincinnati, OH, United States
| | - Joey Breckenridge
- Division of Immunobiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States
- University of Cincinnati, Cincinnati, OH, United States
| | - Erica Culberson
- University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Andrew Ghastine
- Division of Immunobiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States
| | - Virginie Tardif
- Normandy University, UniRouen, Institut National de la Santé et de la Recherche Médicale (INSERM), UMR1096 (EnVI Laboratory), Rouen, France
| | - Rana Herro
- Division of Immunobiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States
- Department of Pediatrics, University of Cincinnati, Cincinnati, OH, United States
| |
Collapse
|
15
|
Wortel CM, Liem SI, van Leeuwen NM, Boonstra M, Fehres CM, Stöger L, Huizinga TW, Toes RE, De Vries-Bouwstra J, Scherer HU. Anti-topoisomerase, but not anti-centromere B cell responses in systemic sclerosis display active, Ig-secreting cells associated with lung fibrosis. RMD Open 2023; 9:e003148. [PMID: 37507206 PMCID: PMC10387632 DOI: 10.1136/rmdopen-2023-003148] [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: 03/10/2023] [Accepted: 07/10/2023] [Indexed: 07/30/2023] Open
Abstract
OBJECTIVES Almost all patients with systemic sclerosis (SSc) harbour autoantibodies. Anti-topoisomerase antibodies (ATA) and anti-centromere antibodies (ACA) are most prevalent and associate with distinct clinical phenotypes. B cell responses underlying these phenotypes are ill-defined. To understand how B cell autoreactivity and disease pathology connect, we determined phenotypic and functional characteristics of autoreactive B cells in ATA-positive and ACA-positive patients. METHODS Levels and isotypes of autoantibodies secreted by ex vivo cultured peripheral blood mononuclear cells from patients with ATA-positive (n=22) and ACA-positive (n=20) SSc were determined. Antibody secreting cells (ASCs) were isolated by cell sorting and cultured separately. Correlations were studied between the degree of spontaneous autoantibody production and the presence and degree of interstitial lung disease (ILD). RESULTS Circulating B cells secreting either ATA-immunoglobulin G (IgG) or ACA-IgG on stimulation was readily detectable in patients. The ATA response, but not the ACA response, showed additional secretion of autoreactive IgA. ATA-IgG and ATA-IgA were also secreted spontaneously. Additional cell sorting confirmed the presence of ATA-secreting plasmablasts. The degree of spontaneous ATA-secretion was higher in patients with ILD than in those without (p<0.001) and correlated with the degree of pulmonary fibrosis (p<0.001). CONCLUSION In contrast to ACA-positive patients, ATA-positive patients show signs of recent activation of the B cell response that hallmarks this disease. The degree of activation correlates with the presence and severity of ILD, the most deleterious disease manifestation. This could explain differential responsiveness to B cell depleting therapy. The abundant and spontaneous secretion of ATA-IgG and ATA-IgA may point toward a continuously activating trigger.
Collapse
Affiliation(s)
- Corrie M Wortel
- Department of Rheumatology, Leiden University Medical Center, Leiden, The Netherlands
| | - Sophie Ie Liem
- Department of Rheumatology, Leiden University Medical Center, Leiden, The Netherlands
| | - Nina M van Leeuwen
- Department of Rheumatology, Leiden University Medical Center, Leiden, The Netherlands
| | - Maaike Boonstra
- Department of Rheumatology, Leiden University Medical Center, Leiden, The Netherlands
| | - Cynthia M Fehres
- Department of Rheumatology, Leiden University Medical Center, Leiden, The Netherlands
| | - Lauran Stöger
- Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Tom Wj Huizinga
- Department of Rheumatology, Leiden University Medical Center, Leiden, The Netherlands
| | - René Em Toes
- Department of Rheumatology, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Hans U Scherer
- Department of Rheumatology, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
16
|
Bell S, Young JA, List EO, Basu R, Geitgey DK, Lach G, Lee K, Swegan D, Caggiano LJ, Okada S, Kopchick JJ, Berryman DE. Increased Fibrosis in White Adipose Tissue of Male and Female bGH Transgenic Mice Appears Independent of TGF-β Action. Endocrinology 2023; 164:7069260. [PMID: 36869769 DOI: 10.1210/endocr/bqad038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 02/21/2023] [Accepted: 02/24/2023] [Indexed: 03/05/2023]
Abstract
Fibrosis is a pathological state caused by excess deposition of extracellular matrix proteins in a tissue. Male bovine growth hormone (bGH) transgenic mice experience metabolic dysfunction with a marked decrease in lifespan and with increased fibrosis in several tissues including white adipose tissue (WAT), which is more pronounced in the subcutaneous (Sc) depot. The current study expanded on these initial findings to evaluate WAT fibrosis in female bGH mice and the role of transforming growth factor (TGF)-β in the development of WAT fibrosis. Our findings established that female bGH mice, like males, experience a depot-dependent increase in WAT fibrosis, and bGH mice of both sexes have elevated circulating levels of several markers of collagen turnover. Using various methods, TGF-β signaling was found unchanged or decreased-as opposed to an expected increase-despite the marked fibrosis in WAT of bGH mice. However, acute GH treatments in vivo, in vitro, or ex vivo did elicit a modest increase in TGF-β signaling in some experimental systems. Finally, single nucleus RNA sequencing confirmed no perturbation in TGF-β or its receptor gene expression in any WAT cell subpopulations of Sc bGH WAT; however, a striking increase in B lymphocyte infiltration in bGH WAT was observed. Overall, these data suggest that bGH WAT fibrosis is independent of the action of TGF-β and reveals an intriguing shift in immune cells in bGH WAT that should be further explored considering the increasing importance of B cell-mediated WAT fibrosis and pathology.
Collapse
Affiliation(s)
- Stephen Bell
- Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701, USA
- Edison Biotechnology Institute, Ohio University, Athens, OH 45701, USA
| | - Jonathan A Young
- Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701, USA
- Edison Biotechnology Institute, Ohio University, Athens, OH 45701, USA
| | - Edward O List
- Edison Biotechnology Institute, Ohio University, Athens, OH 45701, USA
| | - Reetobrata Basu
- Edison Biotechnology Institute, Ohio University, Athens, OH 45701, USA
| | | | - Grace Lach
- Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701, USA
- Edison Biotechnology Institute, Ohio University, Athens, OH 45701, USA
| | - Kevin Lee
- Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701, USA
- Diabetes Institute, Ohio University, Athens, OH 45701, USA
| | - Deborah Swegan
- College of Arts and Sciences, Ohio University, Athens, OH 45701, USA
| | | | - Shigeru Okada
- Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701, USA
- Edison Biotechnology Institute, Ohio University, Athens, OH 45701, USA
| | - John J Kopchick
- Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701, USA
- Edison Biotechnology Institute, Ohio University, Athens, OH 45701, USA
- Diabetes Institute, Ohio University, Athens, OH 45701, USA
| | - Darlene E Berryman
- Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701, USA
- Edison Biotechnology Institute, Ohio University, Athens, OH 45701, USA
- Diabetes Institute, Ohio University, Athens, OH 45701, USA
| |
Collapse
|
17
|
Kanemitsu-Okada K, Abe M, Nakamura Y, Miyake T, Watanabe T, Yoshida O, Koizumi Y, Hirooka M, Tokumoto Y, Matsuura B, Koizumi M, Hiasa Y. Role of B Cell-Activating Factor in Fibrosis Progression in a Murine Model of Non-Alcoholic Steatohepatitis. Int J Mol Sci 2023; 24:ijms24032509. [PMID: 36768854 PMCID: PMC9916461 DOI: 10.3390/ijms24032509] [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: 11/29/2022] [Revised: 01/23/2023] [Accepted: 01/25/2023] [Indexed: 01/31/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is the most prevalent chronic liver disease all over the world. Therapeutic strategies targeting its multidirectional pathways are required. Particularly, fibrosis is closely associated with its prognosis. We previously found that B cell-activating factor (BAFF) is associated with severity of NAFLD. Here, we determined the direct in vivo role of BAFF in the development of liver fibrosis. Histological and biochemical analyses were performed using wild-type and BAFF-deficient mice. We established a murine model of non-alcoholic steatohepatitis (NASH) using carbon tetrachloride injection accompanied by high-fat/high-cholesterol diet feeding. Additionally, in vitro analysis using mouse macrophage-like cell line RAW264.7 and primary hepatic stellate cells was performed. Hepatic steatosis and inflammation, and most importantly, the progression of liver fibrosis, were ameliorated in BAFF-deficient mice compared to those wild-type mice in our model. Additionally, BAFF deficiency reduced the number of CD11c+ M1-type macrophages in the liver. Moreover, BAFF stimulated RAW264.7 cells to secrete nitric oxide and tumor necrosis factor α, which drove the activation of hepatic stellate cells. This indicates that BAFF plays a crucial role in NASH development and may be a promising therapeutic target for NASH.
Collapse
|
18
|
Esnault S, Jarjour NN. Development of Adaptive Immunity and Its Role in Lung Remodeling. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1426:287-351. [PMID: 37464127 DOI: 10.1007/978-3-031-32259-4_14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/20/2023]
Abstract
Asthma is characterized by airflow limitations resulting from bronchial closure, which can be either reversible or fixed due to changes in airway tissue composition and structure, also known as remodeling. Airway remodeling is defined as increased presence of mucins-producing epithelial cells, increased thickness of airway smooth muscle cells, angiogenesis, increased number and activation state of fibroblasts, and extracellular matrix (ECM) deposition. Airway inflammation is believed to be the main cause of the development of airway remodeling in asthma. In this chapter, we will review the development of the adaptive immune response and the impact of its mediators and cells on the elements defining airway remodeling in asthma.
Collapse
|
19
|
Pinyowiwat P, Rutnin S, Chanprapaph K. Scleroderma-Like Lupus Panniculitis: A Case Report and Literature Review. Clin Cosmet Investig Dermatol 2023; 16:995-1001. [PMID: 37065790 PMCID: PMC10103709 DOI: 10.2147/ccid.s405553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 04/04/2023] [Indexed: 04/18/2023]
Abstract
Sclerodermic or scleroderma-like lupus erythematosus panniculitis (SLEP) shares both clinical and histopathological features between lupus panniculitis and localized scleroderma. It is exceedingly rare. We herein report a case of SLEP manifested with a solitary, firm-to-hard, erythematous plaque in an Asian woman. This patient responded well to intralesional corticosteroid and antimalarials. We have reviewed the pathogenesis of fibrosis in patients with chronic cutaneous lupus erythematosus as well as documented cases of SLEP in the literature.
Collapse
Affiliation(s)
- Prinpat Pinyowiwat
- Division of Dermatology, Department of Medicine, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Suthinee Rutnin
- Division of Dermatology, Department of Medicine, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Kumutnart Chanprapaph
- Division of Dermatology, Department of Medicine, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
- Correspondence: Kumutnart Chanprapaph, Division of Dermatology, Department of Medicine, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, 270 Rama IV Road, Ratchatewi, Bangkok, 10400, Thailand, Tel +662-201-1141, Fax +662-201-1211, Email
| |
Collapse
|
20
|
Kim S, Park HJ, Lee SI. The Microbiome in Systemic Sclerosis: Pathophysiology and Therapeutic Potential. Int J Mol Sci 2022; 23:ijms232416154. [PMID: 36555792 PMCID: PMC9853331 DOI: 10.3390/ijms232416154] [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: 09/30/2022] [Revised: 12/06/2022] [Accepted: 12/15/2022] [Indexed: 12/23/2022] Open
Abstract
Systemic sclerosis (SSc), also known as scleroderma, is an autoimmune disease with unknown etiology characterized by multi-organ fibrosis. Despite substantial investigation on SSc-related cellular and molecular mechanisms, effective therapies are still lacking. The skin, lungs, and gut are the most affected organs in SSc, which act as physical barriers and constantly communicate with colonized microbiota. Recent reports have documented a unique microbiome signature, which may be the pathogenic trigger or driver of SSc. Since gut microbiota influences the efficacy and toxicity of oral drugs, evaluating drug-microbiota interactions has become an area of interest in disease treatment. The existing evidence highlights the potential of the microbial challenge as a novel therapeutic option in SSc. In this review, we have summarized the current knowledge about molecular mechanisms of SSc and highlighted the underlying role of the microbiome in SSc pathogenesis. We have also discussed the latest therapeutic interventions using microbiomes in SSc, including drug-microbiota interactions and animal disease models. This review aims to elucidate the pathophysiological connection and therapeutic potential of the microbiome in SSc. Insights into the microbiome will significantly improve our understanding of etiopathogenesis and developing therapeutics for SSc.
Collapse
|
21
|
Fang D, Chen B, Lescoat A, Khanna D, Mu R. Immune cell dysregulation as a mediator of fibrosis in systemic sclerosis. Nat Rev Rheumatol 2022; 18:683-693. [DOI: 10.1038/s41584-022-00864-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/04/2022] [Indexed: 11/11/2022]
|
22
|
Chancheewa B, Asawanonda P, Noppakun N, Kumtornrut C. Myofibroblasts, B Cells, and Mast Cells in Different Types of Long-Standing Acne Scars. Skin Appendage Disord 2022; 8:469-475. [PMID: 36407643 PMCID: PMC9672862 DOI: 10.1159/000524566] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Accepted: 04/06/2022] [Indexed: 09/08/2023] Open
Abstract
Acne scars are classified into various types based on their appearances, ranging from hypertrophic to atrophic. Abnormal wound healing processes play an important role in the pathogenesis of scars; however, the exact mechanisms involved in various scar appearances have still not been elucidated. In this study, we used immunofluorescence and immunohistochemistry techniques to detect the presence of myofibroblasts, B cells, and mast cells in each type of acne scar persisting longer than 6 months. We found the highest density of myofibroblasts in hypertrophic acne scars, while in the other atrophic scars, we could not identify any myofibroblast-rich areas in our specimens. B-cell infiltration was mild and found in only 23% (4/17) of all acne scar specimens. Interestingly, mast cells were identified in all specimens, ranging from minimal to high density, and a high number of mast cells in acne scars were associated with obesity. In conclusion, myofibroblasts are abundant only in hypertrophic acne scars, and mast cells, but not B cells, might play an important role in the pathogenesis of long-standing acne scars.
Collapse
Affiliation(s)
- Bussabong Chancheewa
- Division of Dermatology, Department of Medicine, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Pravit Asawanonda
- Division of Dermatology, Department of Medicine, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Nopadon Noppakun
- Division of Dermatology, Department of Medicine, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Chanat Kumtornrut
- Division of Dermatology, Department of Medicine, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Division of Dermatology, Department of Medicine, King Chulalongkorn Memorial Hospital, The Thai Red Cross Society, Bangkok, Thailand
| |
Collapse
|
23
|
Hajam EY, Panikulam P, Chu CC, Jayaprakash H, Majumdar A, Jamora C. The expanding impact of T-regs in the skin. Front Immunol 2022; 13:983700. [PMID: 36189219 PMCID: PMC9521603 DOI: 10.3389/fimmu.2022.983700] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 08/26/2022] [Indexed: 11/29/2022] Open
Abstract
As the interface between the body and the environment, the skin functions as the physical barrier against external pathogens and toxic agents. In addition, the skin is an immunologically active organ with a plethora of resident adaptive and innate immune cells, as well as effector molecules that provide another layer of protection in the form of an immune barrier. A major subpopulation of these immune cells are the Foxp3 expressing CD4 T cells or regulatory T cells (T-regs). The canonical function of T-regs is to keep other immune cells in check during homeostasis or to dissipate a robust inflammatory response following pathogen clearance or wound healing. Interestingly, recent data has uncovered unconventional roles that vary between different tissues and we will highlight the emerging non-lymphoid functions of cutaneous T-regs. In light of the novel functions of other immune cells that are routinely being discovered in the skin, their regulation by T-regs implies that T-regs have executive control over a broad swath of biological activities in both homeostasis and disease. The blossoming list of non-inflammatory functions, whether direct or indirect, suggests that the role of T-regs in a regenerative organ such as the skin will be a field ripe for discovery for decades to come.
Collapse
Affiliation(s)
- Edries Yousaf Hajam
- IFOM ETS- The AIRC Institute of Molecular Oncology Joint Research Laboratory, Centre for Inflammation and Tissue Homeostasis, Institute for Stem Cell Science and Regenerative Medicine (inStem), Bangalore, Karnataka, India
- School of Chemical and Biotechnology, Shanmugha Arts, Science, Technology and Research Academy (SASTRA) University, Thanjavur, Tamil Nadu, India
| | - Patricia Panikulam
- IFOM ETS- The AIRC Institute of Molecular Oncology Joint Research Laboratory, Centre for Inflammation and Tissue Homeostasis, Institute for Stem Cell Science and Regenerative Medicine (inStem), Bangalore, Karnataka, India
| | | | - Haarshadri Jayaprakash
- IFOM ETS- The AIRC Institute of Molecular Oncology Joint Research Laboratory, Centre for Inflammation and Tissue Homeostasis, Institute for Stem Cell Science and Regenerative Medicine (inStem), Bangalore, Karnataka, India
| | | | - Colin Jamora
- IFOM ETS- The AIRC Institute of Molecular Oncology Joint Research Laboratory, Centre for Inflammation and Tissue Homeostasis, Institute for Stem Cell Science and Regenerative Medicine (inStem), Bangalore, Karnataka, India
| |
Collapse
|
24
|
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
|
25
|
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: 23] [Impact Index Per Article: 11.5] [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
|
26
|
Ledoult E, Jendoubi M, Collet A, Guerrier T, Largy A, Speca S, Vivier S, Bray F, Figeac M, Hachulla E, Labalette M, Leprêtre F, Sebda S, Sanges S, Rolando C, Sobanski V, Dubucquoi S, Launay D. Simple gene signature to assess murine fibroblast polarization. Sci Rep 2022; 12:11748. [PMID: 35817787 PMCID: PMC9273630 DOI: 10.1038/s41598-022-15640-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 06/27/2022] [Indexed: 11/09/2022] Open
Abstract
We provide an original multi-stage approach identifying a gene signature to assess murine fibroblast polarization. Prototypic polarizations (inflammatory/fibrotic) were induced by seeded mouse embryonic fibroblasts (MEFs) with TNFα or TGFß1, respectively. The transcriptomic and proteomic profiles were obtained by RNA microarray and LC-MS/MS. Gene Ontology and pathways analysis were performed among the differentially expressed genes (DEGs) and proteins (DEPs). Balb/c mice underwent daily intradermal injections of HOCl (or PBS) as an experimental murine model of inflammation-mediated fibrosis in a time-dependent manner. As results, 1456 and 2215 DEGs, and 289 and 233 DEPs were respectively found in MEFs in response to TNFα or TGFß1, respectively. Among the most significant pathways, we combined 26 representative genes to encompass the proinflammatory and profibrotic polarizations of fibroblasts. Based on principal component analysis, this signature deciphered baseline state, proinflammatory polarization, and profibrotic polarization as accurately as RNA microarray and LC-MS/MS did. Then, we assessed the gene signature on dermal fibroblasts isolated from the experimental murine model. We observed a proinflammatory polarization at day 7, and a mixture of a proinflammatory and profibrotic polarizations at day 42 in line with histological findings. Our approach provides a small-size and convenient gene signature to assess murine fibroblast polarization.
Collapse
Affiliation(s)
- Emmanuel Ledoult
- Inserm, U1286, 4Ème Étage Centre, Place Verdun, 59000, Lille, France. .,INFINITE - Institute for Translational Research in Inflammation, Univ. Lille, 59000, 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, 59000, Lille, France.
| | - Manel Jendoubi
- Inserm, U1286, 4Ème Étage Centre, Place Verdun, 59000, Lille, France.,INFINITE - Institute for Translational Research in Inflammation, Univ. Lille, 59000, Lille, France
| | - Aurore Collet
- Inserm, U1286, 4Ème Étage Centre, Place Verdun, 59000, Lille, France.,INFINITE - Institute for Translational Research in Inflammation, Univ. Lille, 59000, 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, 59000, Lille, France
| | - Thomas Guerrier
- Inserm, U1286, 4Ème Étage Centre, Place Verdun, 59000, Lille, France.,INFINITE - Institute for Translational Research in Inflammation, Univ. Lille, 59000, Lille, France.,Laboratoire d'Immunologie, Pole Biologie et d'Anatomopathologie, CHU Lille, 59000, Lille, France
| | - Alexis Largy
- INFINITE - Institute for Translational Research in Inflammation, Univ. Lille, 59000, Lille, France
| | - Silvia Speca
- Inserm, U1286, 4Ème Étage Centre, Place Verdun, 59000, Lille, France.,INFINITE - Institute for Translational Research in Inflammation, Univ. Lille, 59000, Lille, France
| | - Solange Vivier
- Inserm, U1286, 4Ème Étage Centre, Place Verdun, 59000, Lille, France.,INFINITE - Institute for Translational Research in Inflammation, Univ. Lille, 59000, Lille, France
| | - Fabrice Bray
- CNRS, UAR 3290 - MSAP - Miniaturisation Pour La Synthèse, Univ. Lille, l'Analyse et la Protéomique, 59000, Lille, France
| | - Martin Figeac
- CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, US 41 - UMS 2014 - PLBS, Univ. Lille, 59000, Lille, France
| | - Eric Hachulla
- Inserm, U1286, 4Ème Étage Centre, Place Verdun, 59000, Lille, France.,INFINITE - Institute for Translational Research in Inflammation, Univ. Lille, 59000, 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, 59000, Lille, France
| | - Myriam Labalette
- Inserm, U1286, 4Ème Étage Centre, Place Verdun, 59000, Lille, France.,INFINITE - Institute for Translational Research in Inflammation, Univ. Lille, 59000, Lille, France.,Laboratoire d'Immunologie, Pole Biologie et d'Anatomopathologie, CHU Lille, 59000, Lille, France
| | - Frédéric Leprêtre
- CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, US 41 - UMS 2014 - PLBS, Univ. Lille, 59000, Lille, France
| | - Shéhérazade Sebda
- CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, US 41 - UMS 2014 - PLBS, Univ. Lille, 59000, Lille, France
| | - Sébastien Sanges
- Inserm, U1286, 4Ème Étage Centre, Place Verdun, 59000, Lille, France.,INFINITE - Institute for Translational Research in Inflammation, Univ. Lille, 59000, 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, 59000, Lille, France
| | - Christian Rolando
- CNRS, UAR 3290 - MSAP - Miniaturisation Pour La Synthèse, Univ. Lille, l'Analyse et la Protéomique, 59000, Lille, France.,Shrieking Sixties, 1-3 Allée Lavoisier, 59650, Villeneuve-d'Ascq, France
| | - Vincent Sobanski
- Inserm, U1286, 4Ème Étage Centre, Place Verdun, 59000, Lille, France.,INFINITE - Institute for Translational Research in Inflammation, Univ. Lille, 59000, 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, 59000, Lille, France.,Institut Universitaire de France (IUF), 75005, Paris, France
| | - Sylvain Dubucquoi
- Inserm, U1286, 4Ème Étage Centre, Place Verdun, 59000, Lille, France.,INFINITE - Institute for Translational Research in Inflammation, Univ. Lille, 59000, Lille, France.,Laboratoire d'Immunologie, Pole Biologie et d'Anatomopathologie, CHU Lille, 59000, Lille, France
| | - David Launay
- Inserm, U1286, 4Ème Étage Centre, Place Verdun, 59000, Lille, France.,INFINITE - Institute for Translational Research in Inflammation, Univ. Lille, 59000, 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, 59000, Lille, France
| |
Collapse
|
27
|
Liossis SNC, Staveri C. The Role of B Cells in Scleroderma Lung Disease Pathogenesis. Front Med (Lausanne) 2022; 9:936182. [PMID: 35860745 PMCID: PMC9289134 DOI: 10.3389/fmed.2022.936182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Accepted: 06/14/2022] [Indexed: 11/13/2022] Open
Abstract
Systemic sclerosis (SSc) is a chronic, autoimmune, multisystem disease characterized by tissue fibrosis that, apart from the skin, may affect the lungs among other organs. B cells have been found in tissue lymphocytic infiltrates; in the lungs are encountered in lymphoid aggregates. The abnormal and hyperreactive B cell in SSc may initiate and perpetuate the fibrotic process via incompletely understood mechanisms. Studies in animal models of SSc have demonstrated that B cell dysregulation is an early event in disease pathogenesis. Functional disturbances of BCR signaling such as decreased inhibitory CD22 signal transduction or augmented CD19-mediated signaling result in prolonged B cell activation. Antagonism of BAFF, a cytokine known for his central role in B cell survival and maturation, not only suppresses the production of fibrogenic cytokines such as IL-6 and IL-10, but also amplifies antifibrogenic cytokine secretion such as IFN-γ and it finally contributes to skin fibrosis attenuation. B cells subsets in SSc patients display several abnormalities. Naïve B cells are increased, in contrast to switched memory B cells that are not only decreased but also activated. Disturbances in the expression of molecules that are involved in B cell tuning have also been described. Interestingly, a distinct B cell population characterized by anergy and exhaustion has been found to be increased in patients with SSc-ILD. Another B cell subset, the CD30+GM-Beff, is capable to differentiate monocytes to dendritic cells and is increased in SSc patients with ILD. Of note, patients with SSc-ILD exhibit increased expression of the inhibitory receptor FcγRIIB on naïve and double negative B cells aiming perhaps to counterbalance the abnormal B cell activation. Studies of B cell targeted treatments have demonstrated promising clinical efficacy. Therefore, B cell eliminating therapies could be integrated into the therapeutic armamentarium of patients suffering from SSc-ILD aiming to at least stabilize the fibrotic lung process.
Collapse
Affiliation(s)
- Stamatis-Nick C. Liossis
- Division of Rheumatology, Department of Internal Medicine, Patras University Hospital, Patras, Greece
- Division of Rheumatology, Department of Internal Medicine, University of Patras Medical School, Patras, Greece
- *Correspondence: Stamatis-Nick C. Liossis
| | - Chrysanthi Staveri
- Division of Rheumatology, Department of Internal Medicine, Patras University Hospital, Patras, Greece
| |
Collapse
|
28
|
Fabricating a Novel Three-Dimensional Skin Model Using Silica Nonwoven Fabrics (SNF). APPLIED SCIENCES-BASEL 2022. [DOI: 10.3390/app12136537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Silica nonwoven fabrics (SNF) prepared using electrospinning have high biocompatibility, thermal stability, and porosity that allows growing three-dimensional culture of cells. In this study, we used SNF to construct a three-dimensional artificial skin model consisting of epidermal and dermal layers with immortalized and primary human cell lines, creating a novel model that minimizes tissue shrinkage. As a result, SNF dermal/epidermal models have enhanced functions in the basement membrane, whereas Collagen dermal/epidermal models have advantages in keratinization and barrier functions. The SNF dermal/epidermal model with mechanical strength formed a basement membrane mimicking structure, suggesting the construction of a stable skin model. Next, we constructed three-dimensional skin models consisting of SNF and collagen. In the combination models, the expression of genes in the basement membrane was significantly increased compared with that in the Collagen dermal/epidermal model, and the gene for keratinization was increased compared with that in the SNF dermal/epidermal model. We believe that the combination model can be a biomimetic model that takes advantage of both SNF and collagen and can be applied to various basic research. Our new skin model is expected to be an alternative method for skin testing to improve the shrinkage of the collagen matrix gel.
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: 26] [Impact Index Per Article: 13.0] [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
|
Rosendahl AH, Schönborn K, Krieg T. Pathophysiology of systemic sclerosis (scleroderma). Kaohsiung J Med Sci 2022; 38:187-195. [PMID: 35234358 DOI: 10.1002/kjm2.12505] [Citation(s) in RCA: 54] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Accepted: 01/18/2022] [Indexed: 12/14/2022] Open
Abstract
Systemic sclerosis (scleroderma) is an autoimmune-triggered chronic fibrosing disease that affects the skin and many other organs. Its pathophysiology is complex and involves an early endothelial damage, an inflammatory infiltrate and a resulting fibrotic reaction. Based on a predisposing genetic background, an altered balance of the acquired and the innate immune system leads to the release of many cytokines and chemokines as well as autoantibodies, which induce the activation of fibroblasts with the formation of myofibroblasts and the deposition of a stiff and rigid connective tissue. A curative treatment is still not available but remarkable progress has been made in the management of organ complications. In addition, several breakthroughs in the pathophysiology have led to new therapeutic concepts. Based on these, many new compounds have been developed during the last years, which target these different pathways and offer specific therapeutic approaches.
Collapse
Affiliation(s)
- Ann-Helen Rosendahl
- Translational Matrix Biology, University of Cologne, Medical Faculty, Cologne, Germany
| | - Katrin Schönborn
- Translational Matrix Biology, University of Cologne, Medical Faculty, Cologne, Germany.,Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - Thomas Krieg
- Translational Matrix Biology, University of Cologne, Medical Faculty, Cologne, Germany.,Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany.,Department of Dermatology, University Hospital of Cologne, Cologne, Germany.,Cologne Excellence Cluster on Cellular Stress Responses in Ageing-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| |
Collapse
|
31
|
Moog MT, Hinze C, Bormann T, Aschenbrenner F, Knudsen L, DeLuca DS, Jonigk D, Neubert L, Welte T, Gauldie J, Kolb M, Maus UA. B Cells Are Not Involved in the Regulation of Adenoviral TGF-β1- or Bleomycin-Induced Lung Fibrosis in Mice. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 208:1259-1271. [PMID: 35149532 DOI: 10.4049/jimmunol.2100767] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Accepted: 12/27/2021] [Indexed: 06/14/2023]
Abstract
Idiopathic pulmonary fibrosis (IPF) is an irreversible, age-related diffuse parenchymal lung disease of poorly defined etiology. Many patients with IPF demonstrate distinctive lymphocytic interstitial infiltrations within remodeled lung tissue with uncertain pathogenetic relevance. Histopathological examination of explant lung tissue of patients with IPF revealed accentuated lymphoplasmacellular accumulations in close vicinity to, or even infiltrating, remodeled lung tissue. Similarly, we found significant accumulations of B cells interfused with T cells within remodeled lung tissue in two murine models of adenoviral TGF-β1 or bleomycin (BLM)-induced lung fibrosis. Such B cell accumulations coincided with significantly increased lung collagen deposition, lung histopathology, and worsened lung function in wild-type (WT) mice. Surprisingly, B cell-deficient µMT knockout mice exhibited similar lung tissue remodeling and worsened lung function upon either AdTGF-β1 or BLM as for WT mice. Comparative transcriptomic profiling of sorted B cells collected from lungs of AdTGF-β1- and BLM-exposed WT mice identified a large set of commonly regulated genes, but with significant enrichment observed for Gene Ontology terms apparently not related to lung fibrogenesis. Collectively, although we observed B cell accumulations in lungs of IPF patients as well as two experimental models of lung fibrosis, comparative profiling of characteristic features of lung fibrosis between WT and B cell-deficient mice did not support a major involvement of B cells in lung fibrogenesis in mice.
Collapse
Affiliation(s)
- Marie T Moog
- Division of Experimental Pneumology, Hannover Medical School, Hannover, Germany
| | - Christopher Hinze
- Division of Experimental Pneumology, Hannover Medical School, Hannover, Germany
| | - Tina Bormann
- Division of Experimental Pneumology, Hannover Medical School, Hannover, Germany
| | | | - Lars Knudsen
- Institute of Functional and Applied Anatomy, Hannover Medical School, Hannover, Germany
| | - David S DeLuca
- German Center for Lung Research, partner site Biomedical Research in Endstage and Obstructive Lung Disease Hanover, Hannover, Germany
| | - Danny Jonigk
- German Center for Lung Research, partner site Biomedical Research in Endstage and Obstructive Lung Disease Hanover, Hannover, Germany
- Institute of Pathology, Hannover Medical School, Hannover, Germany
| | - Lavinia Neubert
- German Center for Lung Research, partner site Biomedical Research in Endstage and Obstructive Lung Disease Hanover, Hannover, Germany
- Institute of Pathology, Hannover Medical School, Hannover, Germany
| | - Tobias Welte
- German Center for Lung Research, partner site Biomedical Research in Endstage and Obstructive Lung Disease Hanover, Hannover, Germany
- Clinic for Pneumology, Hannover Medical School, Hannover, Germany; and
| | - Jack Gauldie
- Department of Medicine, Pathology, and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Martin Kolb
- Department of Medicine, Pathology, and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Ulrich A Maus
- Division of Experimental Pneumology, Hannover Medical School, Hannover, Germany;
- German Center for Lung Research, partner site Biomedical Research in Endstage and Obstructive Lung Disease Hanover, Hannover, Germany
| |
Collapse
|
32
|
Hinchcliff M, Garcia-Milian R, Di Donato S, Dill K, Bundschuh E, Galdo FD. Cellular and Molecular Diversity in Scleroderma. Semin Immunol 2021; 58:101648. [PMID: 35940960 DOI: 10.1016/j.smim.2022.101648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
With the increasing armamentarium of high-throughput tools available at manageable cost, it is attractive and informative to determine the molecular underpinnings of patient heterogeneity in systemic sclerosis (SSc). Given the highly variable clinical outcomes of patients labelled with the same diagnosis, unravelling the cellular and molecular basis of disease heterogeneity will be crucial to predicting disease risk, stratifying management and ultimately informing a patient-centered precision medicine approach. Herein, we summarise the findings of the past several years in the fields of genomics, transcriptomics, and proteomics that contribute to unraveling the cellular and molecular heterogeneity of SSc. Expansion of these findings and their routine integration with quantitative analysis of histopathology and imaging studies into clinical care promise to inform a scientifically driven patient-centred personalized medicine approach to SSc in the near future.
Collapse
Affiliation(s)
- Monique Hinchcliff
- Yale School of Medicine, Department of Internal Medicine, Section of Rheumatology, Allergy & Immunology, USA.
| | | | - Stefano Di Donato
- Raynaud's and Scleroderma Programme, Leeds Institute of Rheumatic and Musculoskeletal Medicine and NIHR Biomedical Research Centre, University of Leeds, UK
| | | | - Elizabeth Bundschuh
- Yale School of Medicine, Department of Internal Medicine, Section of Rheumatology, Allergy & Immunology, USA
| | - Francesco Del Galdo
- Raynaud's and Scleroderma Programme, Leeds Institute of Rheumatic and Musculoskeletal Medicine and NIHR Biomedical Research Centre, University of Leeds, UK.
| |
Collapse
|
33
|
Mouchet N, Vu N, Turlin B, Rioux-Leclercq N, Jouneau S, Samson M, Amiot L. HLA-G Is Widely Expressed by Mast Cells in Regions of Organ Fibrosis in the Liver, Lung and Kidney. Int J Mol Sci 2021; 22:ijms222212490. [PMID: 34830373 PMCID: PMC8618089 DOI: 10.3390/ijms222212490] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 10/18/2021] [Accepted: 11/03/2021] [Indexed: 01/14/2023] Open
Abstract
We previously demonstrated that mast cells expressing HLA-G are associated with regions of hepatitis C virus-induced liver fibrosis. Here, we aimed to determine whether HLA-G expression in mast cells is specific to viral etiology, the liver, or to the general process of fibrosis. We enumerated HLA-G+ cells and mast cells by the immunohistochemistry of (i) liver blocks from 41 cases of alcoholic cirrhosis, (ii) 10 of idiopathic pulmonary fibrosis (IPF), and (iii) 10 of renal fibrosis. The nature of the HLA-G+ cells was specified by multiplex immunofluorescence using software. More than half of all HLA-G+ cells were mast cells in fibrotic areas of alcoholic cirrhosis and IPF. In the kidneys, subjected to fibrosis, the HLA-G+ cells were indeed mast cells but could not be counted. Moreover, in certain cases of the liver and lung, we observed a number of cellular nodes, which were secondary or tertiary follicles, in which HLA-G was highly expressed by B lymphocytes. In conclusion, HLA-G+ mast cells could be observed in the fibrotic regions of all organs studied. Previous studies suggest a protective role for HLA-G+ mast cells against inflammation and fibrosis. The observed follicles with B lymphocytes that express HLA-G may also reinforce their antifibrotic role.
Collapse
Affiliation(s)
- Nicolas Mouchet
- Univ Rennes, INSERM, CNRS, Biosit, Core Facility H2P2, F-35000 Rennes, France;
- France BioImaging, Biogenouest, IBISA, F-35000 Rennes, France
| | - Nicolas Vu
- Univ Rennes, INSERM, EHESP, IRSET (Institut de Recherche en Santé, Environnement et Travail)-UMR_S 1085, F-35000 Rennes, France; (N.V.); (M.S.)
| | - Bruno Turlin
- Univ Rennes, CHU Rennes, F-35000 Rennes, France;
| | - Nathalie Rioux-Leclercq
- Univ Rennes, CHU Rennes, INSERM, EHESP, IRSET (Institut de Recherche en Santé, Environnement et Travail)-UMR_S 1085, F-35000 Rennes, France; (N.R.-L.); (S.J.)
| | - Stéphane Jouneau
- Univ Rennes, CHU Rennes, INSERM, EHESP, IRSET (Institut de Recherche en Santé, Environnement et Travail)-UMR_S 1085, F-35000 Rennes, France; (N.R.-L.); (S.J.)
| | - Michel Samson
- Univ Rennes, INSERM, EHESP, IRSET (Institut de Recherche en Santé, Environnement et Travail)-UMR_S 1085, F-35000 Rennes, France; (N.V.); (M.S.)
| | - Laurence Amiot
- Univ Rennes, CHU Rennes, INSERM, EHESP, IRSET (Institut de Recherche en Santé, Environnement et Travail)-UMR_S 1085, F-35000 Rennes, France; (N.R.-L.); (S.J.)
- Correspondence: ; Tel.: +(330)-223-23-48-62
| |
Collapse
|
34
|
Abstract
From the clinical standpoint, systemic sclerosis (SSc) is characterized by skin and internal organ fibrosis, diffuse fibroproliferative vascular modifications, and autoimmunity. Clinical presentation and course are highly heterogenous and life expectancy variably affected mostly dependent on lung and heart involvement. SSc touches more women than men with differences in disease severity and environmental exposure. Pathogenetic events originate from altered homeostasis favored by genetic predisposition, environmental cues and a variety of endogenous and exogenous triggers. Epigenetic modifications modulate SSc pathogenesis which strikingly associate profound immune-inflammatory dysregulation, abnormal endothelial cell behavior, and cell trans-differentiation into myofibroblasts. SSc myofibroblasts show enhanced survival and enhanced extracellular matrix deposition presenting altered structure and altered physicochemical properties. Additional cell types of likely pathogenic importance are pericytes, platelets, and keratinocytes in conjunction with their relationship with vessel wall cells and fibroblasts. In SSc, the profibrotic milieu is favored by cell signaling initiated in the one hand by transforming growth factor-beta and related cytokines and in the other hand by innate and adaptive type 2 immune responses. Radical oxygen species and invariant receptors sensing danger participate to altered cell behavior. Conventional and SSc-specific T cell subsets modulate both fibroblasts as well as endothelial cell dysfunction. Beside autoantibodies directed against ubiquitous antigens important for enhanced clinical classification, antigen-specific agonistic autoantibodies may have a pathogenic role. Recent studies based on single-cell RNAseq and multi-omics approaches are revealing unforeseen heterogeneity in SSc cell differentiation and functional states. Advances in system biology applied to the wealth of data generated by unbiased screening are allowing to subgroup patients based on distinct pathogenic mechanisms. Deciphering heterogeneity in pathogenic mechanisms will pave the way to highly needed personalized therapeutic approaches.
Collapse
|
35
|
Beurier P, Ricard L, Eshagh D, Malard F, Siblany L, Fain O, Mohty M, Gaugler B, Mekinian A. TFH cells in systemic sclerosis. J Transl Med 2021; 19:375. [PMID: 34461933 PMCID: PMC8407089 DOI: 10.1186/s12967-021-03049-0] [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: 06/15/2021] [Accepted: 08/20/2021] [Indexed: 12/17/2022] Open
Abstract
Systemic sclerosis is an autoimmune disease characterized by excessive dermal fibrosis with progression to internal organs, vascular impairment and immune dysregulation evidenced by the infiltration of inflammatory cells in affected tissues and the production of auto antibodies. While the pathogenesis remains unclear, several data highlight that T and B cells deregulation is implicated in the disease pathogenesis. Over the last decade, aberrant responses of circulating T follicular helper cells, a subset of CD4 T cells which are able to localise predominantly in the B cell follicles through a high level of chemokine receptor CXCR5 expression are described in pathogenesis of several autoimmune diseases and chronic graft-versus-host-disease. In the present review, we summarized the observed alteration of number and frequency of circulating T follicular helper cells in systemic sclerosis. We described their role in aberrant B cell activation and differentiation though interleukine-21 secretion. We also clarified T follicular helper-like cells involvement in fibrogenesis in both human and mouse model. Finally, because T follicular helper cells are involved in both fibrosis and autoimmune abnormalities in systemic sclerosis patients, we presented the different strategies could be used to target T follicular helper cells in systemic sclerosis, the therapeutic trials currently being carried out and the future perspectives from other auto-immune diseases and graft-versus-host-disease models.
Collapse
Affiliation(s)
- Pauline Beurier
- INSERM UMRs 938, Centre de Recherche Saint-Antoine, AP-HP, Hôpital Saint-Antoine, Service de Médecine Interne and Inflammation-Immunopathology-Biotherapy Department (DHU i2B), Sorbonne Université, 75012, Paris, France.,Sorbonne Université, Paris, France
| | - Laure Ricard
- INSERM UMRs 938, Centre de Recherche Saint-Antoine, AP-HP, Hôpital Saint-Antoine, Service de Médecine Interne and Inflammation-Immunopathology-Biotherapy Department (DHU i2B), Sorbonne Université, 75012, Paris, France.,Sorbonne Université, Paris, France.,Service D'Hématologie Clinique, AP-HP, Hôpital Saint-Antoine, 75012, Paris, France
| | - Deborah Eshagh
- INSERM UMRs 938, Centre de Recherche Saint-Antoine, AP-HP, Hôpital Saint-Antoine, Service de Médecine Interne and Inflammation-Immunopathology-Biotherapy Department (DHU i2B), Sorbonne Université, 75012, Paris, France.,Sorbonne Université, Paris, France
| | - Florent Malard
- INSERM UMRs 938, Centre de Recherche Saint-Antoine, AP-HP, Hôpital Saint-Antoine, Service de Médecine Interne and Inflammation-Immunopathology-Biotherapy Department (DHU i2B), Sorbonne Université, 75012, Paris, France.,Sorbonne Université, Paris, France.,Service D'Hématologie Clinique, AP-HP, Hôpital Saint-Antoine, 75012, Paris, France
| | - Lama Siblany
- INSERM UMRs 938, Centre de Recherche Saint-Antoine, AP-HP, Hôpital Saint-Antoine, Service de Médecine Interne and Inflammation-Immunopathology-Biotherapy Department (DHU i2B), Sorbonne Université, 75012, Paris, France.,Sorbonne Université, Paris, France
| | - Olivier Fain
- INSERM UMRs 938, Centre de Recherche Saint-Antoine, AP-HP, Hôpital Saint-Antoine, Service de Médecine Interne and Inflammation-Immunopathology-Biotherapy Department (DHU i2B), Sorbonne Université, 75012, Paris, France.,Sorbonne Université, Paris, France
| | - Mohamad Mohty
- INSERM UMRs 938, Centre de Recherche Saint-Antoine, AP-HP, Hôpital Saint-Antoine, Service de Médecine Interne and Inflammation-Immunopathology-Biotherapy Department (DHU i2B), Sorbonne Université, 75012, Paris, France.,Sorbonne Université, Paris, France.,Service D'Hématologie Clinique, AP-HP, Hôpital Saint-Antoine, 75012, Paris, France
| | - Béatrice Gaugler
- INSERM UMRs 938, Centre de Recherche Saint-Antoine, AP-HP, Hôpital Saint-Antoine, Service de Médecine Interne and Inflammation-Immunopathology-Biotherapy Department (DHU i2B), Sorbonne Université, 75012, Paris, France
| | - Arsène Mekinian
- INSERM UMRs 938, Centre de Recherche Saint-Antoine, AP-HP, Hôpital Saint-Antoine, Service de Médecine Interne and Inflammation-Immunopathology-Biotherapy Department (DHU i2B), Sorbonne Université, 75012, Paris, France. .,Sorbonne Université, Paris, France. .,Service de Médecine Interne and Inflammation-Immunopathology-Biotherapy Department (DMU 3iD), AP-HP, Hôpital Saint-Antoine, 75012, Paris, France.
| |
Collapse
|
36
|
Ali MF, Egan AM, Shaughnessy GF, Anderson DK, Kottom TJ, Dasari H, Van Keulen VP, Aubry MC, Yi ES, Limper AH, Peikert T, Carmona EM. Antifibrotics Modify B-Cell-induced Fibroblast Migration and Activation in Patients with Idiopathic Pulmonary Fibrosis. Am J Respir Cell Mol Biol 2021; 64:722-733. [PMID: 33689587 DOI: 10.1165/rcmb.2020-0387oc] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
B-cell activation is increasingly linked to numerous fibrotic lung diseases, and it is well known that aggregates of lymphocytes form in the lung of many of these patients. Activation of B-cells by pattern recognition receptors (PRRs) drives the release of inflammatory cytokines, chemokines, and metalloproteases important in the pathophysiology of pulmonary fibrosis. However, the specific mechanisms of B-cell activation in patients with idiopathic pulmonary fibrosis (IPF) are poorly understood. Herein, we have demonstrated that B-cell activation by microbial antigens contributes to the inflammatory and profibrotic milieu seen in patients with IPF. B-cell stimulation by CpG and β-glucan via PRRs resulted in activation of mTOR-dependent and independent pathways. Moreover, we showed that the B-cell-secreted inflammatory milieu is specific to the inducing antigen and causes differential fibroblast migration and activation. B-cell responses to infectious agents and subsequent B-cell-mediated fibroblast activation are modifiable by antifibrotics, but each seems to exert a specific and different effect. These results suggest that, upon PRR activation by microbial antigens, B-cells can contribute to the inflammatory and fibrotic changes seen in patients with IPF, and antifibrotics are able to at least partially reverse these responses.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Eunhee S Yi
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota
| | - Andrew H Limper
- Thoracic Diseases Research Unit.,Division of Pulmonary and Critical Care Medicine
| | - Tobias Peikert
- Thoracic Diseases Research Unit.,Division of Pulmonary and Critical Care Medicine.,Department of Immunology and
| | - Eva M Carmona
- Thoracic Diseases Research Unit.,Division of Pulmonary and Critical Care Medicine.,Department of Immunology and
| |
Collapse
|
37
|
Ly NTM, Ueda-Hayakawa I, Nguyen CTH, Huynh TNM, Kishimoto I, Fujimoto M, Okamoto H. Imbalance toward TFH 1 cells playing a role in aberrant B cell differentiation in systemic sclerosis. Rheumatology (Oxford) 2021; 60:1553-1562. [PMID: 33175976 DOI: 10.1093/rheumatology/keaa669] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 09/07/2020] [Indexed: 11/14/2022] Open
Abstract
OBJECTIVE SSc is a connective tissue disease with multisystem disorder induced by the inflammation and fibrosis following T and B cell abnormalities. Follicular helper CD4+ T (TFH) cells play a crucial role in the formation of germinal centres and specialize in interacting to aid B cell differentiation. We aimed to investigate TFH cells and their subsets to evaluate their involvement with B cell alteration in SSc. METHOD Circulating TFH cells (cTFH), B cells and their subsets were assessed by flow cytometry. The concentration of serum cytokines was measured by cytokine array assay. Immunohistochemistry and IF were performed to evaluate the migration of TFH cells in SSc skin lesions. RESULTS The proportion of cTFH cells did not differ from controls, but their subsets were imbalanced in SSc patients. The frequency of TFH 1 was increased and correlated with ACA titre, serum IgM or CRP levels of patients, and cytokine concentrations of IL-21 and IL-6 that induce B cell differentiation in SSc. cTFH cells from SSc showed activated phenotype with expressing higher cytokine levels compared with controls. The frequency of TFH 17 was also increased, but was not correlated with a high level of Th17 cytokines in patients' sera. Furthermore, infiltration of TFH cells was found in skin lesion of SSc patients. CONCLUSION We here describe an imbalance of cTFH toward TFH 1 that may induce B cell alteration through IL-21 and IL-6 pathways and promote inflammation, contributing to the pathogenesis of SSc disease.
Collapse
Affiliation(s)
- Nhung Thi My Ly
- Department of Dermatology, Kansai Medical University, Hirakata, Japan
- Department of Dermatology and Venereology, University of Medicine and Pharmacy at Ho Chi Minh City, Ho Chi Minh, Vietnam, Japan
| | - Ikuko Ueda-Hayakawa
- Department of Dermatology, Kansai Medical University, Hirakata, Japan
- Department of Dermatology, Osaka University, Suita, Japan
| | - Chuyen Thi Hong Nguyen
- Department of Dermatology, Kansai Medical University, Hirakata, Japan
- Department of Dermatology and Venereology, University of Medicine and Pharmacy at Ho Chi Minh City, Ho Chi Minh, Vietnam, Japan
| | | | - Izumi Kishimoto
- Department of Dermatology, Kansai Medical University, Hirakata, Japan
| | | | - Hiroyuki Okamoto
- Department of Dermatology, Kansai Medical University, Hirakata, Japan
| |
Collapse
|
38
|
Wu Q, Liu Y, Xie Y, Wei S, Liu Y. Identification of Potential ceRNA Network and Patterns of Immune Cell Infiltration in Systemic Sclerosis-Associated Interstitial Lung Disease. Front Cell Dev Biol 2021; 9:622021. [PMID: 34222222 PMCID: PMC8248550 DOI: 10.3389/fcell.2021.622021] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Accepted: 05/26/2021] [Indexed: 02/05/2023] Open
Abstract
Purpose Systemic sclerosis-associated interstitial lung disease (SSc-ILD) is one of the most severe complications of systemic sclerosis (SSc) and is the leading cause of SSc-related deaths. However, the precise pathogenesis of pulmonary fibrosis in SSc-ILD remains unknown. This study aimed to evaluate the competing endogenous RNA (ceRNA) regulatory network and immune cell infiltration patterns in SSc-ILD. Methods One microRNA (miRNA) and three messenger RNA (mRNA) microarray datasets were obtained from the Gene Expression Omnibus (GEO) database. Then, the differentially expressed miRNAs (DEmiRs) and mRNAs (DEMs) between SSc-ILD patients and normal controls were identified, respectively, followed by the prediction of the target genes and target lncRNAs of DEmiRs. The overlapping genes between DEmiRs target genes and DEMs were identified as core mRNAs to construct the ceRNA network. In addition, the “Cell Type Identification by Estimating Relative Subsets of Known RNA Transcripts (CIBERSORT)” algorithm was used to analyze the composition of infiltrating immune cells in lung tissues of SSc-ILD patients and controls, and differentially expressed immune cells were recognized. The correlation between immune cells and core mRNAs was evaluated by Pearson correlation analysis. Results Totally, 42 SSc-ILD lung tissues and 18 normal lung tissues were included in this study. We identified 35 DEmiRs and 142 DEMs and predicted 1,265 target genes of DEmiRs. Then, 9 core mRNAs related to SSc-ILD were recognized, which were the overlapping genes between DEmiRs target genes and DEMs. Meanwhile, 9 DEmiRs related to core mRNAs were identified reversely, and their target lncRNAs were predicted. In total, 9 DEmiRs, 9 core mRNAs, and 51 predicted lncRNAs were integrated to construct the ceRNA regulatory network of SSc-ILD. In addition, 9 types of immune cells were differentially expressed in lung tissues between SSc-ILD patients and controls. Some core mRNAs, such as COL1A1, FOS, and EDN1, were positively or negatively correlated with the number of infiltrating immune cells. Conclusion This is the first comprehensive study to construct the potential ceRNA regulatory network and analyze the composition of infiltrating immune cells in lung tissues of SSc-ILD patients, which improves our understanding of the pathogenesis of SSc-ILD.
Collapse
Affiliation(s)
- Qiuhong Wu
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, China
| | - Yang Liu
- Lung Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Yan Xie
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, China
| | - Shixiong Wei
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, China
| | - Yi Liu
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, China.,Rare Diseases Center, West China Hospital, Sichuan University, Chengdu, China.,Institute of Immunology and Inflammation, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
39
|
B cells as target for immunotherapy in rheumatic diseases - current status. Immunol Lett 2021; 236:12-19. [PMID: 34077805 DOI: 10.1016/j.imlet.2021.05.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Revised: 05/16/2021] [Accepted: 05/25/2021] [Indexed: 01/16/2023]
Abstract
This mini-review is a short overview of different therapeutical strategies targeting B cells in systemic autoimmune rheumatic diseases, mainly: rheumatoid arthritis (RA), systemic lupus erythematosus (SLE) and primary Sjogren Syndrome (pSS). Many strategies and their rationale are discussed in this review: B cells' depletion (anti-CD20, anti-CD22), long-lived plasma cells depletion (anti-CD19, anti-CD27, anti-CD38 and anti-CD138), changing activation of B cells (anti-BAFF) and inhibiting proteasomes in plasma cells (bortezomib). The past successful therapies and less successful are shown, and the possible reasons for failures are discussed.
Collapse
|
40
|
De Pieri A, Korman BD, Jüngel A, Wuertz-Kozak K. Engineering Advanced In Vitro Models of Systemic Sclerosis for Drug Discovery and Development. Adv Biol (Weinh) 2021; 5:e2000168. [PMID: 33852183 PMCID: PMC8717409 DOI: 10.1002/adbi.202000168] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 01/13/2021] [Accepted: 01/19/2021] [Indexed: 12/19/2022]
Abstract
Systemic sclerosis (SSc) is a complex multisystem disease with the highest case-specific mortality among all autoimmune rheumatic diseases, yet without any available curative therapy. Therefore, the development of novel therapeutic antifibrotic strategies that effectively decrease skin and organ fibrosis is needed. Existing animal models are cost-intensive, laborious and do not recapitulate the full spectrum of the disease and thus commonly fail to predict human efficacy. Advanced in vitro models, which closely mimic critical aspects of the pathology, have emerged as valuable platforms to investigate novel pharmaceutical therapies for the treatment of SSc. This review focuses on recent advancements in the development of SSc in vitro models, sheds light onto biological (e.g., growth factors, cytokines, coculture systems), biochemical (e.g., hypoxia, reactive oxygen species) and biophysical (e.g., stiffness, topography, dimensionality) cues that have been utilized for the in vitro recapitulation of the SSc microenvironment, and highlights future perspectives for effective drug discovery and validation.
Collapse
Affiliation(s)
- Andrea De Pieri
- Dr. A. De Pieri, Prof. K. Wuertz-Kozak, Department of Biomedical Engineering, Rochester Institute of Technology (RIT), 106 Lomb Memorial Rd., Rochester, NY, 14623, USA
| | - Benjamin D Korman
- Prof. B. D. Korman, Department of Medicine, Division of Allergy, Immunology and Rheumatology, University of Rochester Medical Center, Rochester, NY, 14623, USA
| | - Astrid Jüngel
- Prof. A. Jüngel, Center of Experimental Rheumatology, University Clinic of Rheumatology, Balgrist University Hospital, University Hospital Zurich, Zurich, 8008, Switzerland
- Prof. A. Jüngel, Department of Physical Medicine and Rheumatology, Balgrist University Hospital, University of Zurich, Zurich, 8008, Switzerland
| | - Karin Wuertz-Kozak
- Dr. A. De Pieri, Prof. K. Wuertz-Kozak, Department of Biomedical Engineering, Rochester Institute of Technology (RIT), 106 Lomb Memorial Rd., Rochester, NY, 14623, USA
- Prof. K. Wuertz-Kozak, Schön Clinic Munich Harlaching, Spine Center, Academic Teaching Hospital and Spine Research Institute of the Paracelsus Medical University Salzburg (Austria), Munich, 81547, Germany
| |
Collapse
|
41
|
Romano E, Rosa I, Fioretto BS, Cerinic MM, Manetti M. The Role of Pro-fibrotic Myofibroblasts in Systemic Sclerosis: from Origin to Therapeutic Targeting. Curr Mol Med 2021; 22:209-239. [PMID: 33823766 DOI: 10.2174/0929867328666210325102749] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 03/02/2021] [Accepted: 03/09/2021] [Indexed: 11/22/2022]
Abstract
Systemic sclerosis (SSc, scleroderma) is a complex connective tissue disorder characterized by multisystem clinical manifestations resulting from immune dysregulation/autoimmunity, vasculopathy and, most notably, progressive fibrosis of the skin and internal organs. In recent years, it has emerged that the main drivers of SSc-related tissue fibrosis are myofibroblasts, a type of mesenchymal cells with both the extracellular matrix-synthesizing features of fibroblasts and the cytoskeletal characteristics of contractile smooth muscle cells. The accumulation and persistent activation of pro-fibrotic myofibroblasts during SSc development and progression result into elevated mechanical stress and reduced matrix plasticity within the affected tissues and may be ascribed to a reduced susceptibility of these cells to pro-apoptotic stimuli, as well as their increased formation from tissue-resident fibroblasts or transition from different cell types. Given the crucial role of myofibroblasts in SSc pathogenesis, finding the way to inhibit myofibroblast differentiation and accumulation by targeting their formation, function and survival may represent an effective approach to hamper the fibrotic process or even halt or reverse established fibrosis. In this review, we discuss the role of myofibroblasts in SSc-related fibrosis, with a special focus on their cellular origin and the signaling pathways implicated in their formation and persistent activation. Furthermore, we provide an overview of potential therapeutic strategies targeting myofibroblasts that may be able to counteract fibrosis in this pathological condition.
Collapse
Affiliation(s)
- Eloisa Romano
- Department of Experimental and Clinical Medicine, Division of Rheumatology, University of Florence, Florence. Italy
| | - Irene Rosa
- Department of Experimental and Clinical Medicine, Division of Rheumatology, University of Florence, Florence. Italy
| | - Bianca Saveria Fioretto
- Department of Experimental and Clinical Medicine, Division of Rheumatology, University of Florence, Florence. Italy
| | - Marco Matucci Cerinic
- Department of Experimental and Clinical Medicine, Division of Rheumatology, University of Florence, Florence. Italy
| | - Mirko Manetti
- Department of Experimental and Clinical Medicine, Section of Anatomy and Histology, University of Florence, Florence. Italy
| |
Collapse
|
42
|
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: 2] [Impact Index Per Article: 0.7] [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
|
43
|
Lima-Júnior JR, Arruda LCM, Gonçalves MS, Dias JBE, Moraes DA, Covas DT, Simões BP, Oliveira MC, Malmegrim KCR. Autologous hematopoietic stem cell transplantation restores the suppressive capacity of regulatory B cells in systemic sclerosis patients. Rheumatology (Oxford) 2021; 60:5538-5548. [PMID: 33724344 DOI: 10.1093/rheumatology/keab257] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Accepted: 03/07/2021] [Indexed: 12/17/2022] Open
Abstract
OBJECTIVES The rationale of autologous hematopoietic stem cell transplantation (AHSCT) for autoimmune diseases is that high-dose immunosuppression eradicates autoreactive T and B cells, and the infused autologous hematopoietic stem cells promote reconstitution of a naive and self-tolerant immune system. The aim of this study was to evaluate the reconstitution of different B cell subsets, both quantitatively and functionally, in systemic sclerosis (SSc) patients treated with AHSCT. METHODS Peripheral blood was harvested from twenty-two SSc patients before transplantation and at 30, 60, 120, 180 and 360 days post-AHSCT. Immunophenotyping of B cell subsets, B cell cytokine production, signaling pathways, and suppressive capacity of regulatory B cells (Bregs) were assessed by flow cytometry. RESULTS Naïve B cell frequencies increased from 60 to 360 days post-AHSCT, compared to pre-transplantation. Conversely, memory B cell frequencies decreased during the same period. Plasma cell frequencies transiently decreased at 60 days post-AHSCT. IL-10-producing Bregs CD19+CD24hiCD38hi and CD19+CD24hiCD27+ frequencies increased at 180 days. Moreover, the phosphorylation of ERK1/2 and p38MAPK proteins increased in B cells reconstituted post-AHSCT. Notably, CD19+CD24hiCD38hi Bregs recovered their ability to suppress production of Th1 cytokines by CD4+ T cells at 360 days post-AHSCT. Finally, IL-6 and TGF-β1-producing B cells decreased following AHSCT. CONCLUSION Taken together, these results suggest improvements in immunoregulatory and anti-fibrotic mechanisms after AHSCT for SSc, which may contribute to reestablishment of self-tolerance and clinical remission.
Collapse
Affiliation(s)
- João R Lima-Júnior
- Graduate Program on Biosciences and Biotechnology, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil.,Center for Cell-based Therapy, Regional Hemotherapy Center of the Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Lucas C M Arruda
- Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| | - Maynara S Gonçalves
- Basic and Applied Immunology Program, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil.,Center for Cell-based Therapy, Regional Hemotherapy Center of the Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Juliana B E Dias
- Division of Clinical Immunology, Department of Internal Medicine, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Daniela A Moraes
- Division of Clinical Immunology, Department of Internal Medicine, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Dimas T Covas
- Center for Cell-based Therapy, Regional Hemotherapy Center of the Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Belinda P Simões
- Center for Cell-based Therapy, Regional Hemotherapy Center of the Ribeirão Preto Medical School, 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.,Division of Clinical Immunology, Department of Internal Medicine, 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
| |
Collapse
|
44
|
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: 3.0] [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
|
45
|
Vacchi C, Manfredi A, Cassone G, Erre GL, Salvarani C, Sebastiani M. Efficacy and safety of rituximab in the treatment of connective tissue disease-related interstitial lung disease. Drugs Context 2021; 10:2020-8-7. [PMID: 33505478 PMCID: PMC7813433 DOI: 10.7573/dic.2020-8-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 11/30/2020] [Indexed: 02/07/2023] Open
Abstract
Interstitial lung disease (ILD) represents a severe pulmonary complication of connective tissue diseases, rheumatoid arthritis (RA), and antineutrophil cytoplasmic antibody-associated vasculitis. Treatment of ILD, mainly based on immunosuppression, remains challenging. Rituximab (RTX), a monoclonal antibody binding to CD20, is considered a valuable therapeutic choice in cases of refractory ILD. Here, we review the available efficacy and safety data on the use of RTX in the treatment of rheumatic disease-related ILD. Despite controversial efficacy data, RTX seems to be able to stabilize or improve ILD related to RA and antisynthetase syndrome and in established and severe ILD complicating systemic sclerosis. Fewer data are available regarding ILD related to Sjögren syndrome, systemic lupus erythematosus, and antineutrophil cytoplasmic antibody-associated vasculitis. To date, few prospective studies are available and randomized trials are still ongoing with the purpose of exploring the role of RTX in this condition, including the supposed relationship between efficacy and ILD radiologic patterns and safety data, up to now derived mainly from RA studies. Despite an overall acceptable safety profile, concerns remain regarding an increased infectious disease risk in patients with ILD as well as possible lung toxicity and the increased rate of immune-mediated reactions in patients with connective tissue diseases. In conclusion, RTX is a relevant therapeutic option for rheumatic disease-related ILD despite the existing uncertainties; ongoing trials are expected to clarify its use.
Collapse
Affiliation(s)
- Caterina Vacchi
- Clinical and Experimental Medicine PhD Program, University of Modena and Reggio Emilia, Modena, Italy
- Chair and Rheumatology Unit, University of Modena and Reggio Emilia, Azienda Ospedaliero-Universitaria Policlinico di Modena, Modena, Italy
| | - Andreina Manfredi
- Chair and Rheumatology Unit, University of Modena and Reggio Emilia, Azienda Ospedaliero-Universitaria Policlinico di Modena, Modena, Italy
| | - Giulia Cassone
- Clinical and Experimental Medicine PhD Program, University of Modena and Reggio Emilia, Modena, Italy
- Chair and Rheumatology Unit, University of Modena and Reggio Emilia, Azienda Ospedaliero-Universitaria Policlinico di Modena, Modena, Italy
- Rheumatology Unit, IRCCS Arcispedale Santa Maria Nuova, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Gian Luca Erre
- Rheumatology Unit, Azienda Ospedaliero-Universitaria di Sassari, Sassari, Italy
| | - Carlo Salvarani
- Chair and Rheumatology Unit, University of Modena and Reggio Emilia, Azienda Ospedaliero-Universitaria Policlinico di Modena, Modena, Italy
- Rheumatology Unit, IRCCS Arcispedale Santa Maria Nuova, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Marco Sebastiani
- Chair and Rheumatology Unit, University of Modena and Reggio Emilia, Azienda Ospedaliero-Universitaria Policlinico di Modena, Modena, Italy
| |
Collapse
|
46
|
Rossi D, Sciascia S, Cecchi I, Saracco M, Montabone E, Modena V, Pellerito R, Carignola R, Roccatello D. A 3-Year Observational Study of Patients with Progressive Systemic Sclerosis Treated with an Intensified B Lymphocyte Depletion Protocol: Clinical and Immunological Response. J Clin Med 2021; 10:E292. [PMID: 33466837 PMCID: PMC7830314 DOI: 10.3390/jcm10020292] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 01/03/2021] [Accepted: 01/06/2021] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND B-cells have been suggested to play a role in the pathogenesis of systemic sclerosis (SSc), representing, therefore, a potential therapeutic target. OBJECTIVES We aimed at investigating the 36-month outcomes of 20 SSc patients who underwent an intensified B-depletion therapy (IBCDT) scheme, including both Rituximab (RTX) and cyclophosphamide (CYC). METHODS Data from 20 severe patients (18 females and 2 males, mean age 66.7 ± 11.0 years) with diffuse SSc (anti-topoisomerase I antibody in 95%) patients with multiorgan involvement including interstitial lung disease (ILD) treated with an IBCDT were prospectively collected. IBCDT comprehended: RTX 375 mg/m2 administered for four weekly doses (on days 1, 8, 15, and 22), followed by two additional doses after 30 and 60 days, in addition to two administrations of 10 mg/kg of intravenous CYC plus three methylprednisolone pulses (15 mg/kg) and subsequently followed by oral prednisone rapidly tapered to low minimum dosage of 5 mg daily. In addition, 10 patients with more severe functional respiratory impairment at baseline were also treated with RTX 500 mg every 4 months during the first year and two times a year during the second and the third year. RESULTS After 36 months of follow-up, we recorded significant amelioration in N-terminal-pro-brain natriuretic peptide (NT-proBNP) levels (mean 385.4 ± 517 pg/mL at baseline to 279 ± 543 after 36 months). In addition, a significant radiological improvement of ILD in 20% of patients (4/20) and a radiological stabilization with no sign of progression of interstitial involvement in 13/20 (65%) were documented. A total of 3 out of 20 (15%) patients experienced a worsening of the ILD. No patient showed further decrease in functional respiratory parameters, including forced vital capacity, forced expiratory volume in one second, and mean values of diffusing capacity for carbon monoxide Moreover, no patient showed any change in the ejection fraction and pulmonary artery pressure when comparing values at baseline and after 24 and 36 months of observation. No severe infection, renal flare, RTX-related side effects were observed. No patient died. CONCLUSIONS Our findings support that the IBCDT was well tolerated and might be a promising therapeutic option for the management of SSc, especially in those subjects with multiorgan involvement that includes ILD.
Collapse
Affiliation(s)
- Daniela Rossi
- Center of Research of Rheumatology, Nephrology and Rare Diseases, Coordinating Center of the Interregional Network of Rare Diseases of Piedmont and Aosta Valley, SCDU CMID-Nephrology and Dialysis G. Bosco Hospital and University of Turin, 10154 Turin, Italy; (D.R.); (S.S.); (I.C.); (V.M.)
| | - Savino Sciascia
- Center of Research of Rheumatology, Nephrology and Rare Diseases, Coordinating Center of the Interregional Network of Rare Diseases of Piedmont and Aosta Valley, SCDU CMID-Nephrology and Dialysis G. Bosco Hospital and University of Turin, 10154 Turin, Italy; (D.R.); (S.S.); (I.C.); (V.M.)
| | - Irene Cecchi
- Center of Research of Rheumatology, Nephrology and Rare Diseases, Coordinating Center of the Interregional Network of Rare Diseases of Piedmont and Aosta Valley, SCDU CMID-Nephrology and Dialysis G. Bosco Hospital and University of Turin, 10154 Turin, Italy; (D.R.); (S.S.); (I.C.); (V.M.)
| | - Marta Saracco
- Rheumatology Unit, Mauriziano Umberto I Hospital, 10128 Turin, Italy; (M.S.); (R.P.)
| | - Erika Montabone
- Internal Medicine, San Luigi Gonzaga Hospital Orbassano, 10043 Turin, Italy; (E.M.); (R.C.)
| | - Vittorio Modena
- Center of Research of Rheumatology, Nephrology and Rare Diseases, Coordinating Center of the Interregional Network of Rare Diseases of Piedmont and Aosta Valley, SCDU CMID-Nephrology and Dialysis G. Bosco Hospital and University of Turin, 10154 Turin, Italy; (D.R.); (S.S.); (I.C.); (V.M.)
| | - Raffaele Pellerito
- Rheumatology Unit, Mauriziano Umberto I Hospital, 10128 Turin, Italy; (M.S.); (R.P.)
| | - Renato Carignola
- Internal Medicine, San Luigi Gonzaga Hospital Orbassano, 10043 Turin, Italy; (E.M.); (R.C.)
| | - Dario Roccatello
- Center of Research of Rheumatology, Nephrology and Rare Diseases, Coordinating Center of the Interregional Network of Rare Diseases of Piedmont and Aosta Valley, SCDU CMID-Nephrology and Dialysis G. Bosco Hospital and University of Turin, 10154 Turin, Italy; (D.R.); (S.S.); (I.C.); (V.M.)
| |
Collapse
|
47
|
Varrica C, Dias HS, Reis C, Carvalheiro M, Simões S. Targeted delivery in scleroderma fibrosis. Autoimmun Rev 2020; 20:102730. [PMID: 33338593 DOI: 10.1016/j.autrev.2020.102730] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Accepted: 10/02/2020] [Indexed: 12/29/2022]
Abstract
Systemic sclerosis (SSc) is considered one of the most challenging and difficult to treat among rheumatic disorders, due to its severity, multiorgan manifestation and different outcomes. It manifests fibrosis in different organs, mostly in skin and lungs. The skin fibrosis expression is considered the first sign of the disease and usually it is followed by internal organ fibrosis. An aberrant immune system activation seems to relate to the expression of the disease, but even environmental influences and dysregulation of many molecules signalling pathways are involved in the development of the disease. Current therapies are limited and characterized by multiple side effects: systemic route is the elective administration route, which decreases patient adherence to the therapy, as they are often already bothered by pain and disfigurement. Treatments available are organ-based, originally indicated for other conditions and there is no therapy available to reduce the fibroblast population size within existing fibrotic lesions. Disease-modifying therapies or immunomodulatory agents that are highly effective in other rheumatic diseases have shown disappointing results in SSc. There are thus no standardized and effective treatments for this disease, and there are even unanswered questions related to the insurgence of the pathology and all the mechanisms involved. An ideal approach could be considered "targeted therapy" that will be an increasingly attainable objective insofar as our understanding of the disease improves. The advantages in identifying the molecule and the signalling pathways involved in the pathology have helped to find some novel compounds for the therapy of scleroderma fibrosis or following innovative uses for already-approved drugs, corroborated by many clinical studies.
Collapse
Affiliation(s)
- Carla Varrica
- University of Pavia, Corso Strada Nuova, 65, 27100 Pavia, Italy
| | - Helena Sofia Dias
- Faculty of Pharmacy, Universidade de Lisboa, Avenida Professor Gama Pinto, 1649-003 Lisboa, Portugal
| | - Catarina Reis
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Avenida Professor Gama Pinto, 1649-003 Lisboa, Portugal; IBEB, Biophysics and Biomedical Engineering, Faculty of Sciences, Universidade de Lisboa, Campo Grande, 1649-016 Lisboa, Portugal
| | - Manuela Carvalheiro
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Avenida Professor Gama Pinto, 1649-003 Lisboa, Portugal
| | - Sandra Simões
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Avenida Professor Gama Pinto, 1649-003 Lisboa, Portugal.
| |
Collapse
|
48
|
Möckel T, Basta F, Weinmann-Menke J, Schwarting A. B cell activating factor (BAFF): Structure, functions, autoimmunity and clinical implications in Systemic Lupus Erythematosus (SLE). Autoimmun Rev 2020; 20:102736. [PMID: 33333233 DOI: 10.1016/j.autrev.2020.102736] [Citation(s) in RCA: 88] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Accepted: 08/28/2020] [Indexed: 12/30/2022]
Abstract
The B cell activating factor (BAFF), or B lymphocyte stimulator (BLyS), is a B cell survival factor which supports autoreactive B cells and prevents their deletion. BAFF expression is closely linked with autoimmunity and is enhanced by genetic alterations and viral infections. Furthermore, BAFF seems to be involved in adipogenesis, atherosclerosis, neuro-inflammatory processes and ischemia reperfusion (I/R) injury. BAFF is commonly overexpressed in Systemic Lupus Erythematosus (SLE) and strongly involved in the pathogenesis of the disease. The relationship between BAFF levels, disease activity and damage accrual in SLE is controversial, but growing evidence is emerging on its role in renal involvement. Belimumab, a biologic BAFF inhibitor, has been the first biologic agent licensed for SLE therapy so far. As Rituximab (RTX) has been shown to increase BAFF levels following B cell depletion, the combination therapy of RTX plus belimumab (being evaluated in two RCT) seems to be a valuable option for several clinical scenarios. In this review we will highlight the growing body of evidence of immune and non-immune related BAFF expression in experimental and clinical settings.
Collapse
Affiliation(s)
- Tamara Möckel
- Department of Internal Medicine I, Division of Rheumatology and Clinical Immunology, University Medical Center of the Johannes Gutenberg University, Mainz, Germany.
| | - Fabio Basta
- Acura Rheumatology Center Rhineland Palatinate, Bad Kreuznach, Germany
| | - Julia Weinmann-Menke
- Department of Internal Medicine I, Division of Nephrology, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Andreas Schwarting
- Department of Internal Medicine I, Division of Rheumatology and Clinical Immunology, University Medical Center of the Johannes Gutenberg University, Mainz, Germany; Acura Rheumatology Center Rhineland Palatinate, Bad Kreuznach, Germany
| |
Collapse
|
49
|
Riley LA, Merryman WD. Cadherin-11 and cardiac fibrosis: A common target for a common pathology. Cell Signal 2020; 78:109876. [PMID: 33285242 DOI: 10.1016/j.cellsig.2020.109876] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 12/01/2020] [Accepted: 12/02/2020] [Indexed: 02/06/2023]
Abstract
Cardiac fibrosis represents an enormous health concern as it is prevalent in nearly every form of cardiovascular disease, the leading cause of death worldwide. Fibrosis is characterized by the activation of fibroblasts into myofibroblasts, a contractile cell type that secretes significant amounts of extracellular matrix components; however, the onset of this condition is also due to persistent inflammation and the cellular responses to a changing mechanical environment. In this review, we provide an overview of the pro-fibrotic, pro-inflammatory, and biomechanical mechanisms that lead to cardiac fibrosis in cardiovascular diseases. We then discuss cadherin-11, an intercellular adhesion protein present on both myofibroblasts and inflammatory cells, as a potential link for all three of the fibrotic mechanisms. Since experimentally blocking cadherin-11 dimerization prevents fibrotic diseases including cardiac fibrosis, understanding how this protein can be targeted for therapeutic use could lead to better treatments for patients with heart disease.
Collapse
Affiliation(s)
- Lance A Riley
- Department of Biomedical Engineering, Vanderbilt University, USA
| | - W David Merryman
- Department of Biomedical Engineering, Vanderbilt University, USA.
| |
Collapse
|
50
|
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: 12] [Impact Index Per Article: 3.0] [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
|