1
|
Chepy A, Collet A, Launay D, Dubucquoi S, Sobanski V. Autoantibodies in systemic sclerosis: From disease bystanders to pathogenic players. J Transl Autoimmun 2025; 10:100272. [PMID: 39917316 PMCID: PMC11799969 DOI: 10.1016/j.jtauto.2025.100272] [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: 12/01/2024] [Revised: 01/15/2025] [Accepted: 01/20/2025] [Indexed: 02/09/2025] Open
Abstract
Autoantibodies (Aab) are recognized as key indicators in the diagnosis, classification, and monitoring of systemic autoimmune diseases (AID). Recent studies have expanded knowledge through the discovery of new antigenic targets, advanced methods for measuring Aab levels, and understanding their possible pathogenic roles in AID. This narrative review uses systemic sclerosis (SSc) as an example to highlight the importance of Aab associated with HEp-2 immunofluorescence assay positivity (traditionally referred as antinuclear antibodies [ANA]), exploring recent developments in the field. Firstly, we outline the various types of ANA found in SSc and their links with specific disease features. Newly discovered antibodies shed light on SSc cases where Aab had previously gone unidentified. Secondly, we emphasize the necessity for novel quantitative techniques to track Aab levels over time by gathering data regarding the timing of Aab occurrence relative to SSc symptoms and the relationships between Aab concentrations and disease severity. Finally, we discuss the experimental findings suggesting a potential direct role of Aab in the development of SSc. The advancements surrounding Aab provide insights into new disease mechanisms and may lead to innovative diagnostic and treatment approaches.
Collapse
Affiliation(s)
- Aurélien Chepy
- Univ. Lille, Inserm, CHU Lille, U1286 – INFINITE – Institute for Translational Research in Inflammation, Lille, France
- CHU Lille, Département de Médecine interne et Immunologie Clinique, Centre de Référence des Maladies Auto-immunes Systémiques Rares du Nord et Nord-Ouest, Méditerranée et Guadeloupe (CeRAINOM), Lille, France
| | - Aurore Collet
- Univ. Lille, Inserm, CHU Lille, U1286 – INFINITE – Institute for Translational Research in Inflammation, Lille, France
- CHU Lille, Institut d’Immunologie, Lille, France
| | - David Launay
- Univ. Lille, Inserm, CHU Lille, U1286 – INFINITE – Institute for Translational Research in Inflammation, Lille, France
- CHU Lille, Département de Médecine interne et Immunologie Clinique, Centre de Référence des Maladies Auto-immunes Systémiques Rares du Nord et Nord-Ouest, Méditerranée et Guadeloupe (CeRAINOM), Lille, France
| | - Sylvain Dubucquoi
- Univ. Lille, Inserm, CHU Lille, U1286 – INFINITE – Institute for Translational Research in Inflammation, Lille, France
- CHU Lille, Institut d’Immunologie, Lille, France
| | - Vincent Sobanski
- Univ. Lille, Inserm, CHU Lille, U1286 – INFINITE – Institute for Translational Research in Inflammation, Lille, France
- CHU Lille, Département de Médecine interne et Immunologie Clinique, Centre de Référence des Maladies Auto-immunes Systémiques Rares du Nord et Nord-Ouest, Méditerranée et Guadeloupe (CeRAINOM), Lille, France
- Institut Universitaire de France (IUF), Paris, France
| |
Collapse
|
2
|
Rosen ABI, Sanyal A, Hutchins T, Werner G, Berkowitz JS, Tabib T, Lafyatis R, Jacobe H, Das J, Torok KS. Unique and shared transcriptomic signatures underlying localized scleroderma pathogenesis identified using interpretable machine learning. JCI Insight 2025; 10:e185758. [PMID: 40197368 PMCID: PMC11981619 DOI: 10.1172/jci.insight.185758] [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: 08/12/2024] [Accepted: 02/18/2025] [Indexed: 04/10/2025] Open
Abstract
Using transcriptomic profiling at single-cell resolution, we investigated cell-intrinsic and cell-extrinsic signatures associated with pathogenesis and inflammation-driven fibrosis in both adult and pediatric patients with localized scleroderma (LS). We performed single-cell RNA-Seq on adult and pediatric patients with LS and healthy controls. We then analyzed the single-cell RNA-Seq data using an interpretable factor analysis machine learning framework, significant latent factor interaction discovery and exploration (SLIDE), which moves beyond predictive biomarkers to infer latent factors underlying LS pathophysiology. SLIDE is a recently developed latent factor regression-based framework that comes with rigorous statistical guarantees regarding identifiability of the latent factors, corresponding inference, and FDR control. We found distinct differences in the characteristics and complexity in the molecular signatures between adult and pediatric LS. SLIDE identified cell type-specific determinants of LS associated with age and severity and revealed insights into signaling mechanisms shared between LS and systemic sclerosis (SSc), as well as differences in onset of the disease in the pediatric compared with adult population. Our analyses recapitulate known drivers of LS pathology and identify cellular signaling modules that stratify LS subtypes and define a shared signaling axis with SSc.
Collapse
Affiliation(s)
- Aaron BI Rosen
- Center for Systems Immunology, Departments of Immunology and Computational & Systems Biology
| | | | | | | | - Jacob S. Berkowitz
- Center for Systems Immunology, Departments of Immunology and Computational & Systems Biology
| | - Tracy Tabib
- Center for Systems Immunology, Departments of Immunology and Computational & Systems Biology
| | - Robert Lafyatis
- Division of Rheumatology and Clinical Immunology, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Heidi Jacobe
- Department of Dermatology, University of Texas Southwestern, Dallas, Texas, USA
| | - Jishnu Das
- Center for Systems Immunology, Departments of Immunology and Computational & Systems Biology
| | | |
Collapse
|
3
|
Chepy A, Vivier S, Bray F, Chauvet C, Lescoat A, Elhannani A, Figeac M, Guilbert L, Leprêtre F, Bourel L, Hachulla E, Rolando C, Lecureur V, Dubucquoi S, Launay D, Sobanski V. Immunoglobulins G from Patients with Systemic Sclerosis Modify the Molecular Signatures of Endothelial Cells. RMD Open 2025; 11:e004290. [PMID: 40122572 PMCID: PMC11931898 DOI: 10.1136/rmdopen-2024-004290] [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: 02/29/2024] [Accepted: 01/28/2025] [Indexed: 03/25/2025] Open
Abstract
OBJECTIVE Antinuclear antibodies (ANA) are powerful biomarkers in systemic sclerosis (SSc). Functional antibodies (FA) might be implicated in vasculopathy, in which endothelial cells (EC) are key players. We aimed to explore the effect of purified IgG from patients with SSc on omics signatures of EC and examine the influence of ANA serotypes and FA. METHODS EC were cultured in the presence of purified IgG from patients with SSc, patients with systemic lupus erythematosus (SLE) or healthy controls (HC). EC omics profiles were analysed by liquid chromatography with tandem mass spectrometry (LC-MS/MS) and RNA sequencing. EC proteome induced by IgG from patients with SSc was confirmed with an external validation cohort. RESULTS In the derivation cohort, principal component analysis (PCA) using proteomics data showed three distinct groups of subjects: a first one including mostly anti-topoisomerase-I positive patients (ATA+), a second one including mostly anti-centromere positive patients and a third group comprising anti-RNA polymerase-III positive patients, SLE and HC. In transcriptomics, PCA distinguished one group composed of ATA+patients only from a second group mixing ATA+patients with other individuals. The validation cohort confirmed the existence of two groups of distinct EC proteome profiles and clinical severity in ATA+patients. In both SSc cohorts, no association between FA presence and proteomic profiles was observed. Quantitative proteomics measured the most discriminant proteins in EC exposed to purified IgG. CONCLUSION Purified IgG from patients with SSc can modify EC proteome and transcriptome. The observed changes closely associate with ANA serotype.
Collapse
Affiliation(s)
- Aurélien Chepy
- Univ. Lille, Inserm, CHU Lille, U1286 - INFINITE - Institute for Translational Research in Inflammation, Lille, France
- CHU Lille, Département de Médecine Interne et Immunologie Clinique, Centre de Référence des Maladies Auto-Immunes et Auto-Inflammatoires Rares du Nord, Nord-Ouest, Méditerranée et Guadeloupe (CeRAINOM), Lille, France
| | - Solange Vivier
- Univ. Lille, Inserm, CHU Lille, U1286 - INFINITE - Institute for Translational Research in Inflammation, Lille, France
| | - Fabrice Bray
- Univ. Lille, CNRS, USR 3290 - MSAP - Miniaturisation pour la Synthèse, l'Analyse et la Protéomique, Lille, France
| | - Clément Chauvet
- Univ. Lille, Inserm, CHU Lille, U1286 - INFINITE - Institute for Translational Research in Inflammation, Lille, France
| | - Alain Lescoat
- Service de Médecine Interne et Immunologie Clinique, CHU Rennes, Université Rennes, Rennes, France
- Inserm, EHESP, Irset (Institut de Recherche en Santé, Environnement et Travail) - UMR_S 1085, Rennes, France
| | - Abderrahmane Elhannani
- Univ. Lille, Inserm, CHU Lille, U1286 - INFINITE - Institute for Translational Research in Inflammation, Lille, France
- CHU Lille, Département de Médecine Interne et Immunologie Clinique, Centre de Référence des Maladies Auto-Immunes et Auto-Inflammatoires Rares du Nord, Nord-Ouest, Méditerranée et Guadeloupe (CeRAINOM), Lille, France
| | - Martin Figeac
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, US 41 - UAR 2014 - PLBS, Lille, France
| | - Lucile Guilbert
- Univ. Lille, Inserm, CHU Lille, U1286 - INFINITE - Institute for Translational Research in Inflammation, Lille, France
- CHU Lille, Institut d'Immunologie, Lille, France
| | - Frédéric Leprêtre
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, US 41 - UAR 2014 - PLBS, Lille, France
| | - Louisa Bourel
- Univ. Lille, Inserm, CHU Lille, U1286 - INFINITE - Institute for Translational Research in Inflammation, Lille, France
| | - Eric Hachulla
- Univ. Lille, Inserm, CHU Lille, U1286 - INFINITE - Institute for Translational Research in Inflammation, Lille, France
- CHU Lille, Département de Médecine Interne et Immunologie Clinique, Centre de Référence des Maladies Auto-Immunes et Auto-Inflammatoires Rares du Nord, Nord-Ouest, Méditerranée et Guadeloupe (CeRAINOM), Lille, France
| | - Christian Rolando
- Univ. Lille, CNRS, USR 3290 - MSAP - Miniaturisation pour la Synthèse, l'Analyse et la Protéomique, Lille, France
| | - Valérie Lecureur
- Inserm, EHESP, Irset (Institut de Recherche en Santé, Environnement et Travail) - UMR_S 1085, Rennes, France
| | - Sylvain Dubucquoi
- Univ. Lille, Inserm, CHU Lille, U1286 - INFINITE - Institute for Translational Research in Inflammation, Lille, France
- CHU Lille, Institut d'Immunologie, Lille, France
| | - David Launay
- Univ. Lille, Inserm, CHU Lille, U1286 - INFINITE - Institute for Translational Research in Inflammation, Lille, France
- CHU Lille, Département de Médecine Interne et Immunologie Clinique, Centre de Référence des Maladies Auto-Immunes et Auto-Inflammatoires Rares du Nord, Nord-Ouest, Méditerranée et Guadeloupe (CeRAINOM), Lille, France
| | - Vincent Sobanski
- Univ. Lille, Inserm, CHU Lille, U1286 - INFINITE - Institute for Translational Research in Inflammation, Lille, France
- CHU Lille, Département de Médecine Interne et Immunologie Clinique, Centre de Référence des Maladies Auto-Immunes et Auto-Inflammatoires Rares du Nord, Nord-Ouest, Méditerranée et Guadeloupe (CeRAINOM), Lille, France
- Institut Universitaire de France (IUF), Paris, France
| |
Collapse
|
4
|
Abraham DJ, Black CM, Denton CP, Distler JHW, Domsic R, Feghali-Bostwick C, Gourh P, Hinchcliff M, Kolling F, Kuwana M, Lafyatis R, Landegren U, Mahoney JM, Martin J, Matucci-Cerinic M, McMahan ZH, Mora AL, Mouthon L, Rabinovitch M, Rojas M, Rubin K, Trojanowska M, Varga J, Whitfield ML, Gabrielli A, Krieg T. An international perspective on the future of systemic sclerosis research. Nat Rev Rheumatol 2025; 21:174-187. [PMID: 39953141 DOI: 10.1038/s41584-024-01217-2] [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: 12/20/2024] [Indexed: 02/17/2025]
Abstract
Systemic sclerosis (SSc) remains a challenging and enigmatic systemic autoimmune disease, owing to its complex pathogenesis, clinical and molecular heterogeneity, and the lack of effective disease-modifying treatments. Despite a century of research in SSc, the interconnections among microvascular dysfunction, autoimmune phenomena and tissue fibrosis in SSc remain unclear. The absence of validated biomarkers and reliable animal models complicates diagnosis and treatment, contributing to high morbidity and mortality. Advances in the past 5 years, such as single-cell RNA sequencing, next-generation sequencing, spatial biology, transcriptomics, genomics, proteomics, metabolomics, microbiome profiling and artificial intelligence, offer new avenues for identifying the early pathogenetic events that, once treated, could change the clinical history of SSc. Collaborative global efforts to integrate these approaches are crucial to developing a comprehensive, mechanistic understanding and enabling personalized therapies. Challenges include disease classification, clinical heterogeneity and the establishment of robust biomarkers for disease activity and progression. Innovative clinical trial designs and patient-centred approaches are essential for developing effective treatments. Emerging therapies, including cell-based and fibroblast-targeting treatments, show promise. Global cooperation, standardized protocols and interdisciplinary research are vital for advancing SSc research and improving patient outcomes. The integration of advanced research techniques holds the potential for important breakthroughs in the diagnosis, treatment and care of individuals with SSc.
Collapse
Affiliation(s)
- David J Abraham
- Department of Inflammation and Rare Diseases, UCL Centre for Rheumatology, UCL Division of Medicine, Royal Free Hospital Campus, London, UK.
| | - Carol M Black
- Department of Inflammation and Rare Diseases, UCL Centre for Rheumatology, UCL Division of Medicine, Royal Free Hospital Campus, London, UK
| | - Christopher P Denton
- Department of Inflammation and Rare Diseases, UCL Centre for Rheumatology, UCL Division of Medicine, Royal Free Hospital Campus, London, UK
| | - Jörg H W Distler
- Department of Rheumatology, University Hospital Düsseldorf, Medical Faculty of the Heinrich-Heine University, Düsseldorf, Germany
- Hiller Research Center, University Hospital Düsseldorf, Medical Faculty of Heinrich Heine University, Düsseldorf, Germany
| | - Robyn Domsic
- Division of Rheumatology, Department of Internal Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Carol Feghali-Bostwick
- Department of Medicine, Division of Rheumatology and Immunology, Medical University of South Carolina, Charleston, SC, USA
| | - Pravitt Gourh
- Scleroderma Genomics and Health Disparities Unit, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Monique Hinchcliff
- Division of Rheumatology, Allergy and Immunology, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Fred Kolling
- Geisel School of Medicine at Dartmouth, Hanover, NH, USA
| | - Masataka Kuwana
- Department of Allergy and Rheumatology. Nippon Medical School Graduate School of Medicine, Tokyo, Japan
| | - Robert Lafyatis
- Division of Rheumatology and Clinical Immunology. University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Ulf Landegren
- Department of Immunology, Genetics and Pathology, Research programme: Molecular Tools and Functional Genomics, Uppsala University, Uppsala, Sweden
| | | | - Javier Martin
- Department of Cell Biology and Immunology, Institute of Parasitology and Biomedicine López-Neyra, CSIC, Granada, Spain
| | - Marco Matucci-Cerinic
- Unit of Immunology, Rheumatology, Allergy and Rare Diseases and Inflammation, fibrosis and aging Initiative, IRCCS Ospedle San Raffaele and Vita Salute University San Raffaele, Milan, Italy
| | - Zsuzsanna H McMahan
- Department of Internal Medicine, Division of Rheumatology, UTHealth Houston, Houston, TX, USA
| | - Ana L Mora
- Division of Pulmonary, Critical Care and Sleep Medicine, Davis Heart and Lung research Institute, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Luc Mouthon
- Department of Internal Medicine, Reference Center for Rare Systemic Autoimmune and Auto-Inflammatory diseases in Île-de-France, East and West, Cochin Hospital, Public Assistance-Hospitals of Paris, Paris-Centre, Paris Cité University, Paris, France
| | - Marlene Rabinovitch
- Department of Paediatrics, Stanford University School of Medicine, Stanford, CA, USA
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
- Vera Moulton Wall Center for Pulmonary Vascular Diseases, Stanford University School of Medicine, Stanford, CA, USA
- Basic Science and Engineering (BASE) Initiative, Betty Irene Moore Children's Heart Center, Lucile Packard Children's Hospital, Stanford University School of Medicine, Stanford, CA, USA
| | - Mauricio Rojas
- Division of Pulmonary, Critical Care and Sleep Medicine, Davis Heart and Lung research Institute, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Kristofer Rubin
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Maria Trojanowska
- Boston University, Department of Medicine, Arthritis & Autoimmune Diseases Research Center, Boston, MA, USA
| | - John Varga
- Division of Rheumatology, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, USA
| | - Michael L Whitfield
- Department of Biomedical Data Science, Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover, NH, USA
| | - Armando Gabrielli
- Hiller Research Center, University Hospital Düsseldorf, Medical Faculty of Heinrich Heine University, Düsseldorf, Germany.
- Foundation of Molecular Medicine and Cellular Therapy Polytechnic University of Marche, Via Tronto, Ancona, Italy.
| | - Thomas Krieg
- Translational Matrix Biology, Cologne Excellence Cluster on Cellular Stress Responses in Ageing-Associated Diseases (CECAD) and Center for Molecular Medicine (CMMC) University of Cologne, Cologne, Germany.
| |
Collapse
|
5
|
Huang J, Hu Y, Wang S, Liu Y, Sun X, Wang X, Yu H. Single-cell RNA sequencing in autoimmune diseases: New insights and challenges. Pharmacol Ther 2025; 267:108807. [PMID: 39894174 DOI: 10.1016/j.pharmthera.2025.108807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Revised: 01/02/2025] [Accepted: 01/29/2025] [Indexed: 02/04/2025]
Abstract
Autoimmune diseases involve a variety of cell types, yet the intricacies of their individual roles within molecular mechanisms and therapeutic strategies remain poorly understood. Single-cell RNA sequencing (scRNA-seq) offers detailed insights into transcriptional diversity at the single-cell level, significantly advancing research in autoimmune diseases. This article explores how scRNA-seq enhances the understanding of cellular heterogeneity and its potential applications in the etiology, diagnosis, treatment, and prognosis of autoimmune diseases. By revealing a comprehensive cellular landscape, scRNA-seq illuminates the functional regulation of different cell subtypes during disease progression. It aids in identifying diagnostic and prognostic markers, and analyzing cell communication networks to uncover potential therapeutic targets. Despite its valuable contributions, addressing the limitations of scRNA-seq is essential for making further advancements.
Collapse
Affiliation(s)
- Jialing Huang
- Department of Immunology, Special Key Laboratory of Ocular Diseases of Guizhou Province, Zunyi Medical University, Guizhou, China
| | - Yuelin Hu
- Department of Immunology, Special Key Laboratory of Ocular Diseases of Guizhou Province, Zunyi Medical University, Guizhou, China
| | - Shuqing Wang
- Department of Immunology, Special Key Laboratory of Ocular Diseases of Guizhou Province, Zunyi Medical University, Guizhou, China
| | - Yuefang Liu
- School of Basic Medical Sciences, Special Key Laboratory of Gene Detection and Therapy of Guizhou Province, Zunyi Medical University, Guizhou, China
| | - Xin Sun
- School of Basic Medical Sciences, Special Key Laboratory of Gene Detection and Therapy of Guizhou Province, Zunyi Medical University, Guizhou, China
| | - Xin Wang
- School of Basic Medical Sciences, Special Key Laboratory of Gene Detection and Therapy of Guizhou Province, Zunyi Medical University, Guizhou, China
| | - Hongsong Yu
- Department of Immunology, Special Key Laboratory of Ocular Diseases of Guizhou Province, Zunyi Medical University, Guizhou, China; Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Guizhou, China.
| |
Collapse
|
6
|
Rosa I, Romano E, Fioretto BS, Manetti M. Autoantibodies as putative biomarkers and triggers of cell dysfunctions in systemic sclerosis. Curr Opin Rheumatol 2025; 37:51-63. [PMID: 39046085 DOI: 10.1097/bor.0000000000001035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/25/2024]
Abstract
PURPOSE OF REVIEW Antinuclear autoantibodies represent a serological hallmark of systemic sclerosis (SSc), with anticentromere, antitopoisomerase-I, and anti-RNA polymerase III antibodies routinely assessed for diagnosis, clinical subset classification, and prognosis. In addition, an increasing number of autoantibodies have been demonstrated to play a pathogenic role by mediating different SSc manifestations. This review aims to give an overview on autoantibodies as putative biomarkers in SSc and discuss their possible pathogenic role as triggers of cell dysfunctions. RECENT FINDINGS Over the years, different autoantibodies have been proposed as biomarkers aiding in diagnosis, disease subtype classification, disease progression prediction, organ involvement, as well as in understanding treatment response. Increasing literature also indicates functional autoantibodies as direct contributors to SSc pathogenesis by exerting agonistic or antagonistic activities on their specific cognate targets. SUMMARY In SSc, search and validation of novel autoantibodies with higher diagnostic specificity and more accurate predictive values are increasingly needed for early diagnosis and specific follow-up, and to define the best therapeutic option according to different disease subsets. Moreover, since autoantibodies are also emerging as functional pathogenic players, a better unraveling of their possible pathomechanisms becomes essential to identify new targets and develop promising therapeutic agents able to neutralize their effects.
Collapse
Affiliation(s)
- Irene Rosa
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | | | | | | |
Collapse
|
7
|
Rouvière B, Le Dantec C, Bettacchioli E, Beretta L, Foulquier N, Cao C, Jamin C, Pers JO, Kerick M, Martin J, Alarcón-Riquelme ME, de Moreuil C, Cornec D, Hillion S. Stratification according to autoantibody status in systemic sclerosis reveals distinct molecular signatures. Ann Rheum Dis 2024:ard-2024-225925. [PMID: 39438128 DOI: 10.1136/ard-2024-225925] [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: 04/08/2024] [Accepted: 10/07/2024] [Indexed: 10/25/2024]
Abstract
OBJECTIVES Systemic sclerosis (SSc) is a heterogeneous disease, complicating its management. Its complexity and the insufficiency of clinical manifestations alone to delineate homogeneous patient groups further challenge this task. However, autoantibodies could serve as relevant markers for the pathophysiological mechanisms driving the disease. Identifying specific immunological mechanisms based on patients' serological statuses might facilitate a deeper understanding of the diversity of the disease. METHODS A cohort of 206 patients with SSc enrolled in the PRECISESADS cross-sectional study was examined. Patients were stratified based on their anti-centromere (ACA) and anti-SCL70 (SCL70) antibody statuses. Comprehensive omics analyses including transcriptomic, flow cytometric, cytokine and metabolomic data were analysed to characterise the differences between these patient groups. RESULTS Patients with SCL70 antibodies showed severe clinical features such as diffuse cutaneous sclerosis and pulmonary fibrosis and were biologically distinguished by unique transcriptomic profiles. They exhibit a pro-inflammatory and fibrotic signature associated with impaired tissue remodelling and increased carnitine metabolism. Conversely, ACA-positive patients exhibited an immunomodulation and tissue homeostasis signature and increased phospholipid metabolism. CONCLUSIONS Patients with SSc display varying biological profiles based on their serological status. The findings highlight the potential utility of serological status as a discriminating factor in disease severity and suggest its relevance in tailoring treatment strategies and future research directions.
Collapse
Affiliation(s)
- Bénedicte Rouvière
- U1227, LBAI, Univ Brest, Inserm, and CHU Brest, Brest, France, Brest, France
| | | | | | - Lorenzo Beretta
- Referral Center for Systemic Autoimmune Diseases, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico di Milano, Milan, Italy
| | - Nathan Foulquier
- U1227, LBAI, Univ Brest, Inserm, and CHU Brest, Brest, France, Brest, France
| | - Celine Cao
- U1227, LBAI, Univ Brest, Inserm, and CHU Brest, Brest, France, Brest, France
| | - Christophe Jamin
- U1227, LBAI, Univ Brest, Inserm, and CHU Brest, Brest, France, Brest, France
| | | | - Martin Kerick
- Instituto de Parasitologia y Biomedicina Lopez-Neyra, Granada, Granada, Spain
| | - Javier Martin
- Institute of Parasitology and Biomedicine López-Neyra, CSIC, Granada, Spain, Granada, Spain
| | - Marta Eugenia Alarcón-Riquelme
- Centre for Genomics and Oncological Research (GENYO), Pfizer-University of Granada-Andalusian Regional Government, Granada, Spain
- Institute for Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | | | - Divi Cornec
- U1227, LBAI, Univ Brest, Inserm, and CHU Brest, Brest, France, Brest, France
| | - Sophie Hillion
- U1227, LBAI, Univ Brest, Inserm, and CHU Brest, Brest, France, Brest, France
| |
Collapse
|
8
|
Liang M, Wang L, Tian X, Wang K, Zhu X, Huang L, Li Q, Ye W, Chen C, Yang H, Wu W, Chen X, Zhu X, Xue Y, Wan W, Wu Y, Lu L, Wang J, Zou H, Ying T, Zhou F. Identification and validation of anti-protein arginine methyltransferase 5 (PRMT5) antibody as a novel biomarker for systemic sclerosis (SSc). Ann Rheum Dis 2024; 83:1144-1155. [PMID: 38684324 PMCID: PMC11420721 DOI: 10.1136/ard-2024-225596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2024] [Accepted: 04/17/2024] [Indexed: 05/02/2024]
Abstract
OBJECTIVES In the complex panorama of autoimmune diseases, the characterisation of pivotal contributing autoantibodies that are involved in disease progression remains challenging. This study aimed to employ a global antibody profiling strategy to identify novel antibodies and investigate their association with systemic sclerosis (SSc). METHODS We implemented this strategy by conducting immunoprecipitation (IP) following on-bead digestion with the sera of patients with SSc or healthy donors, using antigen pools derived from cell lysates. The enriched antigen-antibody complex was proceeded with mass spectrometry (MS)-based quantitative proteomics and over-represented by bioinformatics analysis. The candidate antibodies were then orthogonally validated in two independent groups of patients with SSc. Mice were immunised with the target antigen, which was subsequently evaluated by histological examination and RNA sequencing. RESULTS The IP-MS analysis, followed by validation in patients with SSc, revealed a significant elevation in anti-PRMT5 antibodies among patients with SSc. These antibodies exhibited robust diagnostic accuracy in distinguishing SSc from healthy controls and other autoimmune conditions, including systemic lupus erythematosus and Sjögren's syndrome, with an area under the curve ranging from 0.900 to 0.988. The elevation of anti-PRMT5 antibodies was verified in a subsequent independent group with SSc using an additional method, microarray. Notably, 31.11% of patients with SSc exhibited seropositivity for anti-PRMT5 antibodies. Furthermore, the titres of anti-PRMT5 antibodies demonstrated a correlation with the progression or regression trajectory in SSc. PRMT5 immunisation displayed significant inflammation and fibrosis in both the skin and lungs of mice. This was concomitant with the upregulation of multiple proinflammatory and profibrotic pathways, thereby underscoring a potentially pivotal role of anti-PRMT5 antibodies in SSc. CONCLUSIONS This study has identified anti-PRMT5 antibodies as a novel biomarker for SSc.
Collapse
Affiliation(s)
- Minrui Liang
- Department of Rheumatology, Huashan Hospital, Fudan University, Shanghai, China
- Institute of Rheumatology, Immunology and Allergy, Fudan University, Shanghai, China
- Huashan Rare Disease Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Lingbiao Wang
- Department of Rheumatology, Huashan Hospital, Fudan University, Shanghai, China
- Institute of Rheumatology, Immunology and Allergy, Fudan University, Shanghai, China
- Huashan Rare Disease Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Xiaolong Tian
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS) and Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, Shanghai Engineering Research Center for Synthetic Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Kun Wang
- Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xiaoyi Zhu
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS) and Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, Shanghai Engineering Research Center for Synthetic Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Linlin Huang
- Department of Rheumatology, Huashan Hospital, Fudan University, Shanghai, China
- Institute of Rheumatology, Immunology and Allergy, Fudan University, Shanghai, China
- Huashan Rare Disease Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Qing Li
- Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Wenjing Ye
- Department of Rheumatology, Huashan Hospital, Fudan University, Shanghai, China
- Institute of Rheumatology, Immunology and Allergy, Fudan University, Shanghai, China
- Huashan Rare Disease Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Chen Chen
- Department of Rheumatology, Huashan Hospital, Fudan University, Shanghai, China
- Institute of Rheumatology, Immunology and Allergy, Fudan University, Shanghai, China
- Department of Emergency Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Haihua Yang
- Department of Respiratory and Critical Care Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Wanqing Wu
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
| | - Xiangjun Chen
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
| | - Xiaoxia Zhu
- Department of Rheumatology, Huashan Hospital, Fudan University, Shanghai, China
- Institute of Rheumatology, Immunology and Allergy, Fudan University, Shanghai, China
- Huashan Rare Disease Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Yu Xue
- Department of Rheumatology, Huashan Hospital, Fudan University, Shanghai, China
- Institute of Rheumatology, Immunology and Allergy, Fudan University, Shanghai, China
- Huashan Rare Disease Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Weiguo Wan
- Department of Rheumatology, Huashan Hospital, Fudan University, Shanghai, China
- Institute of Rheumatology, Immunology and Allergy, Fudan University, Shanghai, China
- Huashan Rare Disease Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Yanling Wu
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS) and Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, Shanghai Engineering Research Center for Synthetic Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Liwei Lu
- Department of Pathology, The University of Hong Kong, Hong Kong, China
| | - Jiucun Wang
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, and Human Phenome Institute, Fudan University, Shanghai, China
| | - Hejian Zou
- Department of Rheumatology, Huashan Hospital, Fudan University, Shanghai, China
- Institute of Rheumatology, Immunology and Allergy, Fudan University, Shanghai, China
- Huashan Rare Disease Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Tianlei Ying
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS) and Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, Shanghai Engineering Research Center for Synthetic Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Feng Zhou
- Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
9
|
Tian N, Cheng H, Du Y, Wang X, Lei Y, Liu X, Chen M, Xu Z, Wang L, Yin H, Fu R, Li D, Zhou P, Lu L, Yin Z, Dai SM, Li B. Cannabinoid receptor 2 selective agonist alleviates systemic sclerosis by inhibiting Th2 differentiation through JAK/SOCS3 signaling. J Autoimmun 2024; 147:103233. [PMID: 38797049 DOI: 10.1016/j.jaut.2024.103233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 02/09/2024] [Accepted: 04/16/2024] [Indexed: 05/29/2024]
Abstract
Systemic sclerosis (SSc) poses a significant challenge in autoimmunology, characterized by the development of debilitating fibrosis of skin and internal organs. The pivotal role of dysregulated T cells, notably the skewed polarization toward Th2 cells, has been implicated in the vascular damage and progressive fibrosis observed in SSc. In this study, we explored the underlying mechanisms by which cannabinoid receptor 2 (CB2) highly selective agonist HU-308 restores the imbalance of T cells to alleviate SSc. Using a bleomycin-induced SSc (BLM-SSc) mouse model, we demonstrated that HU-308 effectively attenuates skin and lung fibrosis by specifically activating CB2 on CD4+ T cells to inhibit the polarization of Th2 cells in BLM-SSc mice, which was validated by Cnr2-specific-deficient mice. Different from classical signaling downstream of G protein-coupled receptors (GPCRs), HU-308 facilitates the expression of SOCS3 protein and subsequently impedes the IL2/STAT5 signaling pathway during Th2 differentiation. The deficiency of SOCS3 partially mitigated the impact of HU-308. Analysis of a cohort comprising 80 SSc patients and 82 healthy controls revealed an abnormal elevation in the Th2/Th1 ratio in SSc patients. The proportion of Th2 cells showed a significant positive correlation with mRSS score and positivity of anti-Scl-70. Administration of HU-308 to PBMCs and peripheral CD4+ T cells from SSc patients led to the upregulation of SOCS3, which effectively suppressed the aberrantly activated STAT5 signaling pathway and the proportion of CD4+IL4+ T cells. In conclusion, our findings unveil a novel mechanism by which the CB2 agonist HU-308 ameliorates fibrosis in SSc by targeting and reducing Th2 responses. These insights provide a foundation for future therapeutic approaches in SSc by modulating Th2 responses.
Collapse
Affiliation(s)
- Na Tian
- Department of Rheumatology and Immunology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China; Center for Immune-Related Diseases at Shanghai Institute of Immunology, Department of Respiratory and Critical Care Medicine of Ruijin Hospital, Department of Thoracic Surgery of Ruijin Hospital, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Hao Cheng
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Department of Respiratory and Critical Care Medicine of Ruijin Hospital, Department of Thoracic Surgery of Ruijin Hospital, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Department of Rheumatism and Immunology, Peking University Shenzhen Hospital, Guangdong, China; Center for Cancer Immunology Research, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, China.
| | - Yu Du
- Department of Rheumatology and Immunology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Xiaoxia Wang
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Department of Respiratory and Critical Care Medicine of Ruijin Hospital, Department of Thoracic Surgery of Ruijin Hospital, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Songjiang Research Institute, Songjiang District Central Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Yi Lei
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Department of Respiratory and Critical Care Medicine of Ruijin Hospital, Department of Thoracic Surgery of Ruijin Hospital, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Xinnan Liu
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Department of Respiratory and Critical Care Medicine of Ruijin Hospital, Department of Thoracic Surgery of Ruijin Hospital, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Miao Chen
- Department of Rheumatology and Immunology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Zhan Xu
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Department of Respiratory and Critical Care Medicine of Ruijin Hospital, Department of Thoracic Surgery of Ruijin Hospital, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Lingbiao Wang
- Division of Rheumatology, Huashan Hospital, Fudan University, Shanghai, China.
| | - Hanlin Yin
- Department of Rheumatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Rong Fu
- Core Facility of Basic Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Dan Li
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Department of Respiratory and Critical Care Medicine of Ruijin Hospital, Department of Thoracic Surgery of Ruijin Hospital, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Penghui Zhou
- State Key Laboratory of Oncology in Southern China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China.
| | - Liangjing Lu
- Department of Rheumatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Zhinan Yin
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, Zhuhai People's Hospital Affiliated with Jinan University, Jinan University, Zhuhai, China; State Key Laboratory of Bioactive Molecules and Druggability Assessment, The Biomedical Translational Research Institute, Health Science Center (School of Medicine), Jinan University, Guangzhou, China; Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education, Guangzhou, China.
| | - Sheng-Ming Dai
- Department of Rheumatology and Immunology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Bin Li
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Department of Respiratory and Critical Care Medicine of Ruijin Hospital, Department of Thoracic Surgery of Ruijin Hospital, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Department of Integrated TCM & Western Medicine, Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China; Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University, Shanghai, China; Department of Oncology, Department of Hepatobiliary Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China.
| |
Collapse
|
10
|
Zhu P, Deng W. Single-Cell RNA-Sequencing Analyses Identify APLNR, INS-IGF2, RGCC Genes May Be Involved in the Pathogenesis of Systemic Sclerosis Skin. Clin Cosmet Investig Dermatol 2024; 17:1059-1069. [PMID: 38742168 PMCID: PMC11090198 DOI: 10.2147/ccid.s456593] [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: 01/26/2024] [Accepted: 04/21/2024] [Indexed: 05/16/2024]
Abstract
Background Systemic sclerosis represents a persistent autoimmune disorder marked with fibrosis affecting both skin and other organs, which leads to a diminished quality of life and increased mortality. The affected skin provides a valuable opportunity to explore the pathogenesis of systemic sclerosis. Nevertheless, the roles of various cell populations within scleroderma remain intricate. Methods We conducted a comprehensive reanalysis of recently published single-cell RNA-sequencing data from skin tissue cells in scleroderma. Through the utilization of Seurat, irGSEA, AUCell packages, and WGCNA analysis, we aimed to unveil crucial genes associated with the disease's etiological factors. Our investigation involved the characterization of heterogeneous pathway activities in both healthy and SSc-affected skin. Furthermore, we employed immunofluorescence techniques to validate the expression patterns of hub genes and differentially expressed genes. Results The Endothelial-to-Mesenchymal Transition (EndMT) pathway was upregulated in SSc skin. Notably, the M4 module within Endothelial cell subpopulation 1 exhibited a strong association with EndMT. Furthermore, we identified three overexpressed genes (APLNR, INS-IGF2, RGCC) that demonstrated a significant correlation with EndMT. Importantly, their expression levels were markedly higher in skin of individuals with SSc when compared to healthy controls. Conclusion APLNR, INS-IGF2 and RGCC serve as potential key players in the pathogenesis of SSc skin through EndMT-dependent mechanisms.
Collapse
Affiliation(s)
- Peiqiu Zhu
- Department of Dermatology and Venereology, Beijing Jishuitan Hospital, Capital Medical University, Beijing, People’s Republic of China
| | - Weiwei Deng
- Department of Dermatology, Dermatology Hospital of Southern Medical University, Guangzhou, People’s Republic of China
| |
Collapse
|
11
|
Shi Z, Liu Z, Wei Y, Zhang R, Deng Y, Li D. The role of dermal fibroblasts in autoimmune skin diseases. Front Immunol 2024; 15:1379490. [PMID: 38545113 PMCID: PMC10965632 DOI: 10.3389/fimmu.2024.1379490] [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: 01/31/2024] [Accepted: 03/01/2024] [Indexed: 04/18/2024] Open
Abstract
Fibroblasts are an important subset of mesenchymal cells in maintaining skin homeostasis and resisting harmful stimuli. Meanwhile, fibroblasts modulate immune cell function by secreting cytokines, thereby implicating their involvement in various dermatological conditions such as psoriasis, vitiligo, and atopic dermatitis. Recently, variations in the subtypes of fibroblasts and their expression profiles have been identified in these prevalent autoimmune skin diseases, implying that fibroblasts may exhibit distinct functionalities across different diseases. In this review, from the perspective of their fundamental functions and remarkable heterogeneity, we have comprehensively collected evidence on the role of fibroblasts and their distinct subpopulations in psoriasis, vitiligo, atopic dermatitis, and scleroderma. Importantly, these findings hold promise for guiding future research directions and identifying novel therapeutic targets for treating these diseases.
Collapse
Affiliation(s)
| | | | | | | | | | - Dong Li
- Department of Dermatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
12
|
Willenborg S, Satzinger S, Eming SA. [Skin fibrosis : Novel insights in pathophysiology and treatment]. DERMATOLOGIE (HEIDELBERG, GERMANY) 2024; 75:218-224. [PMID: 38351374 DOI: 10.1007/s00105-024-05299-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 01/10/2024] [Indexed: 02/24/2024]
Abstract
The pathogenesis of fibrosing alterations in the skin and other organ systems is not yet sufficiently understood and current therapeutic options are limited. Fibrosing diseases of the skin lead to a loss of function, which can subsequently be accompanied by serious impairments in quality of life, increased morbidity and ultimately increased mortality. There are currently only a few pharmacological and therapeutic approaches approved to prevent or ameliorate fibrosing diseases. Furthermore, tissue-specific versus common, non-organ-specific pathophysiological cellular and molecular mechanisms are not resolved. The development of new, cause-based and therefore likely more efficient therapeutic approaches is urgently needed. This represents a major challenge, but also opens up the opportunity for special contributions to improve this medically unsolved problem. Here we present important findings from recent years with a focus on the role of the immune response in fibrogenesis.
Collapse
Affiliation(s)
- Sebastian Willenborg
- Klinik und Poliklinik für Dermatologie und Venerologie, Uniklinik Köln, Kerpener Str. 62, 50937, Köln, Deutschland
| | - Sabrina Satzinger
- Klinik und Poliklinik für Dermatologie und Venerologie, Uniklinik Köln, Kerpener Str. 62, 50937, Köln, Deutschland
| | - Sabine A Eming
- Klinik und Poliklinik für Dermatologie und Venerologie, Uniklinik Köln, Kerpener Str. 62, 50937, Köln, Deutschland.
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, Köln, Deutschland.
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Köln, Deutschland.
- Institute of Zoology, Developmental Biology Unit, University of Cologne, Köln, Deutschland.
| |
Collapse
|