1
|
Son HH, Moon SJ. Pathogenesis of systemic sclerosis: an integrative review of recent advances. JOURNAL OF RHEUMATIC DISEASES 2025; 32:89-104. [PMID: 40134549 PMCID: PMC11931279 DOI: 10.4078/jrd.2024.0129] [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: 11/05/2024] [Revised: 11/07/2024] [Accepted: 11/07/2024] [Indexed: 03/27/2025]
Abstract
Systemic sclerosis (SSc), or scleroderma, is a complex autoimmune connective tissue disease characterized by autoimmunity, vasculopathy, and progressive organ fibrosis, leading to severe organ dysfunction. The disease begins with a vascular injury triggered by autoimmune responses and environmental factors against a backdrop of genetic predisposition. This injury impairs angiogenesis and vasculogenesis, resulting in capillary loss and arteriolar constriction, which promotes immune cell infiltration and sustained inflammation within affected tissues. These vascular anomalies cause severe complications, including pulmonary artery hypertension, scleroderma renal crisis, and skin ulcers. Chronic inflammation fosters persistent fibroblast activation, resulting in extensive fibrosis that defines SSc. This review synthesizes the latest research on pathogenesis of SSc, highlighting the shift from fundamental research to a precision therapeutic approach. It explores the potential of technologies like flow cytometry and single-cell RNA sequencing to investigate pathogenic cell subtypes. These platforms integrate transcriptomic, genomic, proteomic, and epigenomic data to uncover insights into the underlying mechanisms of SSc pathogenesis. This review advocates for a multidisciplinary, patient-centric approach that harnesses recent scientific advances, directing future SSc research toward personalized and precise interventions.
Collapse
Affiliation(s)
- Ha-Hee Son
- Division of Rheumatology, Department of Internal Medicine, Yeouido St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Su-Jin Moon
- Division of Rheumatology, Department of Internal Medicine, Yeouido St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| |
Collapse
|
2
|
He D, Shannon CP, Hirota JA, Ask K, Ryerson CJ, Tebbutt SJ. Diagnostic potential of genomic blood biomarkers of pulmonary fibrosis in a prospective cohort. PLoS One 2024; 19:e0314876. [PMID: 39625896 PMCID: PMC11614250 DOI: 10.1371/journal.pone.0314876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Accepted: 11/18/2024] [Indexed: 12/06/2024] Open
Abstract
Fibrotic interstitial lung diseases (ILDs) result from excessive deposition of extracellular matrix (ECM) proteins in the lung, causing irreversible damage to the lung architecture. Clinical management of ILDs differs depending on the diagnosis, but differentiation between subtypes can be difficult and better clinical biomarkers are needed. In this study, we use a 166-gene NanoString assay to investigate whether there are ILD subtype-specific transcripts in whole blood. We identified one transcript, killer cell lectin like receptor 1 (KLRF1), as differentially expressed between idiopathic pulmonary fibrosis (IPF) and systemic sclerosis-associated ILD (SSc-ILD), and identified two transcripts (VCAN, LTK) associated with IPF expression against other ILD subtypes. These findings were validated by examining their expression in ILD lung, with KLRF1 expression significantly higher in SSc-ILD compared to IPF and hypersensitivity pneumonitis (HP) samples. Taken together, this pilot study provides support for the use of the peripheral transcriptome in identifying diagnostic biomarkers of ILD with biological relevance.
Collapse
MESH Headings
- Humans
- Biomarkers/blood
- Idiopathic Pulmonary Fibrosis/blood
- Idiopathic Pulmonary Fibrosis/genetics
- Idiopathic Pulmonary Fibrosis/diagnosis
- Male
- Female
- Prospective Studies
- Middle Aged
- Lung Diseases, Interstitial/blood
- Lung Diseases, Interstitial/genetics
- Lung Diseases, Interstitial/diagnosis
- Scleroderma, Systemic/blood
- Scleroderma, Systemic/genetics
- Scleroderma, Systemic/complications
- Scleroderma, Systemic/diagnosis
- Aged
- Alveolitis, Extrinsic Allergic/blood
- Alveolitis, Extrinsic Allergic/diagnosis
- Alveolitis, Extrinsic Allergic/genetics
- Transcriptome
- Pilot Projects
- Lectins, C-Type/genetics
- Lectins, C-Type/blood
- Pulmonary Fibrosis/blood
- Pulmonary Fibrosis/genetics
- Pulmonary Fibrosis/diagnosis
- Genomics/methods
Collapse
Affiliation(s)
- Daniel He
- Department of Medicine, Division of Respiratory Medicine, University of British Columbia, Vancouver, BC, Canada
- Centre for Heart Lung Innovation, St Paul’s Hospital, Vancouver, BC, Canada
- Prevention of Organ Failure (PROOF) Centre of Excellence, Vancouver, BC, Canada
| | - Casey P. Shannon
- Centre for Heart Lung Innovation, St Paul’s Hospital, Vancouver, BC, Canada
- Prevention of Organ Failure (PROOF) Centre of Excellence, Vancouver, BC, Canada
- Providence Health Care Research Institute, Providence Research, Vancouver, BC, Canada
| | - Jeremy A. Hirota
- Department of Medicine, Division of Respiratory Medicine, University of British Columbia, Vancouver, BC, Canada
- Department of Medicine, McMaster University, Hamilton, ON, Canada
- Department of Medicine, Firestone Institute for Respiratory Health–Division of Respirology, McMaster University, Hamilton, ON, Canada
- Department of Biology, University of Waterloo, Waterloo, Ontario, Canada
| | - Kjetil Ask
- Department of Medicine, McMaster University, Hamilton, ON, Canada
- Department of Medicine, Firestone Institute for Respiratory Health–Division of Respirology, McMaster University, Hamilton, ON, Canada
| | - Christopher J. Ryerson
- Department of Medicine, Division of Respiratory Medicine, University of British Columbia, Vancouver, BC, Canada
- Centre for Heart Lung Innovation, St Paul’s Hospital, Vancouver, BC, Canada
- Providence Health Care Research Institute, Providence Research, Vancouver, BC, Canada
| | - Scott J. Tebbutt
- Department of Medicine, Division of Respiratory Medicine, University of British Columbia, Vancouver, BC, Canada
- Centre for Heart Lung Innovation, St Paul’s Hospital, Vancouver, BC, Canada
- Prevention of Organ Failure (PROOF) Centre of Excellence, Vancouver, BC, Canada
- Providence Health Care Research Institute, Providence Research, Vancouver, BC, Canada
| |
Collapse
|
3
|
Pezeshkian F, Shahriarirad R, Mahram H. An overview of the role of chemokine CX3CL1 (Fractalkine) and CX3C chemokine receptor 1 in systemic sclerosis. Immun Inflamm Dis 2024; 12:e70034. [PMID: 39392260 PMCID: PMC11467895 DOI: 10.1002/iid3.70034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 09/13/2024] [Accepted: 09/19/2024] [Indexed: 10/12/2024] Open
Abstract
INTRODUCTION Systemic sclerosis (SSc) is a complex autoimmune disease characterized by fibrosis, vascular damage, and immune dysregulation. Fractalkine or chemokine (C-X3-C motif) ligand 1 (CX3CL1), a chemokine and adhesion molecule, along with its receptor CX3CR1, have been implicated in the inflammatory processes of SSc. CX3CL1 functions as both a chemoattractant and an adhesion molecule, guiding immune cell trafficking. This systematic review examines the role of CX3CL1 and its receptor CX3CR1 in the pathogenesis of SSc, with a focus on pulmonary and vascular complications. METHODS A systematic literature search was conducted across databases including PubMed, Scopus, and Web of Science from inception to November 2020. The search focused on studies investigating the CX3CL1/CX3CR1 axis in the context of SSc. RESULTS The review identified elevated CX3CL1 expression in SSc patients, particularly in the skin and lungs, where CX3CR1 is expressed on infiltrating immune cells. Higher levels of CX3CL1 were correlated with the severity of interstitial lung disease in SSc patients, indicating a potential predictive marker for disease progression. CX3CR1-positive monocytes and NK cells were recruited to inflamed tissues, contributing to fibrosis and tissue damage. Animal studies showed that inhibition of the CX3CL1/CX3CR1 axis reduced fibrosis and improved vascular function. CONCLUSION The CX3CL1/CX3CR1 axis plays a key role in immune cell recruitment and fibrosis in SSc. Elevated CX3CL1 levels are associated with lung and vascular complications, making it a potential biomarker for disease progression and a promising therapeutic target.
Collapse
Affiliation(s)
| | - Reza Shahriarirad
- Thoracic and Vascular Surgery Research CenterShiraz University of Medical SciencesShirazIran
| | | |
Collapse
|
4
|
Balog JÁ, Zvara Á, Bukovinszki V, Puskás LG, Balog A, Szebeni GJ. Comparative single-cell multiplex immunophenotyping of therapy-naive patients with rheumatoid arthritis, systemic sclerosis, and systemic lupus erythematosus shed light on disease-specific composition of the peripheral immune system. Front Immunol 2024; 15:1376933. [PMID: 38726007 PMCID: PMC11079270 DOI: 10.3389/fimmu.2024.1376933] [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: 01/26/2024] [Accepted: 04/03/2024] [Indexed: 05/12/2024] Open
Abstract
Introduction Systemic autoimmune diseases (SADs) are a significant burden on the healthcare system. Understanding the complexity of the peripheral immunophenotype in SADs may facilitate the differential diagnosis and identification of potential therapeutic targets. Methods Single-cell mass cytometric immunophenotyping was performed on peripheral blood mononuclear cells (PBMCs) from healthy controls (HCs) and therapy-naive patients with rheumatoid arthritis (RA), progressive systemic sclerosis (SSc), and systemic lupus erythematosus (SLE). Immunophenotyping was performed on 15,387,165 CD45+ live single cells from 52 participants (13 cases/group), using an antibody panel to detect 34 markers. Results Using the t-SNE (t-distributed stochastic neighbor embedding) algorithm, the following 17 main immune cell types were determined: CD4+/CD57- T cells, CD4+/CD57+ T cells, CD8+/CD161- T cells, CD8+/CD161+/CD28+ T cells, CD8dim T cells, CD3+/CD4-/CD8- T cells, TCRγ/δ T cells, CD4+ NKT cells, CD8+ NKT cells, classic NK cells, CD56dim/CD98dim cells, B cells, plasmablasts, monocytes, CD11cdim/CD172dim cells, myeloid dendritic cells (mDCs), and plasmacytoid dendritic cells (pDCs). Seven of the 17 main cell types exhibited statistically significant frequencies in the investigated groups. The expression levels of the 34 markers in the main populations were compared between HCs and SADs. In summary, 59 scatter plots showed significant differences in the expression intensities between at least two groups. Next, each immune cell population was divided into subpopulations (metaclusters) using the FlowSOM (self-organizing map) algorithm. Finally, 121 metaclusters (MCs) of the 10 main immune cell populations were found to have significant differences to classify diseases. The single-cell T-cell heterogeneity represented 64MCs based on the expression of 34 markers, and the frequency of 23 MCs differed significantly between at least twoconditions. The CD3- non-T-cell compartment contained 57 MCs with 17 MCs differentiating at least two investigated groups. In summary, we are the first to demonstrate the complexity of the immunophenotype of 34 markers over 15 million single cells in HCs vs. therapy-naive patients with RA, SSc, and SLE. Disease specific population frequencies or expression patterns of peripheral immune cells provide a single-cell data resource to the scientific community.
Collapse
Affiliation(s)
- József Á. Balog
- Laboratory of Functional Genomics, Institute of Genetics, HUN-REN Biological Research Centre, Szeged, Hungary
- Core Facility, HUN-REN Biological Research Centre, Szeged, Hungary
| | - Ágnes Zvara
- Laboratory of Functional Genomics, Institute of Genetics, HUN-REN Biological Research Centre, Szeged, Hungary
- Core Facility, HUN-REN Biological Research Centre, Szeged, Hungary
| | - Vivien Bukovinszki
- Department of Rheumatology and Immunology, Faculty of Medicine, Albert Szent-Gyorgyi Health Centre, University of Szeged, Szeged, Hungary
| | - László G. Puskás
- Laboratory of Functional Genomics, Institute of Genetics, HUN-REN Biological Research Centre, Szeged, Hungary
- Core Facility, HUN-REN Biological Research Centre, Szeged, Hungary
| | - Attila Balog
- Department of Rheumatology and Immunology, Faculty of Medicine, Albert Szent-Gyorgyi Health Centre, University of Szeged, Szeged, Hungary
| | - Gábor J. Szebeni
- Laboratory of Functional Genomics, Institute of Genetics, HUN-REN Biological Research Centre, Szeged, Hungary
- Core Facility, HUN-REN Biological Research Centre, Szeged, Hungary
- Department of Internal Medicine, Hematology Centre, Faculty of Medicine University of Szeged, Szeged, Hungary
- Astridbio Technologies Ltd., Szeged, Hungary
| |
Collapse
|
5
|
Shin YJ, Lee JH. Exploring the Molecular and Developmental Dynamics of Endothelial Cell Differentiation. Int J Stem Cells 2024; 17:15-29. [PMID: 37879853 PMCID: PMC10899884 DOI: 10.15283/ijsc23086] [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: 06/11/2023] [Revised: 08/06/2023] [Accepted: 09/05/2023] [Indexed: 10/27/2023] Open
Abstract
The development and differentiation of endothelial cells (ECs) are fundamental processes with significant implications for both health and disease. ECs, which are found in all organs and blood vessels, play a crucial role in facilitating nutrient and waste exchange and maintaining proper vessel function. Understanding the intricate signaling pathways involved in EC development holds great promise for enhancing vascularization, tissue engineering, and vascular regeneration. Hematopoietic stem cells originating from hemogenic ECs, give rise to diverse immune cell populations, and the interaction between ECs and immune cells is vital for maintaining vascular integrity and regulating immune responses. Dysregulation of vascular development pathways can lead to various diseases, including cancer, where tumor-specific ECs promote tumor growth through angiogenesis. Recent advancements in single-cell genomics and in vivo genetic labeling have shed light on EC development, plasticity, and heterogeneity, uncovering tissue-specific gene expression and crucial signaling pathways. This review explores the potential of ECs in various applications, presenting novel opportunities for advancing vascular medicine and treatment strategies.
Collapse
Affiliation(s)
- Yu Jung Shin
- Department of Bioengineering, University of Washington, Seattle, WA, USA
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA
| | - Jung Hyun Lee
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA
- Department of Dermatology, School of Medicine, University of Washington, Seattle, WA, USA
| |
Collapse
|
6
|
Pardos-Gea J, Martin-Fernandez L, Closa L, Ferrero A, Marzo C, Rubio-Rivas M, Mitjavila F, González-Porras JR, Bastida JM, Mateo J, Carrasco M, Bernardo Á, Astigarraga I, Aguinaco R, Corrales I, Garcia-Martínez I, Vidal F. Key Genes of the Immune System and Predisposition to Acquired Hemophilia A: Evidence from a Spanish Cohort of 49 Patients Using Next-Generation Sequencing. Int J Mol Sci 2023; 24:16372. [PMID: 38003562 PMCID: PMC10671092 DOI: 10.3390/ijms242216372] [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: 10/10/2023] [Revised: 11/10/2023] [Accepted: 11/13/2023] [Indexed: 11/26/2023] Open
Abstract
Acquired hemophilia A (AHA) is a rare bleeding disorder caused by the presence of autoantibodies against factor VIII (FVIII). As with other autoimmune diseases, its etiology is complex and its genetic basis is unknown. The aim of this study was to identify the immunogenetic background that predisposes individuals to AHA. HLA and KIR gene clusters, as well as KLRK1, were sequenced using next-generation sequencing in 49 AHA patients. Associations between candidate genes involved in innate and adaptive immune responses and AHA were addressed by comparing the alleles, genotypes, haplotypes, and gene frequencies in the AHA cohort with those in the donors' samples or Spanish population cohort. Two genes of the HLA cluster, as well as rs1049174 in KLRK1, which tags the natural killer (NK) cytotoxic activity haplotype, were found to be linked to AHA. Specifically, A*03:01 (p = 0.024; odds ratio (OR) = 0.26[0.06-0.85]) and DRB1*13:03 (p = 6.8 × 103, OR = 7.56[1.64-51.40]), as well as rs1049174 (p = 0.012), were significantly associated with AHA. In addition, two AHA patients were found to carry one copy each of the low-frequency allele DQB1*03:09 (nallele = 2, 2.04%), which was completely absent in the donors. To the best of our knowledge, this is the first time that the involvement of these specific alleles in the predisposition to AHA has been proposed. Further molecular and functional studies will be needed to unravel their specific contributions. We believe our findings expand the current knowledge on the genetic factors involved in susceptibility to AHA, which will contribute to improving the diagnosis and prognosis of AHA patients.
Collapse
Affiliation(s)
- Jose Pardos-Gea
- Systemic Autoimmune Diseases Unit, Department of Internal Medicine, Vall d’Hebron University Hospital, 08035 Barcelona, Spain
| | - Laura Martin-Fernandez
- Congenital Coagulopathies Laboratory, Blood and Tissue Bank, 08005 Barcelona, Spain
- Transfusional Medicine Group, Vall d’Hebron Research Institute, Autonomous University of Barcelona (VHIR-UAB), 08035 Barcelona, Spain
| | - Laia Closa
- Transfusional Medicine Group, Vall d’Hebron Research Institute, Autonomous University of Barcelona (VHIR-UAB), 08035 Barcelona, Spain
- Histocompatibility and Immunogenetics Laboratory, Blood and Tissue Bank, 08005 Barcelona, Spain
| | - Ainara Ferrero
- Hematology Service, Arnau de Vilanova University Hospital, 25198 Lleida, Spain
| | - Cristina Marzo
- Hematology Service, Arnau de Vilanova University Hospital, 25198 Lleida, Spain
| | - Manuel Rubio-Rivas
- Department of Internal Medicine, Bellvitge University Hospital, Bellvitge Biomedical Research Institute (IDIBELL), University of Barcelona, L’Hospitalet de Llobregat, 08908 Barcelona, Spain; (M.R.-R.)
| | - Francesca Mitjavila
- Department of Internal Medicine, Bellvitge University Hospital, Bellvitge Biomedical Research Institute (IDIBELL), University of Barcelona, L’Hospitalet de Llobregat, 08908 Barcelona, Spain; (M.R.-R.)
| | - José Ramón González-Porras
- Department of Hematology, Complejo Asistencial Universitario de Salamanca (CAUSA), Instituto de Investigación Biomedica de Salamanca (IBSAL), Facultad de Medicina, Universidad de Salamanca (USAL), 37007 Salamanca, Spain
| | - José María Bastida
- Department of Hematology, Complejo Asistencial Universitario de Salamanca (CAUSA), Instituto de Investigación Biomedica de Salamanca (IBSAL), Facultad de Medicina, Universidad de Salamanca (USAL), 37007 Salamanca, Spain
| | - José Mateo
- Thrombosis and Hemostasis Unit, Sant Pau Campus Salut Barcelona, 08025 Barcelona, Spain
| | - Marina Carrasco
- Thrombosis and Hemostasis Unit, Sant Pau Campus Salut Barcelona, 08025 Barcelona, Spain
| | - Ángel Bernardo
- Hematology Service, Central University Hospital of Asturias, 33011 Oviedo, Spain
| | - Itziar Astigarraga
- Department of Pediatrics, Biobizkaia Health Research Institute, Hospital Universitario Cruces, University of the Basque Country UPV/EHU, 48903 Barakaldo, Spain
| | - Reyes Aguinaco
- Hematology Service, University Hospital Joan XXIII, 43002 Tarragona, Spain
| | - Irene Corrales
- Congenital Coagulopathies Laboratory, Blood and Tissue Bank, 08005 Barcelona, Spain
- Transfusional Medicine Group, Vall d’Hebron Research Institute, Autonomous University of Barcelona (VHIR-UAB), 08035 Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Instituto Carlos III (ISCIII), 28029 Madrid, Spain
| | - Iris Garcia-Martínez
- Congenital Coagulopathies Laboratory, Blood and Tissue Bank, 08005 Barcelona, Spain
- Transfusional Medicine Group, Vall d’Hebron Research Institute, Autonomous University of Barcelona (VHIR-UAB), 08035 Barcelona, Spain
| | - Francisco Vidal
- Congenital Coagulopathies Laboratory, Blood and Tissue Bank, 08005 Barcelona, Spain
- Transfusional Medicine Group, Vall d’Hebron Research Institute, Autonomous University of Barcelona (VHIR-UAB), 08035 Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Instituto Carlos III (ISCIII), 28029 Madrid, Spain
| |
Collapse
|
7
|
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]
|
8
|
Abstract
PURPOSE OF REVIEW Systemic sclerosis (SSc) is a chronic rheumatic disease that is characterized by immune activation, vasculopathy and fibrosis of the skin and internal organs. It has been proposed that premature onset of ageing pathways and associated senescent changes in cells contribute to the clinical and pathological features of SSc. The aim of this review is to critically review recent insights into the involvement of cellular senescence in SSc. RECENT FINDINGS Cellular senescence plays a critical role in SSc pathogenesis, particularly involving endothelial cells and fibroblasts. Immunosenescence could also contribute to SSc pathogenesis by direct alteration of cellular functions or indirect promotion of defective immune surveillance. Molecular studies have shed some light on how cellular senescence contributes to fibrosis. Recent and planned proof-of-concept trials using senotherapeutics showed promising results in fibrotic diseases, including SSc. SUMMARY There is increasing evidence implicating cellular senescence in SSc. The mechanisms underlying premature cellular senescence in SSc, and its potential role in pathogenesis, merit further investigation. Emerging drugs targeting senescence-related pathways might be potential therapeutic options for SSc.
Collapse
Affiliation(s)
- Pei-Suen Tsou
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
- Scleroderma Program, University of Michigan, Ann Arbor, MI, USA
| | - Bo Shi
- Department of Dermatology, Northwestern University, Chicago, IL, USA
| | - John Varga
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
- Scleroderma Program, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
9
|
Matei AE, Kubánková M, Xu L, Györfi AH, Boxberger E, Soteriou D, Papava M, Prater J, Hong X, Bergmann C, Kräter M, Schett G, Guck J, Distler JHW. Identification of a Distinct Monocyte-Driven Signature in Systemic Sclerosis Using Biophysical Phenotyping of Circulating Immune Cells. Arthritis Rheumatol 2022; 75:768-781. [PMID: 36281753 DOI: 10.1002/art.42394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 09/08/2022] [Accepted: 10/18/2022] [Indexed: 11/11/2022]
Abstract
OBJECTIVE Pathologically activated circulating immune cells, including monocytes, play major roles in systemic sclerosis (SSc). Their functional characterization can provide crucial information with direct clinical relevance. However, tools for the evaluation of pathologic immune cell activation and, in general, of clinical outcomes in SSc are scarce. Biophysical phenotyping (including characterization of cell mechanics and morphology) provides access to a novel, mostly unexplored layer of information regarding pathophysiologic immune cell activation. We hypothesized that the biophysical phenotyping of circulating immune cells, reflecting their pathologic activation, can be used as a clinical tool for the evaluation and risk stratification of patients with SSc. METHODS We performed biophysical phenotyping of circulating immune cells by real-time fluorescence and deformability cytometry (RT-FDC) in 63 SSc patients, 59 rheumatoid arthritis (RA) patients, 28 antineutrophil cytoplasmic antibody-associated vasculitis (AAV) patients, and 22 age- and sex-matched healthy donors. RESULTS We identified a specific signature of biophysical properties of circulating immune cells in SSc patients that was mainly driven by monocytes. Since it is absent in RA and AAV, this signature reflects an SSc-specific monocyte activation rather than general inflammation. The biophysical properties of monocytes indicate current disease activity, the extent of skin or lung fibrosis, and the severity of manifestations of microvascular damage, as well as the risk of disease progression in SSc patients. CONCLUSION Changes in the biophysical properties of circulating immune cells reflect their pathologic activation in SSc patients and are associated with clinical outcomes. As a high-throughput approach that requires minimal preparations, RT-FDC-based biophysical phenotyping of monocytes can serve as a tool for the evaluation and risk stratification of patients with SSc.
Collapse
Affiliation(s)
- Alexandru-Emil Matei
- Department of Rheumatology and Hiller Research Unit, University Hospital Düsseldorf, Medical Faculty of Heinrich Heine University, Düsseldorf, Germany, and Department of Internal Medicine 3-Rheumatology and Immunology and Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander University Erlangen-Nürnberg (FAU), and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Markéta Kubánková
- Max Planck Institute for the Science of Light & Max-Planck-Center für Physik und Medizin, Erlangen, Germany, and Biotechnology Center, Center for Molecular and Cellular Bioengineering, Technische Universität Dresden, Dresden, Germany
| | - Liyan Xu
- Department of Rheumatology and Hiller Research Unit, University Hospital Düsseldorf, Medical Faculty of Heinrich Heine University, Düsseldorf, Germany, and Department of Internal Medicine 3-Rheumatology and Immunology and Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander University Erlangen-Nürnberg (FAU), and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Andrea-Hermina Györfi
- Department of Rheumatology and Hiller Research Unit, University Hospital Düsseldorf, Medical Faculty of Heinrich Heine University, Düsseldorf, Germany, and Department of Internal Medicine 3-Rheumatology and Immunology and Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander University Erlangen-Nürnberg (FAU), and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Evgenia Boxberger
- Department of Internal Medicine 3-Rheumatology and Immunology and Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Despina Soteriou
- Max Planck Institute for the Science of Light & Max-Planck-Center für Physik und Medizin, Erlangen, Germany
| | - Maria Papava
- Department of Internal Medicine 3-Rheumatology and Immunology and Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Julia Prater
- Department of Internal Medicine 3-Rheumatology and Immunology and Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Xuezhi Hong
- Department of Rheumatology and Hiller Research Unit, University Hospital Düsseldorf, Medical Faculty of Heinrich Heine University, Düsseldorf, Germany, and Department of Internal Medicine 3-Rheumatology and Immunology and Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander University Erlangen-Nürnberg (FAU), and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Christina Bergmann
- Department of Internal Medicine 3-Rheumatology and Immunology and Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Martin Kräter
- Max Planck Institute for the Science of Light & Max-Planck-Center für Physik und Medizin, Erlangen, Germany, and Biotechnology Center, Center for Molecular and Cellular Bioengineering, Technische Universität Dresden, Dresden, Germany
| | - Georg Schett
- Department of Internal Medicine 3-Rheumatology and Immunology and Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Jochen Guck
- Max Planck Institute for the Science of Light & Max-Planck-Center für Physik und Medizin, Erlangen, Germany, and Biotechnology Center, Center for Molecular and Cellular Bioengineering, Technische Universität Dresden, Dresden, Germany
| | - Jörg H W Distler
- Department of Rheumatology and Hiller Research Unit, University Hospital Düsseldorf, Medical Faculty of Heinrich Heine University, Düsseldorf, Germany, and Department of Internal Medicine 3-Rheumatology and Immunology and Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander University Erlangen-Nürnberg (FAU), and Universitätsklinikum Erlangen, Erlangen, Germany
| |
Collapse
|
10
|
van Eeden C, Osman MS, Cohen Tervaert JW. Fatigue in ANCA-associated vasculitis (AAV) and systemic sclerosis (SSc): similarities with Myalgic encephalomyelitis/Chronic Fatigue Syndrome (ME/CFS). A critical review of the literature. Expert Rev Clin Immunol 2022; 18:1049-1070. [PMID: 36045606 DOI: 10.1080/1744666x.2022.2116002] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Persistent debilitating fatigue is a frequent complaint in patients with systemic autoimmune rheumatic diseases (SARDs). Fatigue is, however, frequently overlooked in the clinic, and patients who successfully achieve remission of their disease, often still have a lowered quality of life due to its persistence. How similar is this fatigue to Myalgic encephalomyelitis/Chronic Fatigue Syndrome (ME/CFS), what is this fatigue associated with, and what tools/approaches (if any), have resulted in the improvement of fatigue in these patients is poorly defined. AREAS COVERED Similarities between the pathophysiology of ME/CFS, systemic sclerosis (SSc) and primary systemic vasculitides (PSV) are discussed, followed by an in-depth review of the prevalence and correlates of fatigue in these diseases. The authors reviewed literature from MEDLINE, APA PsycInfo, Embase, and CINAHL. EXPERT OPINION Persistent fatigue is a prominent feature in SARDs and may not be associated with components commonly associated with disease activity and/or progression. Immune and metabolic commonalities exist between ME/CFS, SSc, and PSVs - suggesting that common pathways inherent to the diseases and fatigue may be present. We suggest that patients with features of ME/CFS need to be identified by treating physicians, as they may require alternative approaches to therapy to improve their quality of life.
Collapse
Affiliation(s)
- Charmaine van Eeden
- Division of Rheumatology, Department of Medicine, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Mohammed S Osman
- Division of Rheumatology, Department of Medicine, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Jan Willem Cohen Tervaert
- Division of Rheumatology, Department of Medicine, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada.,School for Mental Health and Neurosciences (MHeNs), Maastricht University, Maastricht, Netherlands
| |
Collapse
|
11
|
Hojjatipour T, Aslani S, Salimifard S, Mikaeili H, Hemmatzadeh M, Gholizadeh Navashenaq J, Ahangar Parvin E, Jadidi-Niaragh F, Mohammadi H. NK cells - Dr. Jekyll and Mr. Hyde in autoimmune rheumatic diseases. Int Immunopharmacol 2022; 107:108682. [DOI: 10.1016/j.intimp.2022.108682] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 02/28/2022] [Accepted: 03/02/2022] [Indexed: 02/07/2023]
|
12
|
Gur C, Wang SY, Sheban F, Zada M, Li B, Kharouf F, Peleg H, Aamar S, Yalin A, Kirschenbaum D, Braun-Moscovici Y, Jaitin DA, Meir-Salame T, Hagai E, Kragesteen BK, Avni B, Grisariu S, Bornstein C, Shlomi-Loubaton S, David E, Shreberk-Hassidim R, Molho-Pessach V, Amar D, Tzur T, Kuint R, Gross M, Barboy O, Moshe A, Fellus-Alyagor L, Hirsch D, Addadi Y, Erenfeld S, Biton M, Tzemach T, Elazary A, Naparstek Y, Tzemach R, Weiner A, Giladi A, Balbir-Gurman A, Amit I. LGR5 expressing skin fibroblasts define a major cellular hub perturbed in scleroderma. Cell 2022; 185:1373-1388.e20. [PMID: 35381199 PMCID: PMC7612792 DOI: 10.1016/j.cell.2022.03.011] [Citation(s) in RCA: 76] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 12/26/2021] [Accepted: 03/09/2022] [Indexed: 11/28/2022]
Abstract
Systemic sclerosis (scleroderma, SSc) is an incurable autoimmune disease with high morbidity and mortality rates. Here, we conducted a population-scale single-cell genomic analysis of skin and blood samples of 56 healthy controls and 97 SSc patients at different stages of the disease. We found immune compartment dysfunction only in a specific subtype of diffuse SSc patients but global dysregulation of the stromal compartment, particularly in a previously undefined subset of LGR5+-scleroderma-associated fibroblasts (ScAFs). ScAFs are perturbed morphologically and molecularly in SSc patients. Single-cell multiome profiling of stromal cells revealed ScAF-specific markers, pathways, regulatory elements, and transcription factors underlining disease development. Systematic analysis of these molecular features with clinical metadata associates specific ScAF targets with disease pathogenesis and SSc clinical traits. Our high-resolution atlas of the sclerodermatous skin spectrum will enable a paradigm shift in the understanding of SSc disease and facilitate the development of biomarkers and therapeutic strategies.
Collapse
Affiliation(s)
- Chamutal Gur
- Department of Systems Immunology, Weizmann Institute, Rehovot, Israel; Rheumatology Department, Hadassah Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Israel
| | - Shuang-Yin Wang
- Department of Systems Immunology, Weizmann Institute, Rehovot, Israel.
| | - Fadi Sheban
- Department of Systems Immunology, Weizmann Institute, Rehovot, Israel
| | - Mor Zada
- Department of Systems Immunology, Weizmann Institute, Rehovot, Israel
| | - Baoguo Li
- Department of Systems Immunology, Weizmann Institute, Rehovot, Israel
| | - Fadi Kharouf
- Rheumatology Department, Hadassah Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Israel
| | - Hagit Peleg
- Rheumatology Department, Hadassah Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Israel
| | - Suhail Aamar
- Rheumatology Department, Hadassah Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Israel
| | - Adam Yalin
- Department of Systems Immunology, Weizmann Institute, Rehovot, Israel
| | | | - Yolanda Braun-Moscovici
- Rheumatology Institute, Rambam Health Care Campus, Rappaport Faculty of Medicine, Technion-Israeli Institute of Technology, Haifa, Israel
| | | | - Tomer Meir-Salame
- Flow Cytometry Unit, Department of Biological Services, Weizmann Institute, Rehovot, Israel
| | - Efrat Hagai
- Flow Cytometry Unit, Department of Biological Services, Weizmann Institute, Rehovot, Israel
| | | | - Batia Avni
- Department of Bone Marrow Transplantation, Hadassah Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Israel
| | - Sigal Grisariu
- Department of Bone Marrow Transplantation, Hadassah Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Israel
| | | | | | - Eyal David
- Department of Systems Immunology, Weizmann Institute, Rehovot, Israel
| | - Rony Shreberk-Hassidim
- Dermatology Department, Hadassah Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Israel
| | - Vered Molho-Pessach
- Dermatology Department, Hadassah Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Israel
| | - Dalit Amar
- Plastic Surgery Department, Hadassah Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Israel
| | - Tomer Tzur
- Plastic Surgery Department, Hadassah Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Israel
| | - Rottem Kuint
- Institue of Pulmonology Medicine, Hadassah Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Israel
| | - Moshe Gross
- Orthopedic Department, Hadassah Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Israel
| | - Oren Barboy
- Department of Systems Immunology, Weizmann Institute, Rehovot, Israel
| | - Adi Moshe
- Department of Systems Immunology, Weizmann Institute, Rehovot, Israel
| | | | - Dana Hirsch
- The Department of Veterinary Resources, Weizmann Institute, Rehovot, Israel
| | - Yoseph Addadi
- Life Sciences Core Facilities, Weizmann Institute, Rehovot, Israel
| | - Shlomit Erenfeld
- Department of Bone Marrow Transplantation, Hadassah Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Israel
| | - Moshe Biton
- Department of Biological Regulation, Weizmann Institute, Rehovot, Israel
| | - Tehila Tzemach
- Rheumatology Department, Hadassah Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Israel
| | - Anat Elazary
- Rheumatology Department, Hadassah Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Israel
| | - Yaakov Naparstek
- Rheumatology Department, Hadassah Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Israel
| | - Reut Tzemach
- Department of Systems Immunology, Weizmann Institute, Rehovot, Israel; Rheumatology Institute at the Tel Aviv Sourasky Medical Center, Tel Aviv-Yafo, Israel
| | - Assaf Weiner
- Department of Systems Immunology, Weizmann Institute, Rehovot, Israel
| | - Amir Giladi
- Department of Systems Immunology, Weizmann Institute, Rehovot, Israel
| | - Alexandra Balbir-Gurman
- Rheumatology Institute, Rambam Health Care Campus, Rappaport Faculty of Medicine, Technion-Israeli Institute of Technology, Haifa, Israel
| | - Ido Amit
- Department of Systems Immunology, Weizmann Institute, Rehovot, Israel.
| |
Collapse
|
13
|
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: 59] [Impact Index Per Article: 19.7] [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
|
14
|
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
|
15
|
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: 1.5] [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
|
16
|
Liu M, Liang S, Zhang C. NK Cells in Autoimmune Diseases: Protective or Pathogenic? Front Immunol 2021; 12:624687. [PMID: 33777006 PMCID: PMC7994264 DOI: 10.3389/fimmu.2021.624687] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Accepted: 02/22/2021] [Indexed: 12/12/2022] Open
Abstract
Autoimmune diseases generally result from the loss of self-tolerance (i.e., failure of the immune system to distinguish self from non-self), and are characterized by autoantibody production and hyperactivation of T cells, which leads to damage of specific or multiple organs. Thus, autoimmune diseases can be classified as organ-specific or systemic. Genetic and environmental factors contribute to the development of autoimmunity. Recent studies have demonstrated the contribution of innate immunity to the onset of autoimmune diseases. Natural killer (NK) cells, which are key components of the innate immune system, have been implicated in the development of multiple autoimmune diseases such as systemic lupus erythematosus, type I diabetes mellitus, and autoimmune liver disease. However, NK cells have both protective and pathogenic roles in autoimmunity depending on the NK cell subset, microenvironment, and disease type or stage. In this work, we review the current knowledge of the varied roles of NK cell subsets in systemic and organic-specific autoimmune diseases and their clinical potential as therapeutic targets.
Collapse
Affiliation(s)
- Meifang Liu
- Key Lab for Immunology in Universities of Shandong Province, School of Basic Medical Sciences, Weifang Medical University, Weifang, China
| | - Shujuan Liang
- Key Lab for Immunology in Universities of Shandong Province, School of Basic Medical Sciences, Weifang Medical University, Weifang, China
| | - Cai Zhang
- School of Pharmaceutical Sciences, Cheeloo College of Medicine, Institute of Immunopharmaceutical Sciences, Shandong University, Jinan, China
| |
Collapse
|
17
|
Leleu D, Levionnois E, Laurent P, Lazaro E, Richez C, Duffau P, Blanco P, Sisirak V, Contin-Bordes C, Truchetet ME. Elevated Circulatory Levels of Microparticles Are Associated to Lung Fibrosis and Vasculopathy During Systemic Sclerosis. Front Immunol 2020; 11:532177. [PMID: 33193304 PMCID: PMC7645042 DOI: 10.3389/fimmu.2020.532177] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Accepted: 10/01/2020] [Indexed: 12/25/2022] Open
Abstract
Background Microparticles (MPs) are vesicular structures that derive from multiple cellular sources. MPs play important roles in intercellular communication, regulation of cell signaling or initiation of enzymatic processes. While MPs were characterized in Systemic Sclerosis (SSc) patients, their contribution to SSc pathogenesis remains unknown. Our aim was to investigate the potential role of MPs in SSc pathophysiology and their impact on tissue fibrosis. Methods Ninety-six SSc patients and 37 sex-matched healthy donors (HD) were enrolled in this study in order to quantify and phenotype their plasmatic MPs by flow cytometry. The ability of MPs purified from SSc patients and HD controls to modulate fibroblast's extra-cellular matrix genes expression was evaluated in vitro by reverse transcriptase quantitative polymerase chain reaction. Results SSc patients exhibited a higher concentration of circulatory MPs compared to HD. This difference was exacerbated when we only considered patients that were not treated with methotrexate or targeted disease-modifying antirheumatic drugs. Total circulatory MPs were associated to interstitial lung disease, lung fibrosis and diminished lung functional capacity, but also to vascular involvement such as active digital ulcers. Finally, contrary to HD MPs, MPs from SSc patients stimulated the production of extracellular matrix by fibroblast, demonstrating their profibrotic potential. Conclusions In this study, we provide evidence for a direct profibrotic role of MPs from SSc patients, underpinned by strong clinical associations in a large cohort of patients.
Collapse
Affiliation(s)
- Damien Leleu
- University of Bordeaux, CNRS, ImmunoConcEpT, UMR 5164, Bordeaux, France
- Immunology and Immunogenetic Department, Bordeaux University Hospital, Bordeaux, France
| | | | - Paoline Laurent
- University of Bordeaux, CNRS, ImmunoConcEpT, UMR 5164, Bordeaux, France
| | - Estibaliz Lazaro
- University of Bordeaux, CNRS, ImmunoConcEpT, UMR 5164, Bordeaux, France
- Internal Medicine Department, Bordeaux University Hospital, Bordeaux, France
- Centre national de reference des maladies auto-immunes systémiques rares de l’Est et du Sud-Ouest (RESO), Bordeaux, France
| | - Christophe Richez
- University of Bordeaux, CNRS, ImmunoConcEpT, UMR 5164, Bordeaux, France
- Centre national de reference des maladies auto-immunes systémiques rares de l’Est et du Sud-Ouest (RESO), Bordeaux, France
- Rheumatology Department, Bordeaux University Hospital, Bordeaux, France
| | - Pierre Duffau
- University of Bordeaux, CNRS, ImmunoConcEpT, UMR 5164, Bordeaux, France
- Internal Medicine Department, Bordeaux University Hospital, Bordeaux, France
| | - Patrick Blanco
- University of Bordeaux, CNRS, ImmunoConcEpT, UMR 5164, Bordeaux, France
- Immunology and Immunogenetic Department, Bordeaux University Hospital, Bordeaux, France
| | - Vanja Sisirak
- University of Bordeaux, CNRS, ImmunoConcEpT, UMR 5164, Bordeaux, France
| | - Cecile Contin-Bordes
- University of Bordeaux, CNRS, ImmunoConcEpT, UMR 5164, Bordeaux, France
- Immunology and Immunogenetic Department, Bordeaux University Hospital, Bordeaux, France
| | - Marie-Elise Truchetet
- University of Bordeaux, CNRS, ImmunoConcEpT, UMR 5164, Bordeaux, France
- Centre national de reference des maladies auto-immunes systémiques rares de l’Est et du Sud-Ouest (RESO), Bordeaux, France
- Rheumatology Department, Bordeaux University Hospital, Bordeaux, France
| |
Collapse
|
18
|
Tsai CY, Hsieh SC, Wu TH, Li KJ, Shen CY, Liao HT, Wu CH, Kuo YM, Lu CS, Yu CL. Pathogenic Roles of Autoantibodies and Aberrant Epigenetic Regulation of Immune and Connective Tissue Cells in the Tissue Fibrosis of Patients with Systemic Sclerosis. Int J Mol Sci 2020; 21:ijms21093069. [PMID: 32349208 PMCID: PMC7246753 DOI: 10.3390/ijms21093069] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 04/19/2020] [Accepted: 04/22/2020] [Indexed: 12/13/2022] Open
Abstract
Systemic sclerosis (SSc) is a multi-system autoimmune disease with tissue fibrosis prominent in the skin and lung. In this review, we briefly describe the autoimmune features (mainly autoantibody production and cytokine profiles) and the potential pathogenic contributors including genetic/epigenetic predisposition, and environmental factors. We look in detail at the cellular and molecular bases underlying tissue-fibrosis which include trans-differentiation of fibroblasts (FBs) to myofibroblasts (MFBs). We also state comprehensively the pro-inflammatory and pro-fibrotic cytokines relevant to MFB trans-differentiation, vasculopathy-associated autoantibodies, and fibrosis-regulating microRNAs in SSc. It is conceivable that tissue fibrosis is mainly mediated by an excessive production of TGF-β, the master regulator, from the skewed Th2 cells, macrophages, fibroblasts, myofibroblasts, and keratinocytes. After binding with TGF-β receptors on MFB, the downstream Wnt/β-catenin triggers canonical Smad 2/3 and non-canonical Smad 4 signaling pathways to transcribe collagen genes. Subsequently, excessive collagen fiber synthesis and accumulation as well as tissue fibrosis ensue. In the later part of this review, we discuss limited data relevant to the role of long non-coding RNAs (lncRNAs) in tissue-fibrosis in SSc. It is expected that these lncRNAs may become the useful biomarkers and therapeutic targets for SSc in the future. The prospective investigations in the development of novel epigenetic modifiers are also suggested.
Collapse
Affiliation(s)
- Chang-Youh Tsai
- Division of Allergy, Immunology & Rheumatology, Taipei Veterans General Hospital & National Yang-Ming University, #201 Sec. 2, Shih-Pai Road, Taipei 11217, Taiwan;
- Correspondence: (C.-Y.T.); (C.-L.Y.); Fax: +886-2-28717483 (C.-Y.T.); +886-2-23957801 (C.-L.Y.)
| | - Song-Chou Hsieh
- Department of Internal Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, #7 Chung-Shan South Road, Taipei 10002, Taiwan; (S.-C.H.); (K.-J.L.); (C.-Y.S.); (C.-H.W.); (Y.-M.K.); (C.-S.L.)
| | - Tsai-Hung Wu
- Division of Nephrology, Taipei Veterans General Hospital & National Yang-Ming University, #201 Sec. 2, Shih-Pai Road, Taipei 11217, Taiwan;
| | - Ko-Jen Li
- Department of Internal Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, #7 Chung-Shan South Road, Taipei 10002, Taiwan; (S.-C.H.); (K.-J.L.); (C.-Y.S.); (C.-H.W.); (Y.-M.K.); (C.-S.L.)
| | - Chieh-Yu Shen
- Department of Internal Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, #7 Chung-Shan South Road, Taipei 10002, Taiwan; (S.-C.H.); (K.-J.L.); (C.-Y.S.); (C.-H.W.); (Y.-M.K.); (C.-S.L.)
- Institute of Clinical Medicine, National Taiwan University College of Medicine, #7 Chung-Shan South Road, Taipei 10002, Taiwan
| | - Hsien-Tzung Liao
- Division of Allergy, Immunology & Rheumatology, Taipei Veterans General Hospital & National Yang-Ming University, #201 Sec. 2, Shih-Pai Road, Taipei 11217, Taiwan;
| | - Cheng-Han Wu
- Department of Internal Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, #7 Chung-Shan South Road, Taipei 10002, Taiwan; (S.-C.H.); (K.-J.L.); (C.-Y.S.); (C.-H.W.); (Y.-M.K.); (C.-S.L.)
- Institute of Clinical Medicine, National Taiwan University College of Medicine, #7 Chung-Shan South Road, Taipei 10002, Taiwan
| | - Yu-Min Kuo
- Department of Internal Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, #7 Chung-Shan South Road, Taipei 10002, Taiwan; (S.-C.H.); (K.-J.L.); (C.-Y.S.); (C.-H.W.); (Y.-M.K.); (C.-S.L.)
- Institute of Clinical Medicine, National Taiwan University College of Medicine, #7 Chung-Shan South Road, Taipei 10002, Taiwan
| | - Cheng-Shiun Lu
- Department of Internal Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, #7 Chung-Shan South Road, Taipei 10002, Taiwan; (S.-C.H.); (K.-J.L.); (C.-Y.S.); (C.-H.W.); (Y.-M.K.); (C.-S.L.)
- Institute of Clinical Medicine, National Taiwan University College of Medicine, #7 Chung-Shan South Road, Taipei 10002, Taiwan
| | - Chia-Li Yu
- Department of Internal Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, #7 Chung-Shan South Road, Taipei 10002, Taiwan; (S.-C.H.); (K.-J.L.); (C.-Y.S.); (C.-H.W.); (Y.-M.K.); (C.-S.L.)
- Correspondence: (C.-Y.T.); (C.-L.Y.); Fax: +886-2-28717483 (C.-Y.T.); +886-2-23957801 (C.-L.Y.)
| |
Collapse
|
19
|
Karimizadeh E, Mostafaei S, Aslani S, Gharibdoost F, Xavier RM, Salim PH, Kavosi H, Farhadi E, Mahmoudi M. Evaluation of the association between KIR polymorphisms and systemic sclerosis: a meta-analysis. Adv Rheumatol 2020; 60:8. [PMID: 31924272 DOI: 10.1186/s42358-019-0107-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2018] [Accepted: 12/23/2019] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND The results of investigations on the association between killer cell immunoglobulin-like receptor (KIR) gene polymorphisms and the risk of systemic sclerosis (SSc) are inconsistent. To comprehensively evaluate the influence of KIR polymorphisms on the risk of SSc, this meta-analysis was performed. METHODS A systematic literature search was performed in electronic databases including Scopus and PubMed/MEDLINE to find all available studies involving KIR gene family polymorphisms and SSc risk prior to July 2019. Pooled odds ratios (ORs) and their corresponding 95% confidence intervals (CIs) were measured to detect associations between KIR gene family polymorphisms and SSc risk. RESULTS Five articles, comprising 571 patients and 796 healthy participants, evaluating the KIR gene family polymorphisms were included in the final meta-analysis according to the inclusion and exclusion criteria, and 16 KIR genes were assessed. None of the KIR genes were significantly associated with the risk of SSc. CONCLUSIONS The current meta-analysis provides evidence that KIR genes might not be potential risk factors for SSc risk.
Collapse
Affiliation(s)
- Elham Karimizadeh
- Rheumatology Research Center, Tehran University of Medical Sciences, Shariati Hospital, Kargar Ave, Tehran, Iran
| | - Shayan Mostafaei
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Saeed Aslani
- Rheumatology Research Center, Tehran University of Medical Sciences, Shariati Hospital, Kargar Ave, Tehran, Iran
| | - Farhad Gharibdoost
- Rheumatology Research Center, Tehran University of Medical Sciences, Shariati Hospital, Kargar Ave, Tehran, Iran
| | - Ricardo Machado Xavier
- Universidade Federal do Rio Grande do Sul, Serviço de Reumatologia, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Patricia Hartstein Salim
- Universidade Federal do Rio Grande do Sul, Serviço de Reumatologia, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Hoda Kavosi
- Rheumatology Research Center, Tehran University of Medical Sciences, Shariati Hospital, Kargar Ave, Tehran, Iran
| | - Elham Farhadi
- Rheumatology Research Center, Tehran University of Medical Sciences, Shariati Hospital, Kargar Ave, Tehran, Iran. .,Inflammation Research Center, Tehran University of Medical Sciences, Tehran, Iran.
| | - Mahdi Mahmoudi
- Rheumatology Research Center, Tehran University of Medical Sciences, Shariati Hospital, Kargar Ave, Tehran, Iran. .,Inflammation Research Center, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
20
|
Asano Y, Varga J. Rationally-based therapeutic disease modification in systemic sclerosis: Novel strategies. Semin Cell Dev Biol 2019; 101:146-160. [PMID: 31859147 DOI: 10.1016/j.semcdb.2019.12.007] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 12/12/2019] [Accepted: 12/12/2019] [Indexed: 02/07/2023]
Abstract
Systemic sclerosis (SSc) is a highly challenging chronic condition that is dominated by the pathogenetic triad of vascular damage, immune dysregulation/autoimmunity and fibrosis in multiple organs. A hallmark of SSc is the remarkable degree of molecular and phenotypic disease heterogeneity, which surpasses that of other complex rheumatic diseases. Disease trajectories in SSc are unpredictable and variable from patient to patient. Disease-modifying therapies for SSc are lacking, long-term morbidity is considerable and mortality remains unacceptably high. Currently-used empirical approaches to disease modification have modest and variable clinical efficacy and impact on survival, are expensive and frequently associated with unfavorable side effects, and none can be considered curative. However, research during the past several years is yielding significant advances with therapeutic potential. In particular, the application of unbiased omics-based discovery technologies to large and well-characterized SSc patient cohorts, coupled with hypothesis-testing experimental research using a variety of model systems is revealing new insights into SSc that allow formulation of a more nuanced appreciation of disease heterogeneity, and a deepening understanding of pathogenesis. Indeed, we are now presented with numerous novel and rationally-based strategies for targeted SSc therapy, several of which are currently, or expected to be shortly, undergoing clinical evaluation. In this review, we discuss promising novel therapeutic targets and rationally-based approaches to disease modification that have the potential to improve long-term outcomes in SSc.
Collapse
Affiliation(s)
| | - John Varga
- Northwestern Scleroderma Program, Feinberg School of Medicine, Northwestern University, Chicago, United States.
| |
Collapse
|
21
|
López-Isac E, Acosta-Herrera M, Kerick M, Assassi S, Satpathy AT, Granja J, Mumbach MR, Beretta L, Simeón CP, Carreira P, Ortego-Centeno N, Castellvi I, Bossini-Castillo L, Carmona FD, Orozco G, Hunzelmann N, Distler JHW, Franke A, Lunardi C, Moroncini G, Gabrielli A, de Vries-Bouwstra J, Wijmenga C, Koeleman BPC, Nordin A, Padyukov L, Hoffmann-Vold AM, Lie B, Proudman S, Stevens W, Nikpour M, Vyse T, Herrick AL, Worthington J, Denton CP, Allanore Y, Brown MA, Radstake TRDJ, Fonseca C, Chang HY, Mayes MD, Martin J. GWAS for systemic sclerosis identifies multiple risk loci and highlights fibrotic and vasculopathy pathways. Nat Commun 2019; 10:4955. [PMID: 31672989 PMCID: PMC6823490 DOI: 10.1038/s41467-019-12760-y] [Citation(s) in RCA: 121] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Accepted: 09/30/2019] [Indexed: 12/12/2022] Open
Abstract
Systemic sclerosis (SSc) is an autoimmune disease that shows one of the highest mortality rates among rheumatic diseases. We perform a large genome-wide association study (GWAS), and meta-analysis with previous GWASs, in 26,679 individuals and identify 27 independent genome-wide associated signals, including 13 new risk loci. The novel associations nearly double the number of genome-wide hits reported for SSc thus far. We define 95% credible sets of less than 5 likely causal variants in 12 loci. Additionally, we identify specific SSc subtype-associated signals. Functional analysis of high-priority variants shows the potential function of SSc signals, with the identification of 43 robust target genes through HiChIP. Our results point towards molecular pathways potentially involved in vasculopathy and fibrosis, two main hallmarks in SSc, and highlight the spectrum of critical cell types for the disease. This work supports a better understanding of the genetic basis of SSc and provides directions for future functional experiments.
Collapse
Affiliation(s)
- Elena López-Isac
- Institute of Parasitology and Biomedicine López-Neyra, IPBLN-CSIC, Granada, Spain.
| | | | - Martin Kerick
- Institute of Parasitology and Biomedicine López-Neyra, IPBLN-CSIC, Granada, Spain
| | - Shervin Assassi
- The University of Texas Health Science Center-Houston, Houston, USA
| | - Ansuman T Satpathy
- Center for Personal Dynamic Regulomes, Stanford University School of Medicine, Stanford, CA, USA
- Howard Hughes Medical Institute, Stanford University, Stanford, CA, USA
| | - Jeffrey Granja
- Center for Personal Dynamic Regulomes, Stanford University School of Medicine, Stanford, CA, USA
- Howard Hughes Medical Institute, Stanford University, Stanford, CA, USA
| | - Maxwell R Mumbach
- Center for Personal Dynamic Regulomes, Stanford University School of Medicine, Stanford, CA, USA
- Howard Hughes Medical Institute, Stanford University, Stanford, CA, USA
| | - Lorenzo Beretta
- Referral Center for Systemic Autoimmune Diseases, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico di Milano, Milan, Italy
| | - Carmen P Simeón
- Department of Internal Medicine, Valle de Hebrón Hospital, Barcelona, Spain
| | - Patricia Carreira
- Department of Rheumatology, 12 de Octubre University Hospital, Madrid, Spain
| | | | - Ivan Castellvi
- Department of Rheumatology, Santa Creu i Sant Pau University Hospital, Barcelona, Spain
| | | | - F David Carmona
- Department of Genetics and Institute of Biotechnology, University of Granada, Granada, Spain
| | - Gisela Orozco
- Arthritis Research UK Centre for Genetics and Genomics, Centre for Musculoskeletal Research, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, Oxford Road, Manchester, UK
| | | | - Jörg H W Distler
- Department of Internal Medicine 3, Institute for Clinical Immunology, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Andre Franke
- Institute of Clinical Molecular Biology, Christian-Albrechts-University of Kiel, Kiel, Germany
| | - Claudio Lunardi
- Department of Medicine, Università degli Studi di Verona, Verona, Italy
| | - Gianluca Moroncini
- Clinica Medica, Department of Clinical and Molecular Science, Università Politecnica delle Marche and Ospedali Riuniti, Ancona, Italy
| | - Armando Gabrielli
- Clinica Medica, Department of Clinical and Molecular Science, Università Politecnica delle Marche and Ospedali Riuniti, Ancona, Italy
| | | | - Cisca Wijmenga
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | | | - Annika Nordin
- Division of Rheumatology, Department of Medicine, Karolinska University Hospital, Karolinska Institute, Stockholm, Sweden
| | - Leonid Padyukov
- Division of Rheumatology, Department of Medicine, Karolinska University Hospital, Karolinska Institute, Stockholm, Sweden
| | | | - Benedicte Lie
- Department of Medical Genetics, and the Department of Immunology, University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Susanna Proudman
- Royal Adelaide Hospital and University of Adelaide, Adelaide, SA, Australia
| | | | - Mandana Nikpour
- The University of Melbourne at St. Vincent's Hospital, Melbourne, VIC, Australia
| | - Timothy Vyse
- Department of Medical and Molecular Genetics, King's College London, London, UK
| | - Ariane L Herrick
- Centre for Musculoskeletal Research, The University of Manchester, Salford Royal NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, UK
- NIHR Manchester Biomedical Research Centre, Manchester, UK
| | - Jane Worthington
- Arthritis Research UK Centre for Genetics and Genomics, Centre for Musculoskeletal Research, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, Oxford Road, Manchester, UK
| | - Christopher P Denton
- Centre for Rheumatology, Royal Free and University College Medical School, London, United Kingdom
| | - Yannick Allanore
- Department of Rheumatology A, Cochin Hospital, INSERM U1016, Paris Descartes University, Paris, France
| | - Matthew A Brown
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Translational Research Institute, Princess Alexandra Hospital, Brisbane, QLD, Australia
| | - Timothy R D J Radstake
- Department of Rheumatology & Clinical Immunology, Laboratory of Translational Immunology, department of Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Carmen Fonseca
- Centre for Rheumatology, Royal Free and University College Medical School, London, United Kingdom
| | - Howard Y Chang
- Center for Personal Dynamic Regulomes, Stanford University School of Medicine, Stanford, CA, USA
- Howard Hughes Medical Institute, Stanford University, Stanford, CA, USA
| | - Maureen D Mayes
- The University of Texas Health Science Center-Houston, Houston, USA
| | - Javier Martin
- Institute of Parasitology and Biomedicine López-Neyra, IPBLN-CSIC, Granada, Spain.
| |
Collapse
|
22
|
Brown M, O'Reilly S. The immunopathogenesis of fibrosis in systemic sclerosis. Clin Exp Immunol 2018; 195:310-321. [PMID: 30430560 DOI: 10.1111/cei.13238] [Citation(s) in RCA: 119] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/07/2018] [Indexed: 02/07/2023] Open
Abstract
Systemic sclerosis (SSc) is an idiopathic systemic autoimmune disease. It is characterized by a triad of hallmarks: immune dysfunction, fibrosis and vasculopathy. Immune dysfunction in SSc is characterized by the activation and recruitment of immune cells and the production of autoantibodies and cytokines. How immune abnormalities link the fibrosis and vasculopathy in SSc is poorly understood. A plethora of immune cell types are implicated in the immunopathogenesis of SSc, including T cells, B cells, dendritic cells, mast cells and macrophages. How these different cell types interact to contribute to SSc is complicated, and can involve cell-to-cell interactions and communication via cytokines, including transforming growth factor (TGF)-β, interleukin (IL)-6 and IL-4. We will attempt to review significant and recent research demonstrating the importance of immune cell regulation in the immunopathogenesis of SSc with a particular focus on fibrosis.
Collapse
Affiliation(s)
- M Brown
- Faculty of Health and Life Sciences, Northumbria University, Newcastle upon Tyne, UK
| | - S O'Reilly
- Faculty of Health and Life Sciences, Northumbria University, Newcastle upon Tyne, UK
| |
Collapse
|