1
|
Kumar V, Hertz M, Agro A, Byrne AJ. Type 1 invariant natural killer T cells in chronic inflammation and tissue fibrosis. Front Immunol 2023; 14:1260503. [PMID: 37818376 PMCID: PMC10561218 DOI: 10.3389/fimmu.2023.1260503] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 09/06/2023] [Indexed: 10/12/2023] Open
Abstract
Chronic tissue inflammation often results in fibrosis characterized by the accumulation of extracellular matrix components remodeling normal tissue architecture and function. Recent studies have suggested common immune mechanisms despite the complexity of the interactions between tissue-specific fibroblasts, macrophages, and distinct immune cell populations that mediate fibrosis in various tissues. Natural killer T (NKT) cells recognizing lipid antigens bound to CD1d molecules have been shown to play an important role in chronic inflammation and fibrosis. Here we review recent data in both experimental models and in humans that suggest a key role of type 1 invariant NKT (iNKT) cell activation in the progression of inflammatory cascades leading to recruitment of neutrophils and activation of the inflammasome, macrophages, fibroblasts, and, ultimately, fibrosis. Emerging evidence suggests that iNKT-associated mechanisms contribute to type 1, type 2 and type 3 immune pathways mediating tissue fibrosis, including idiopathic pulmonary fibrosis (IPF). Thus, targeting a pathway upstream of these immune mechanisms, such as the inhibition of iNKT activation, may be important in modulating various fibrotic conditions.
Collapse
Affiliation(s)
- Vipin Kumar
- Laboratory of Immune Regulation, Department of Medicine, University of California San Diego, La Jolla, CA, United States
- GRI Bio, La Jolla, CA, United States
| | | | | | - Adam J. Byrne
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
- School of Medicine and Conway Institute for Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland
| |
Collapse
|
2
|
Colic J, Campochiaro C, Hughes M, Matucci Cerinic M, Dagna L. Investigational drugs for the treatment of scleroderma: what's new? Expert Opin Investig Drugs 2023; 32:601-614. [PMID: 37526079 DOI: 10.1080/13543784.2023.2242762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Accepted: 07/26/2023] [Indexed: 08/02/2023]
Abstract
INTRODUCTION Systemic sclerosis (SSc) is an orphan, chronic, autoimmune, fibrotic disease with unknown etiology characterized by progressive fibrosis of the skin and internal organs. SSc has the highest mortality, the deadliest among the connective tissue diseases, despite the introduction of new treatment options in the past decades. AREAS COVERED The aim of the current systematic review was to investigate new targeted therapy and their impact on disease progression, mainly focusing on phase I and II clinical trials within the past three years. EXPERT OPINION Despite recent groundbreaking advancements in understanding SSc pathophysiology, early diagnosis and early introduction of effective targeted treatments within the optimal window of opportunity to prevent irreversible disease damage still represents a significant clinical challenge. Ongoing significant research for new molecular and epigenetics pathways is of fundamental importance to offer new perspectives on disease phenotype and for the development of personalized treatment strategies.
Collapse
Affiliation(s)
- Jelena Colic
- Department of Rheumatology, Institute of Rheumatology, Belgrade, Serbia
| | - Corrado Campochiaro
- Unit of Immunology, Rheumatology, Allergy and Rare Diseases, IRCCS San Raffaele Hospital, Milano, Italy
- Vita-Salute San Raffaele University, Milano, Italy
| | - Michael Hughes
- Division of Musculoskeletal and Dermatological Sciences, The University of Manchester, Manchester, England
| | - Marco Matucci Cerinic
- Unit of Immunology, Rheumatology, Allergy and Rare Diseases, IRCCS San Raffaele Hospital, Milano, Italy
- Division of Rheumatology, Department of Experimental and Clinical Medicine, Azienda Ospedaliero-Universitaria Careggi (AOUC) and Denothe Centre, University of Florence, Florence, Italy
| | - Lorenzo Dagna
- Unit of Immunology, Rheumatology, Allergy and Rare Diseases, IRCCS San Raffaele Hospital, Milano, Italy
- Vita-Salute San Raffaele University, Milano, Italy
| |
Collapse
|
3
|
Pulmonary Fibrosis as a Result of Acute Lung Inflammation: Molecular Mechanisms, Relevant In Vivo Models, Prognostic and Therapeutic Approaches. Int J Mol Sci 2022; 23:ijms232314959. [PMID: 36499287 PMCID: PMC9735580 DOI: 10.3390/ijms232314959] [Citation(s) in RCA: 76] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 11/24/2022] [Accepted: 11/25/2022] [Indexed: 12/03/2022] Open
Abstract
Pulmonary fibrosis is a chronic progressive lung disease that steadily leads to lung architecture disruption and respiratory failure. The development of pulmonary fibrosis is mostly the result of previous acute lung inflammation, caused by a wide variety of etiological factors, not resolved over time and causing the deposition of fibrotic tissue in the lungs. Despite a long history of study and good coverage of the problem in the scientific literature, the effective therapeutic approaches for pulmonary fibrosis treatment are currently lacking. Thus, the study of the molecular mechanisms underlying the transition from acute lung inflammation to pulmonary fibrosis, and the search for new molecular markers and promising therapeutic targets to prevent pulmonary fibrosis development, remain highly relevant tasks. This review focuses on the etiology, pathogenesis, morphological characteristics and outcomes of acute lung inflammation as a precursor of pulmonary fibrosis; the pathomorphological changes in the lungs during fibrosis development; the known molecular mechanisms and key players of the signaling pathways mediating acute lung inflammation and pulmonary fibrosis, as well as the characteristics of the most common in vivo models of these processes. Moreover, the prognostic markers of acute lung injury severity and pulmonary fibrosis development as well as approved and potential therapeutic approaches suppressing the transition from acute lung inflammation to fibrosis are discussed.
Collapse
|
4
|
McGuire P, Glicksman C, Wixtrom R, Sung CJ, Hamilton R, Lawrence M, Haws M, Ferenz S, Kadin M. Microbes, Histology, Blood Analysis, Enterotoxins, and Cytokines: Findings From the ASERF Systemic Symptoms in Women-Biospecimen Analysis Study: Part 3. Aesthet Surg J 2022; 43:230-244. [PMID: 35980942 PMCID: PMC9896138 DOI: 10.1093/asj/sjac225] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 07/30/2022] [Accepted: 08/01/2022] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND There has been an increasing need to acquire rigorous scientific data to answer the concerns of physicians, patients, and the FDA regarding the self-reported illness identified as breast implant illness (BII). There are no diagnostic tests or specific laboratory values to explain the reported systemic symptoms described by these patients. OBJECTIVES The aim of this study was to determine if there are quantifiable laboratory findings that can be identified in blood, capsule tissue pathology, or microbes that differentiate women with systemic symptoms they attribute to their implants from 2 control groups. METHODS A prospective blinded study enrolled 150 subjects into 3 cohorts: (A) women with systemic symptoms they attribute to implants who requested implant removal; (B) women with breast implants requesting removal or exchange who did not have symptoms attributed to implants; and (C) women undergoing cosmetic mastopexy who have never had any implanted medical device. Capsule tissue underwent detailed analysis and blood was sent from all 3 cohorts to evaluate for markers of inflammation. RESULTS No significant histologic differences were identified between the cohorts, except there were more capsules with synovial metaplasia in the non-BII cohort. There was no statistical difference in thyroid-stimulating hormone, vitamin D levels, or complete blood count with differential between the cohorts. Next-generation sequencing revealed no statistically significant difference in positivity between Cohort A and B. Of the 12 cytokines measured, 3 cytokines, interleukin (IL)-17A, IL-13, and IL-22, were found to be significantly more often elevated in sera of subjects in Cohort A than in Cohorts B or C. The enterotoxin data demonstrated an elevation in immunoglobulin G (IgG) anti-Staphylococcus aureus enterotoxin A in Cohort A. There was no correlation between the presence of IgE or IgG anti-Staphylococcal antibody and a positive next-generation sequencing result. CONCLUSIONS This study adds to the current literature by demonstrating few identifiable biomedical markers to explain the systemic symptoms self-reported by patients with BII.
Collapse
Affiliation(s)
- Patricia McGuire
- Washington University, St Louis, MO, USA and a clinical editor for Aesthetic Surgery Journal
| | - Caroline Glicksman
- Corresponding Author: Dr Caroline Glicksman, 2164 State Highway 35, Building A, Sea Girt, NJ 08750, USA. E-mail:
| | | | - C James Sung
- Warren Alpert School of Medicine of Brown University, Providence, RI, USA
| | - Robert Hamilton
- Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | | | | | | - Marshall Kadin
- Brown University Alpert School of Medicine, Providence, RI, USA
| |
Collapse
|
5
|
Martin Calderon L, Pope JE. Precursors to Systemic Sclerosis and Systemic Lupus Erythematosus: From Undifferentiated Connective Tissue Disease to the Development of Identifiable Connective Tissue Diseases. Front Immunol 2022; 13:869172. [PMID: 35603174 PMCID: PMC9118990 DOI: 10.3389/fimmu.2022.869172] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Accepted: 04/05/2022] [Indexed: 11/13/2022] Open
Abstract
The pathogenesis of connective tissue diseases (CTDs), such as systemic lupus erythematosus (SLE) and systemic sclerosis (SSc), is characterized by derangements of the innate and adaptive immune system, and inflammatory pathways leading to autoimmunity, chronic cytokine production, and chronic inflammation. The diagnosis of these diseases is based on meeting established criteria with symptoms, signs and autoantibodies. However, there are pre-clinical states where criteria are not fulfilled but biochemical and autoimmune derangements are present. Understanding the underlying processes responsible for disease pathogenesis in pre-clinical states, which place patients at increased risk for the development of established connective tissue diseases, represents an opportunity for early identification and potentially enables timely treatment with the goal of limiting disease progression and improved prognosis. This scoping review describes the role of the innate and adaptive immune responses in the pre-clinical states of undifferentiated CTD at risk for SSc and prescleroderma, the evolution of antibodies from nonspecific to specific antinuclear antibodies prior to SLE development, and the signaling pathways and inflammatory markers of fibroblast, endothelial, and T cell activation underlying immune dysregulation in these pre-clinical states.
Collapse
Affiliation(s)
- Leonardo Martin Calderon
- Department of Medicine, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada
| | - Janet E Pope
- Division of Rheumatology, St. Joseph's Health Care, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada
| |
Collapse
|
6
|
Campochiaro C, Allanore Y. An update on targeted therapies in systemic sclerosis based on a systematic review from the last 3 years. Arthritis Res Ther 2021; 23:155. [PMID: 34074331 PMCID: PMC8168022 DOI: 10.1186/s13075-021-02536-5] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2020] [Accepted: 05/19/2021] [Indexed: 11/10/2022] Open
Abstract
New molecular mechanisms that can be targeted with specific drugs have recently emerged for the treatment of systemic sclerosis (SSc) patients. Over the past 3 years, the achievement of one large phase 3 trial has led to the approval by drug agencies of the first drug licenced for SSc-related interstitial lung disease. Given this exciting time in the SSc field, we aimed to perform a systemic literature review of phase 1, phase 2 and phase 3 clinical trials and large observational studies about targeted therapies in SSc. We searched MEDLINE/PubMed, EMBASE, and ClinicalTrials.gov for clinical studies from 2016 with targeted therapies as the primary treatment in patients with SSc for skin or lung involvement as the primary clinical outcome measure. Details on the study characteristics, the trial drug used, the molecular target engaged by the trial drug, the inclusion criteria of the study, the treatment dose, the possibility of concomitant immunosuppression, the endpoints of the study, the duration of the study and the results obtained were reviewed. Of the 973 references identified, 21 (4 conference abstracts and 17 articles) were included in the systematic review. A total of 15 phase 1/phase 2 clinical trials, 2 phase 3 clinical trials and 2 observation studies were analysed. The drugs studied in phase 1/phase 2 studies included the following: inebilizumab, dabigatran, C-82, pomalidomide, rilonacept, romilkimab, tocilizumab, tofacitinib, pirfenidone, lenabasum, abatacept, belimumab, riociguat, SAR100842 and lanifibranor. All but 3 studies were performed in early diffuse SSc patients with different inclusion criteria, while 3 studies were performed in SSc patients with interstitial lung disease (ILD). Phase 3 clinical trials investigated nintedanib and tocilizumab. Nintedanib was investigated in SSc-ILD patients whereas tocilizumab focused on early diffuse SSc patients with inflammatory features. Two observational studies including > 50 patients with rituximab as the targeted drug were also evaluated. All these studies offer a real hope for SSc patients. The future challenges will be to customize patient-specific therapeutics with the goal to develop precision medicine for SSc.
Collapse
Affiliation(s)
- Corrado Campochiaro
- Unit of Immunology, Rheumatology, Allergy and Rare Diseases (UnIRAR) IRCCS San Raffaele Hospital, Vita-Salute San Raffaele University, Via Olgettina 60, 20132, Milan, Italy
| | - Yannick Allanore
- Service de Rhumatologie, Hôpital Cochin, Université de Paris, 27 rue du Faubourg Saint-Jacques, 75014, Paris, France.
| |
Collapse
|
7
|
The Blockade of Tumoral IL1β-Mediated Signaling in Normal Colonic Fibroblasts Sensitizes Tumor Cells to Chemotherapy and Prevents Inflammatory CAF Activation. Int J Mol Sci 2021; 22:ijms22094960. [PMID: 34066976 PMCID: PMC8125420 DOI: 10.3390/ijms22094960] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 04/30/2021] [Accepted: 05/05/2021] [Indexed: 12/24/2022] Open
Abstract
Heterotypic interactions between newly transformed cells and normal surrounding cells define tumor’s fate in incipient carcinomas. Once homeostasis has been lost, normal resident fibroblasts become carcinoma-associated fibroblasts, conferring protumorogenic properties on these normal cells. Here we describe the IL1β-mediated interplay between cancer cells and normal colonic myofibroblasts (NCFs), which bestows differential sensitivity to cytotoxic drugs on tumor cells. We used NCFs, their conditioned media (CM), and cocultures with tumor cells to characterize the IL1β-mediated crosstalk between both cell types. We silenced IL1β in tumor cells to demonstrate that such cells do not exert an influence on NCFs inflammatory phenotype. Our results shows that IL1β is overexpressed in cocultured tumor cells. IL1β enables paracrine signaling in myofibroblasts, converting them into inflammatory-CAFs (iCAF). IL1β-stimulated-NCF-CM induces migration and differential sensitivity to oxaliplatin in colorectal tumor cells. Such chemoprotective effect has not been evidenced for TGFβ1-driven NCFs. IL1β induces the loss of a myofibroblastic phenotype in NCFs and acquisition of iCAF traits. In conclusion, IL1β-secreted by cancer cells modify surrounding normal fibroblasts to confer protumorogenic features on them, particularly tolerance to cytotoxic drugs. The use of IL1β-blocking agents might help to avoid the iCAF traits acquisition and consequently to counteract the protumorogenic actions these cells.
Collapse
|
8
|
Boleto G, Avouac J, Allanore Y. An update on recent randomized clinical trials in systemic sclerosis. Joint Bone Spine 2021; 88:105184. [PMID: 33887472 DOI: 10.1016/j.jbspin.2021.105184] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Accepted: 03/24/2021] [Indexed: 10/21/2022]
Abstract
Systemic sclerosis (SSc) is an autoimmune disorder characterized by a remarkable clinical heterogeneity and variable disease course. In this multifactorial condition, the interaction of several pathogenic pathways related to inflammation, vasculopathy and fibrosis promote organ damage. The understanding of the pathogenesis of SSc has improved in recent years leading to the identification of relevant therapeutic targets. Over the last few years, insightful clinical trials have been published and great progress has been made in developing effective therapeutic options. The positive results of the Safety and Efficacy of Nintedanib in Systemic SClerosIS (SENSCIS) trial have led to the approval by drug agencies of the first drug for SSc-related interstitial lung disease. However, the majority of clinical trials have focused on immunosuppressive therapy. Although benefit trends have been observed, outstanding results have not been achieved and to date no immunosuppressant has been approved in SSc. Clinical trial design has evolved regarding inclusion criteria with the aim to enrich for progressive patients. Endpoints have also been revised to better cover the fields of patients' feelings and functioning. Finally, background synthetic immunosuppressants have been allowed in some trials evaluating targeted therapies opening the door to combination therapies. Herewith, we provide an overview of the most significant clinical trials developed in recent years for the management of SSc.
Collapse
Affiliation(s)
- Gonçalo Boleto
- Department of Rheumatology, Université de Paris, Cochin Hospital, Paris, France
| | - Jérôme Avouac
- Department of Rheumatology, Université de Paris, Cochin Hospital, Paris, France; Inserm U1016, Institut Cochin, CNRS UMR8104, Paris, France
| | - Yannick Allanore
- Department of Rheumatology, Université de Paris, Cochin Hospital, Paris, France; Inserm U1016, Institut Cochin, CNRS UMR8104, Paris, France.
| |
Collapse
|
9
|
The Regenerative Potential of Donkey and Human Milk on the Redox-Sensitive and Proliferative Signaling Pathways of Skin Fibroblasts. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:5618127. [PMID: 33224432 PMCID: PMC7673926 DOI: 10.1155/2020/5618127] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 09/11/2020] [Accepted: 10/14/2020] [Indexed: 11/17/2022]
Abstract
The influence of milk bioactive peptides on skin regenerative potential and rejuvenation is very often limited because of allergic reactions. The current study is aimed at exploring the influence of donkey colostrum and mature milk, human colostrum and mature milk, and β-casein and β-casomorphine-7, on the growth and inflammatory response of the culture of cultured skin fibroblasts exposed to these conditions for twenty-four hours. Their effects on the growth-regulatory kinases and redox-sensitive, proinflammatory transcriptional factor NF-κB were detected by using specific primary antibodies against NF-κB p65, Akt-1, phospho-Akt-1, Erk-1, phospho-Erk-1, JNK, phospho-JNK, phospho-STAT-1, and CD26, while logarithmic integrated fluorescence intensity patterns were recorded by flow cytometry. The downregulation of NF-κB p65 was observed after the exposure of skin fibroblasts to donkey milk and human colostrum, while β-casein and β-casomorphine-7 exerted the opposite effect, which suggests that noncasein bioactive peptides of donkey and human milk may be responsible for anti-inflammatory properties. The exposure to all milk species examined and β-casein leads to the activation of growth-regulatory kinases (Akt1/2/3 kinase, Erk kinase, JNK kinase, and Stat-1 kinase), especially for the p-Erk pathway, which suggests that essential amino acids of casein may be responsible for Erk-induced cell cycle activation and proliferation. The opposite effect was observed when cells were exposed to β-casomorphine-7, which may affect the skin fibroblast survival and their proliferative and regenerative potential. Donkey milk did not significantly change the CD26 antigen expression. In conclusion, our results suggest that among cell signaling molecules, the most sensitive but nonspecific downstream effector is p-Erk kinase, which may point to donkey milk usefulness in wound healing, regenerative, and aesthetic dermatology. The noncasein bioactive peptides of donkey milk may be responsible for the anti-inflammatory property of donkey milk and colostrum, which may indicate the usefulness in the treatment of inflammatory skin diseases.
Collapse
|
10
|
Valentini G, Pope JE. Undifferentiated connective tissue disease at risk for systemic sclerosis: Which patients might be labeled prescleroderma? Autoimmun Rev 2020; 19:102659. [PMID: 32942034 DOI: 10.1016/j.autrev.2020.102659] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Accepted: 04/12/2020] [Indexed: 02/06/2023]
Abstract
Undifferentiated Connective Tissue Disease at risk for Systemic Sclerosis (UCTD-risk-SSc), otherwise referred to as very early-early SSc (very early-early diagnosis of systemic sclerosis VEDOSS), is a condition characterized by Raynaud's phenomenon (RP) and either SSc serum marker autoantibodies or a capillaroscopic scleroderma pattern or both, but without satisfying classification criteria for SSc neither features consistent with SSc sine scleroderma. Approximately half the UCTD-risk-SSc patients develop definite SSc over 5-10 years of follow-up. Identifying patients who will undergo such evolution is an unmet need. Predicting at onset which patients with RP are going to develop SSc over time has long been a research objective and still is an unaccomplished task. The present review is devoted to the critical analysis of the nosographic boundaries of the condition and of items predictive of evolution including serological, capillaroscopic and circulating markers. A weighted score, based on serum antinuclear antibody titre, serum marker antibodies positivity and avascular areas has been developed and may identify in the meanwhile patients to be labeled prescleroderma i.e. those probably developing SSc over time. Future research should be directed to investigate unexplored features, validate and improve the performance of the score and highlight the involved pathways to be contrasted in order to identify a targeted therapy hampering the development of overt SSc.
Collapse
Affiliation(s)
- Gabriele Valentini
- Università degli Studi della Campania "Luigi Vanvitelli", Department of Precision Medicine, Section of Rheumatology, Italy.
| | - Janet E Pope
- Università degli Studi della Campania "Luigi Vanvitelli", Department of Precision Medicine, Section of Rheumatology, Italy; Schulich School of Medicine and Dentistry, University of Western Ontario, Division of Rheumatology, St. Joseph's Health Care, London, Ontario, Canada
| |
Collapse
|
11
|
Arcangeletti MC, D’Accolti M, Maccari C, Soffritti I, Conto FD, Chezzi C, Calderaro A, Ferri C, Caselli E. Impact of Human Cytomegalovirus and Human Herpesvirus 6 Infection on the Expression of Factors Associated with Cell Fibrosis and Apoptosis: Clues for Implication in Systemic Sclerosis Development. Int J Mol Sci 2020; 21:E6397. [PMID: 32899126 PMCID: PMC7504027 DOI: 10.3390/ijms21176397] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 08/26/2020] [Accepted: 08/31/2020] [Indexed: 02/07/2023] Open
Abstract
Systemic sclerosis (SSc) is a severe autoimmune disorder characterized by vasculopathy and multi-organ fibrosis; its etiology and pathogenesis are still largely unknown. Herpesvirus infections, particularly by human cytomegalovirus (HCMV) and human herpesvirus 6 (HHV-6), have been suggested among triggers of the disease based on virological and immunological observations. However, the direct impact of HCMV and/or HHV-6 infection on cell fibrosis and apoptosis at the cell microenvironment level has not yet been clarified. Thus, this study aimed to investigate the effects of HCMV and HHV-6 infection on the induction of pro-fibrosis or pro-apoptosis conditions in primary human dermal fibroblasts, one of the relevant SSc target cells. The analysis, performed by microarray in in vitro HCMV- or HHV-6-infected vs. uninfected cells, using specific panels for the detection of the main cellular factors associated with fibrosis or apoptosis, showed that both viruses significantly modified the expression of at least 30 pro-fibrotic and 20 pro-apoptotic factors. Notably, several recognized pro-fibrotic factors were highly induced, and most of them were reported to be involved in vivo in the multifactorial and multistep pathogenic process of SSc, thus suggesting a potential role of both HCMV and HHV-6.
Collapse
Affiliation(s)
- Maria-Cristina Arcangeletti
- Department of Medicine and Surgery, Unit of Virology, University-Hospital of Parma, University of Parma, 43126 Parma, Italy; (C.M.); (F.D.C.); (C.C.); (A.C.)
| | - Maria D’Accolti
- Department of Chemical and Pharmaceutical Sciences, Section of Microbiology and Medical Genetics, University of Ferrara, 44121 Ferrara, Italy; (M.D.); (I.S.); (E.C.)
| | - Clara Maccari
- Department of Medicine and Surgery, Unit of Virology, University-Hospital of Parma, University of Parma, 43126 Parma, Italy; (C.M.); (F.D.C.); (C.C.); (A.C.)
| | - Irene Soffritti
- Department of Chemical and Pharmaceutical Sciences, Section of Microbiology and Medical Genetics, University of Ferrara, 44121 Ferrara, Italy; (M.D.); (I.S.); (E.C.)
| | - Flora De Conto
- Department of Medicine and Surgery, Unit of Virology, University-Hospital of Parma, University of Parma, 43126 Parma, Italy; (C.M.); (F.D.C.); (C.C.); (A.C.)
| | - Carlo Chezzi
- Department of Medicine and Surgery, Unit of Virology, University-Hospital of Parma, University of Parma, 43126 Parma, Italy; (C.M.); (F.D.C.); (C.C.); (A.C.)
| | - Adriana Calderaro
- Department of Medicine and Surgery, Unit of Virology, University-Hospital of Parma, University of Parma, 43126 Parma, Italy; (C.M.); (F.D.C.); (C.C.); (A.C.)
| | - Clodoveo Ferri
- Department of Medical and Surgical Sciences for Children and Adults, Rheumatology Unit, University-Hospital Policlinico of Modena, University of Modena and Reggio Emilia, 41121 Modena, Italy;
| | - Elisabetta Caselli
- Department of Chemical and Pharmaceutical Sciences, Section of Microbiology and Medical Genetics, University of Ferrara, 44121 Ferrara, Italy; (M.D.); (I.S.); (E.C.)
| |
Collapse
|
12
|
Nguyen JK, Austin E, Huang A, Mamalis A, Jagdeo J. The IL-4/IL-13 axis in skin fibrosis and scarring: mechanistic concepts and therapeutic targets. Arch Dermatol Res 2020; 312:81-92. [PMID: 31493000 PMCID: PMC7008089 DOI: 10.1007/s00403-019-01972-3] [Citation(s) in RCA: 110] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Revised: 07/22/2019] [Accepted: 08/29/2019] [Indexed: 02/06/2023]
Abstract
Skin fibrosis, characterized by excessive fibroblast proliferation and extracellular matrix deposition in the dermis, is the histopathologic hallmark of dermatologic diseases such as systemic sclerosis, hypertrophic scars, and keloids. Effective anti-scarring therapeutics remain an unmet need, underscoring the complex pathophysiologic mechanisms of skin fibrosis. The Th2 cytokines interleukin (IL)-4 and IL-13 have been implicated as key mediators in the pathogenesis of fibroproliferative disorders. The goal of this article is to summarize the current understanding of the role of the IL-4/IL-13 axis in wound healing and skin fibrosis. We conducted a literature search to identify research studies investigating the roles of IL-4 and IL-13 in fibrotic skin diseases. While transforming growth factor-beta has long been regarded as the main driver of fibrotic processes, research into the cellular and molecular biology of wound healing has revealed other pathways that promote scar tissue formation. IL-4 and IL-13 are important mediators of skin fibrosis, supported by evidence from in vitro data, animal models of fibrosis, and clinical studies. Overactive signaling of the IL-4/IL-13 axis contributes to the initiation and perpetuation of fibrotic skin diseases. Further insights into the IL-4/IL-13 axis may reveal potential targets for the development of novel therapies that prevent or treat fibrotic skin diseases.
Collapse
Affiliation(s)
- Julie K Nguyen
- Department of Dermatology, SUNY Downstate Medical Center, 450 Clarkson Avenue MSC 46, Brooklyn, NY, 11203, USA
| | - Evan Austin
- Department of Dermatology, SUNY Downstate Medical Center, 450 Clarkson Avenue MSC 46, Brooklyn, NY, 11203, USA
| | - Alisen Huang
- Department of Dermatology, SUNY Downstate Medical Center, 450 Clarkson Avenue MSC 46, Brooklyn, NY, 11203, USA
| | - Andrew Mamalis
- Department of Dermatology, SUNY Downstate Medical Center, 450 Clarkson Avenue MSC 46, Brooklyn, NY, 11203, USA
| | - Jared Jagdeo
- Department of Dermatology, SUNY Downstate Medical Center, 450 Clarkson Avenue MSC 46, Brooklyn, NY, 11203, USA.
- Dermatology Service, VA New York Harbor Healthcare System, Brooklyn, NY, USA.
| |
Collapse
|
13
|
Abstract
Systemic sclerosis (SSc) has the highest cause-specific mortality of all the connective tissue diseases, and the aetiology of this complex and heterogeneous condition remains an enigma. Current disease-modifying therapies for SSc predominantly target inflammatory and vascular pathways but have variable and unpredictable clinical efficacy, and none is curative. Moreover, many of these therapies possess undesirable safety profiles and have no appreciable effect on long-term mortality. This Review describes the most promising of the existing therapeutic targets for SSc and places them in the context of our evolving understanding of the pathophysiology of this disease. As well as taking an in-depth look at the immune, inflammatory, vascular and fibrotic pathways implicated in the pathogenesis of SSc, this Review discusses emerging treatment targets and therapeutic strategies. The article concludes with an overview of important unanswered questions in SSc research that might inform the design of future studies of treatments aimed at modifying the course of this disease.
Collapse
|
14
|
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
|
15
|
Ma WT, Gao F, Gu K, Chen DK. The Role of Monocytes and Macrophages in Autoimmune Diseases: A Comprehensive Review. Front Immunol 2019; 10:1140. [PMID: 31178867 PMCID: PMC6543461 DOI: 10.3389/fimmu.2019.01140] [Citation(s) in RCA: 203] [Impact Index Per Article: 33.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Accepted: 05/07/2019] [Indexed: 12/19/2022] Open
Abstract
Monocytes (Mo) and macrophages (Mϕ) are key components of the innate immune system and are involved in regulation of the initiation, development, and resolution of many inflammatory disorders. In addition, these cells also play important immunoregulatory and tissue-repairing roles to decrease immune reactions and promote tissue regeneration. Several lines of evidence have suggested a causal link between the presence or activation of these cells and the development of autoimmune diseases. In addition, Mo or Mϕ infiltration in diseased tissues is a hallmark of several autoimmune diseases. However, the detailed contributions of these cells, whether they actually initiate disease or perpetuate disease progression, and whether their phenotype and functional alteration are merely epiphenomena are still unclear in many autoimmune diseases. Additionally, little is known about their heterogeneous populations in different autoimmune diseases. Elucidating the relevance of Mo and Mϕ in autoimmune diseases and the associated mechanisms could lead to the identification of more effective therapeutic strategies in the future.
Collapse
Affiliation(s)
- Wen-Tao Ma
- Veterinary Immunology Laboratory, College of Veterinary Medicine, Northwest A&F University, Yangling, China.,School of Life Sciences, University of Science and Technology of China, Hefei, China
| | - Fei Gao
- Veterinary Immunology Laboratory, College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Kui Gu
- Veterinary Immunology Laboratory, College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - De-Kun Chen
- Veterinary Immunology Laboratory, College of Veterinary Medicine, Northwest A&F University, Yangling, China
| |
Collapse
|
16
|
Yun SM, Kim SH, Kim EH. The Molecular Mechanism of Transforming Growth Factor-β Signaling for Intestinal Fibrosis: A Mini-Review. Front Pharmacol 2019; 10:162. [PMID: 30873033 PMCID: PMC6400889 DOI: 10.3389/fphar.2019.00162] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Accepted: 02/11/2019] [Indexed: 01/01/2023] Open
Abstract
Inflammatory bowel disease is known as the most chronic inflammatory disorder in colon, which subsequently progresses to intestinal obstruction and fistula formation. Many studies to date for the treatment of IBD have been focused on inflammation. However, most of the anti-inflammatory agents do not have anti-fibrotic effects and could not relieve intestinal stricture in IBD patients. Because preventing or reversing intestinal fibrosis in IBD is a major therapeutic target, we analyzed the papers focusing on TGF-β signaling in intestinal fibrosis. TGF-β is a good candidate to treat the intestinal fibrosis in IBD which involves TGF-β signaling pathway, EMT, EndMT, ECM, and other regulators. Understanding the mechanism involved in TGF-β signaling will contribute to the treatment and diagnosis of intestinal fibrosis occurring in IBD as well as the understanding of the molecular mechanisms underlying the pathogenesis.
Collapse
Affiliation(s)
- Sun-Mi Yun
- College of Pharmacy and Institute of Pharmaceutical Sciences, CHA University, Seongnam, South Korea
| | - Seok-Ho Kim
- College of Pharmacy and Institute of Pharmaceutical Sciences, CHA University, Seongnam, South Korea
| | - Eun-Hee Kim
- College of Pharmacy and Institute of Pharmaceutical Sciences, CHA University, Seongnam, South Korea
| |
Collapse
|
17
|
Mao YM, Zhao CN, Leng J, Leng RX, Ye DQ, Zheng SG, Pan HF. Interleukin-13: A promising therapeutic target for autoimmune disease. Cytokine Growth Factor Rev 2018; 45:9-23. [PMID: 30581068 DOI: 10.1016/j.cytogfr.2018.12.001] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Accepted: 12/11/2018] [Indexed: 12/16/2022]
Abstract
Interleukin-13 (IL-13) was previously thought to be a redundant presence of IL-4, but in recent years its role in immunity, inflammation, fibrosis, and allergic diseases has become increasingly prominent. IL-13 can regulate several subtypes of T helper (Th) cells and affect their transformation, including Th1, Th2, T17, etc., thus it may play an important role in immune system. Previous studies have revealed that IL-13 is implicated in the pathogenesis of autoimmune diseases, such as systemic lupus erythematosus (SLE), rheumatoid arthritis (RA), systemic sclerosis (SSc), ulcerative colitis (UC), type 1 diabetes (T1D), sjogren's syndrome (SS), etc. In this review, we will briefly discuss the biological features of IL-13 and summarize recent advances in the role of IL-13 in the development and pathogenesis of autoimmune diseases. This information may provide new perspectives and suggestions for the selection of therapeutic targets for autoimmune diseases.
Collapse
Affiliation(s)
- Yan-Mei Mao
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Anhui Province Key Laboratory of Major Autoimmune Diseases, Hefei, Anhui, 230032, China
| | - Chan-Na Zhao
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Anhui Province Key Laboratory of Major Autoimmune Diseases, Hefei, Anhui, 230032, China
| | - Jing Leng
- Anhui Academy of Medical Sciences, 15 Yonghong Road, Hefei, Anhui, China
| | - Rui-Xue Leng
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Anhui Province Key Laboratory of Major Autoimmune Diseases, Hefei, Anhui, 230032, China
| | - Dong-Qing Ye
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Anhui Province Key Laboratory of Major Autoimmune Diseases, Hefei, Anhui, 230032, China
| | - Song Guo Zheng
- Division of Rheumatology, Penn State College of Medicine, Hershey, PA 17033, USA.
| | - Hai-Feng Pan
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Anhui Province Key Laboratory of Major Autoimmune Diseases, Hefei, Anhui, 230032, China.
| |
Collapse
|
18
|
Wang TY, Chen KY, Jhan KY, Li CH, Jung SM, Wang LC. Temporal-spatial expressions of interleukin-4, interleukin-10, and interleukin-13 in the brains of C57BL/6 and BALB/c mice infected with Angiostrongylus cantonensis: An immunohistochemical study. JOURNAL OF MICROBIOLOGY, IMMUNOLOGY, AND INFECTION = WEI MIAN YU GAN RAN ZA ZHI 2018; 53:592-603. [PMID: 30600200 DOI: 10.1016/j.jmii.2018.10.010] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Revised: 09/21/2018] [Accepted: 10/23/2018] [Indexed: 11/25/2022]
Abstract
BACKGROUND Angiostrongylus cantonensis is an important etiologic agent of eosinophilic meningitis and/or eosinophilic meningoencephalitis in humans. Th2 responses have been considered to be predominant in non-permissive hosts. However, changes of cytokines in the central nervous system of the host remain unclear. The present study was conducted to determine the temporal-spatial expressions of IL-4, IL-10, and IL-13 in the brains of infected C57BL/6 and BALB/c mice by immunohistochemistry. METHODS After infecting each mouse with 25 third-stage larvae (L3), brain specimens were collected on day 7 and day 28 post-infection. Each specimen was cut into five sections and stained with corresponding antibodies of the three cytokines. RESULTS In infected C57BL/6 mice, high IL-4 expressions were found in the isocortex, IL-10 in the isocortex, olfactory area, hippocampus, cerebral nuclei, hypothalamus, cerebellum nuclei, and medulla, and IL-13 in the isocortex and cerebellum. In infected BALB/c mice, IL-4 and IL-10 were highly expressed in the isocortex, olfactory areas, cerebral nuclei, hypothalamus, and cerebellum nuclei and IL-13 in the thalamus and hypothalamus. High levels of the cytokines were usually detected in on day 7 in BALB/c mice and day 28 in C57BL/6 mice. CONCLUSION The special temporal-spatial expression changes of these three cytokines in the infected mouse brain may explain the differences in the survival and the time of occurrence of immune responses in the hosts after A. cantonensis infection.
Collapse
Affiliation(s)
- Tzu-Yi Wang
- Department of Parasitology, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan
| | - Kuang-Yao Chen
- Department of Parasitology, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan
| | - Kai-Yuan Jhan
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan
| | - Chung-Han Li
- Department of Parasitology, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan
| | - Shih-Ming Jung
- Department of Pathology, Chang-Gung Memorial Hospital, Chang-Gung Children Hospital at Linkou and Chang-Gung University, Taoyuan 333, Taiwan.
| | - Lian-Chen Wang
- Department of Parasitology, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan; Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan; Molecular Infectious Disease Research Center, Chang Gung Memorial Hospital, Taoyuan, Taiwan.
| |
Collapse
|
19
|
Koguchi-Yoshioka H, Watanabe R, Fujisawa Y, Ishitsuka Y, Nakamura Y, Okiyama N, Fujimoto M. Skin resident memory T-cell population is not constructed effectively in systemic sclerosis. Br J Dermatol 2018; 180:219-220. [PMID: 30120898 DOI: 10.1111/bjd.17100] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- H Koguchi-Yoshioka
- Department of Dermatology, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8575, Japan
| | - R Watanabe
- Department of Dermatology, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8575, Japan
| | - Y Fujisawa
- Department of Dermatology, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8575, Japan
| | - Y Ishitsuka
- Department of Dermatology, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8575, Japan
| | - Y Nakamura
- Department of Dermatology, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8575, Japan
| | - N Okiyama
- Department of Dermatology, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8575, Japan
| | - M Fujimoto
- Department of Dermatology, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8575, Japan
| |
Collapse
|
20
|
Zhang Y, Tang J, Tian Z, van Velkinburgh JC, Song J, Wu Y, Ni B. Innate Lymphoid Cells: A Promising New Regulator in Fibrotic Diseases. Int Rev Immunol 2016. [PMID: 26222510 DOI: 10.3109/08830185.2015.1068304] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Fibrosis is a consequence of chronic inflammation and the persistent accumulation of extracellular matrix, for which the cycle of tissue injury and repair becomes a predominant feature. Both the innate and adaptive immune systems play key roles in the progress of fibrosis. The recently identified subsets of innate lymphoid cells (ILCs), which are mainly localize to epithelial surfaces, have been characterized as regulators of chronic inflammation and tissue remodeling, representing a functional bridge between the innate and adaptive immunity. Moreover, recent research has implicated ILCs as potential contributing factors to several kinds of fibrosis diseases, such as hepatic fibrosis and pulmonary fibrosis. Here, we will summarize and discuss the key roles of ILCs and their related factors in fibrotic diseases and their potential for translation to the clinic.
Collapse
Affiliation(s)
- Yi Zhang
- a Institute of Immunology, PLA, Third Military Medical University , Chongqing , PR China
| | - Jun Tang
- b Department of Dermatology , 105th Hospital of PLA , Hefei , PR China
| | - Zhiqiang Tian
- a Institute of Immunology, PLA, Third Military Medical University , Chongqing , PR China
| | | | - Jianxun Song
- d Department of Microbiology and Immunology , The Pennsylvania State University College of Medicine , Hershey , PA , USA
| | - Yuzhang Wu
- a Institute of Immunology, PLA, Third Military Medical University , Chongqing , PR China
| | - Bing Ni
- a Institute of Immunology, PLA, Third Military Medical University , Chongqing , PR China
| |
Collapse
|
21
|
Fuschiotti P. Current perspectives on the immunopathogenesis of systemic sclerosis. Immunotargets Ther 2016; 5:21-35. [PMID: 27529059 PMCID: PMC4970639 DOI: 10.2147/itt.s82037] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Systemic sclerosis (SSc or scleroderma) is a progressive and highly debilitating autoimmune disorder characterized by inflammation, vasculopathy, and extensive fibrosis. SSc is highly heterogeneous in its clinical presentation, extent and severity of skin and internal organ involvement, and clinical course and has the highest fatality rate among connective tissue diseases. While clinical outcomes have improved in recent years, no current therapy is able to reverse or slow the natural progression of SSc, a reflection of its complex pathogenesis. Although activation of the immune system has long been recognized, the mechanisms responsible for the initiation of autoimmunity and the role of immune effector pathways in the pathogenesis of SSc remain incompletely understood. This review summarizes recent progress in disease pathogenesis with particular focus on the immunopathogenetic mechanisms of SSc.
Collapse
Affiliation(s)
- Patrizia Fuschiotti
- Division of Rheumatology and Clinical Immunology, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| |
Collapse
|
22
|
Mattoo H, Mahajan VS, Maehara T, Deshpande V, Della-Torre E, Wallace ZS, Kulikova M, Drijvers JM, Daccache J, Carruthers MN, Castelino FV, Stone JR, Stone JH, Pillai S. Clonal expansion of CD4(+) cytotoxic T lymphocytes in patients with IgG4-related disease. J Allergy Clin Immunol 2016; 138:825-838. [PMID: 26971690 DOI: 10.1016/j.jaci.2015.12.1330] [Citation(s) in RCA: 261] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Revised: 11/30/2015] [Accepted: 12/14/2015] [Indexed: 12/24/2022]
Abstract
BACKGROUND IgG4-related disease (IgG4-RD) is a systemic condition of unknown cause characterized by highly fibrotic lesions with dense lymphoplasmacytic infiltrates. CD4(+) T cells constitute the major inflammatory cell population in IgG4-RD lesions. OBJECTIVE We used an unbiased approach to characterize CD4(+) T-cell subsets in patients with IgG4-RD based on their clonal expansion and ability to infiltrate affected tissue sites. METHODS We used flow cytometry to identify CD4(+) effector/memory T cells in a cohort of 101 patients with IgG4-RD. These expanded cells were characterized by means of gene expression analysis and flow cytometry. Next-generation sequencing of the T-cell receptor β chain gene was performed on CD4(+)SLAMF7(+) cytotoxic T lymphocytes (CTLs) and CD4(+)GATA3(+) TH2 cells in a subset of patients to identify their clonality. Tissue infiltration by specific T cells was examined by using quantitative multicolor imaging. RESULTS CD4(+) effector/memory T cells with a cytolytic phenotype were expanded in patients with IgG4-RD. Next-generation sequencing revealed prominent clonal expansions of these CD4(+) CTLs but not CD4(+)GATA3(+) memory TH2 cells in patients with IgG4-RD. The dominant T cells infiltrating a range of inflamed IgG4-RD tissue sites were clonally expanded CD4(+) CTLs that expressed SLAMF7, granzyme A, IL-1β, and TGF-β1. Clinical remission induced by rituximab-mediated B-cell depletion was associated with a reduction in numbers of disease-associated CD4(+) CTLs. CONCLUSIONS IgG4-RD is prominently linked to clonally expanded IL-1β- and TGF-β1-secreting CD4(+) CTLs in both peripheral blood and inflammatory tissue lesions. These active, terminally differentiated, cytokine-secreting effector CD4(+) T cells are now linked to a human disease characterized by chronic inflammation and fibrosis.
Collapse
Affiliation(s)
- Hamid Mattoo
- Massachusetts General Hospital, Harvard Medical School, Boston, Mass
| | - Vinay S Mahajan
- Massachusetts General Hospital, Harvard Medical School, Boston, Mass
| | - Takashi Maehara
- Massachusetts General Hospital, Harvard Medical School, Boston, Mass; Section of Oral and Maxillofacial Oncology, Division of Maxillofacial Diagnostic and Surgical Sciences, Fukuoka, Japan
| | - Vikram Deshpande
- Massachusetts General Hospital, Harvard Medical School, Boston, Mass
| | | | - Zachary S Wallace
- Massachusetts General Hospital, Harvard Medical School, Boston, Mass
| | - Maria Kulikova
- Massachusetts General Hospital, Harvard Medical School, Boston, Mass
| | - Jefte M Drijvers
- Massachusetts General Hospital, Harvard Medical School, Boston, Mass
| | - Joe Daccache
- Massachusetts General Hospital, Harvard Medical School, Boston, Mass
| | | | | | - James R Stone
- Massachusetts General Hospital, Harvard Medical School, Boston, Mass
| | - John H Stone
- Massachusetts General Hospital, Harvard Medical School, Boston, Mass.
| | - Shiv Pillai
- Massachusetts General Hospital, Harvard Medical School, Boston, Mass.
| |
Collapse
|
23
|
Koca SS, Pehlivan Y, Kara M, Alibaz-Oner F, Oztuzcu S, Yilmaz N, Cetin GY, Kisacik B, Ozgen M, Pamuk ON, Direskeneli H, Sayarlioglu M, Onat AM. The IL-33 gene is related to increased susceptibility to systemic sclerosis. Rheumatol Int 2016; 36:579-84. [PMID: 26743213 DOI: 10.1007/s00296-015-3417-8] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2015] [Accepted: 12/23/2015] [Indexed: 12/26/2022]
Abstract
Systemic sclerosis (SSc) is a chronic inflammatory disease characterized by widespread fibrosis of the skin and several visceral organs. The pro-fibrotic potential of interleukin (IL)-33 has been demonstrated by in both in vitro and in vivo settings; moreover, increased level of IL-33 has also been reported in patients with SSc. Therefore, the aim of the present study was to detect the potential association of IL-33 gene polymorphisms on the susceptibility of SSc. A total of 300 SSc patients and 280 healthy controls (HC) were enrolled in this multicentric preliminary candidate gene study. DNA samples were harvested using an appropriate commercial DNA isolation kit. Four single nucleotide polymorphisms (SNPs) of IL-33 gene (rs7044343, rs1157505, rs11792633 and rs1929992) were genotyped using the appropriate commercial primer/probe sets on real-time PCR. There was no significant difference in terms of the allelic distributions and minor allele frequencies of evaluated four IL-33 polymorphisms between the SSc and HC groups (P > 0.05 for all). Moreover, the genotypic distributions of rs1157505, rs11792633 and rs1929992 polymorphisms were not significantly different (P > 0.05 for all). However, CC genotype of rs7044343 SNP was significantly higher in the SSc group compared to the HC group (P = 0.013, OR 1.75, 95 % CI 1.12-2.72). This preliminary candidate gene study demonstrates that rs7044343 polymorphism of IL-33 gene is associated with the susceptibility to the SSc in Turkish population. It may be suggested that IL-33 gene may be a candidate gene to research in SSc.
Collapse
Affiliation(s)
- Suleyman Serdar Koca
- Department of Rheumatology, Faculty of Medicine, Firat University, 23119, Elazig, Turkey.
| | - Yavuz Pehlivan
- Department of Rheumatology, Faculty of Medicine, Uludag University, Bursa, Turkey
| | - Murat Kara
- Department of Medical Genetics, Faculty of Medicine, Sitki Kocman University, Mugla, Turkey
| | - Fatma Alibaz-Oner
- Department of Rheumatology, Faculty of Medicine, Marmara University, Istanbul, Turkey
| | - Serdar Oztuzcu
- Department of Medical Genetics, Faculty of Medicine, Gaziantep University, Gaziantep, Turkey
| | - Neslihan Yilmaz
- Department of Rheumatology, Sisli Florence Nightingale Hospital, Istanbul, Turkey
| | - Gozde Yildirim Cetin
- Department of Rheumatology, Faculty of Medicine, Sutcu Imam University, Kahramanmaras, Turkey
| | - Bunyamin Kisacik
- Department of Rheumatology, Faculty of Medicine, Gaziantep University, Gaziantep, Turkey
| | - Metin Ozgen
- Department of Rheumatology, Faculty of Medicine, 19 Mayis University, Samsun, Turkey
| | - Omer Nuri Pamuk
- Department of Rheumatology, Faculty of Medicine, Trakya University, Edirne, Turkey
| | - Haner Direskeneli
- Department of Rheumatology, Faculty of Medicine, Marmara University, Istanbul, Turkey
| | - Mehmet Sayarlioglu
- Department of Rheumatology, Faculty of Medicine, 19 Mayis University, Samsun, Turkey
| | - Ahmet Mesut Onat
- Department of Rheumatology, Faculty of Medicine, Gaziantep University, Gaziantep, Turkey
| |
Collapse
|
24
|
Baraut J, Farge D, Jean-Louis F, Masse I, Grigore EI, Arruda LCM, Lamartine J, Verrecchia F, Michel L. Transforming growth factor-β increases interleukin-13 synthesis via GATA-3 transcription factor in T-lymphocytes from patients with systemic sclerosis. Arthritis Res Ther 2015; 17:196. [PMID: 26227022 PMCID: PMC4521372 DOI: 10.1186/s13075-015-0708-0] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2015] [Accepted: 07/03/2015] [Indexed: 12/23/2022] Open
Abstract
Introduction Transforming growth factor (TGF)-β and interleukin (IL)-13 play a crucial role in the pathogenesis of systemic sclerosis (SSc), partly through activation of collagen production that leads to fibrosis. The aim of the present study was to determine whether TFG-β alters IL-13 production in T lymphocytes from patients with SSc from that seen in those of healthy donors. Methods IL-13 mRNA and protein synthesis under TFG-β exposure was measured in circulating T lymphocytes from healthy donors and patients with SSc and also in the Jurkat Th2 T-cell line, using quantitative real-time PCR and fluorescence-activated cell sorting analysis, respectively. The involvement of Smad and GATA-3 transcription factors was assessed by using specific inhibitors and small interfering RNA, and the binding capacity of GATA-3 to the IL-13 gene promoter was evaluated by chromatin immunoprecipitation assay. Results TGF-β induced a significant decrease in IL-13 mRNA and protein levels in lymphocytes from healthy donors (mean [±SD] inhibition of 30 % ± 10 % and 20 % ± 7 %, respectively; p < 0.05). In contrast, TGF-β promoted a significant increase in IL-13 mRNA levels and IL-13 synthesis by CD4+ and CD8+ T-cell subtypes from patients with SSc, with respective increases of 2.4 ± 0.3-fold, 1.6 ± 0.05-fold and 2.7 ± 0.02-fold. The involvement of the Smad signaling pathway and upregulation of GATA-3 binding capacity on the IL-13 promoter in lymphocytes from patients with SSc contributed to the effect of TGF-β on IL-13 production. Conclusions These results demonstrate that TGF-β upregulates IL-13 synthesis through GATA-3 expression in the T lymphocytes of patients with SSc, confirming that the GATA-3 transcription factor can be regarded as a novel therapeutic target in patients with SSc.
Collapse
Affiliation(s)
- Julie Baraut
- INSERM U976, Pavillon Bazin, Hôpital Saint-Louis 1, Avenue Claude Vellefaux, 75010, Paris, France.
| | - Dominique Farge
- Unité Clinique de Médecine Interne et Pathologie Vasculaire, UF 04, Hôpital Saint-Louis, AP-HP Assistance Publique des Hôpitaux de Paris, INSERM UMRS 1160, Paris, France.
| | - Francette Jean-Louis
- INSERM U976, Pavillon Bazin, Hôpital Saint-Louis 1, Avenue Claude Vellefaux, 75010, Paris, France.
| | - Ingrid Masse
- Université Claude Bernard Lyon I et CNRS UMR5534, Centre de Génétique et de Physiologie Moléculaire et Cellulaire, Villeurbanne, F-69622, France.
| | - Elena Ivan Grigore
- INSERM U976, Pavillon Bazin, Hôpital Saint-Louis 1, Avenue Claude Vellefaux, 75010, Paris, France.
| | - Lucas C M Arruda
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil. .,Center for Cell-based Therapy, São Paulo Research Foundation (FAPESP), São Paulo, Brazil.
| | - Jérôme Lamartine
- Université Claude Bernard Lyon I et CNRS UMR5534, Centre de Génétique et de Physiologie Moléculaire et Cellulaire, Villeurbanne, F-69622, France.
| | - Franck Verrecchia
- INSERM U957, Université de Nantes, 1 rue Gaston Veil, 44000, Nantes, France.
| | - Laurence Michel
- INSERM U976, Pavillon Bazin, Hôpital Saint-Louis 1, Avenue Claude Vellefaux, 75010, Paris, France.
| |
Collapse
|
25
|
D'Amico F, Skarmoutsou E, Mazzarino MC. The sex bias in systemic sclerosis: on the possible mechanisms underlying the female disease preponderance. Clin Rev Allergy Immunol 2015; 47:334-43. [PMID: 24126759 DOI: 10.1007/s12016-013-8392-9] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Systemic sclerosis is a multifactorial and heterogeneous disease. Genetic and environmental factors are known to interplay in the onset and progression of systemic sclerosis. Sex plays an important and determinant role in the development of such a disorder. Systemic sclerosis shows a significant female preponderance. However, the reason for this female preponderance is incompletely understood. Hormonal status, genetic and epigenetic differences, and lifestyle have been considered in order to explain female preponderance in systemic sclerosis. Sex chromosomes play a determinant role in contributing to systemic sclerosis onset and progression, as well as in its sex-biased prevalence. It is known, in fact, that X chromosome contains many sex- and immuno-related genes, thus contributing to immuno tolerance and sex hormone status. This review focuses mainly on the recent progress on epigenetic mechanisms--exclusively linked to the X chromosome--which would contribute to the development of systemic sclerosis. Furthermore, we report also some hypotheses (dealing with skewed X chromosome inactivation, X gene reactivation, acquired monosomy) that have been proposed in order to justify the female preponderance in autoimmune diseases. However, despite the intensive efforts in elucidating the mechanisms involved in the pathogenesis of systemic sclerosis, many questions remain still unanswered.
Collapse
Affiliation(s)
- Fabio D'Amico
- Department of Bio-medical Sciences, University of Catania, via Androne 83, 95124, Catania, Italy,
| | | | | |
Collapse
|
26
|
Ayano M, Tsukamoto H, Kohno K, Ueda N, Tanaka A, Mitoma H, Akahoshi M, Arinobu Y, Niiro H, Horiuchi T, Akashi K. Increased CD226 Expression on CD8+ T Cells Is Associated with Upregulated Cytokine Production and Endothelial Cell Injury in Patients with Systemic Sclerosis. THE JOURNAL OF IMMUNOLOGY 2015; 195:892-900. [PMID: 26109642 DOI: 10.4049/jimmunol.1403046] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/05/2014] [Accepted: 05/26/2015] [Indexed: 02/06/2023]
Abstract
Systemic sclerosis (SSc) is an autoimmune disease characterized by vascular damage and fibrosis of the skin and internal organs. Because activated and oligoclonally expanded CD8(+) T cells can be detected in peripheral blood and lungs of SSc patients, effector memory CD8(+) T cells may play a critical role for organ involvement in SSc; however, the pathogenic functions of effector memory CD8(+) T cells remain incompletely understood. In this study, we performed DNA microarray analysis of the sort-purified effector memory CD8(+) T cells from SSc patients and healthy controls, and showed that the expression of genes related to immune response and cell adhesion, including CD226 (also known as DNAX accessory molecule-1 [DNAM-1]), was significantly altered. Moreover, detailed analysis of CD226 revealed that CD226(high)CD8(+) T cells were increased in SSc patients (mean, 50.7%) compared with healthy controls (32.9%) and were appreciably associated with the severity of skin sclerosis and interstitial lung disease. Furthermore, CD226(+)CD8(+) T cells produced higher amount of various cytokines than CD226(-) ones, and CD226(high)CD8(+) T cells from SSc patients showed upregulated IL-13 production and positive correlation with the cytotoxic capacity of CD8(+) T cells against HUVECs. Finally, the neutralization of CD226 in CD8(+) T cells impaired costimulation, cytokine productions, and cytolysis against HUVECs. These findings indicate that upregulated CD226 expression on CD8(+) T cells reflects disease severity and is involved in SSc pathogenesis via the production of various cytokines, including profibrotic IL-13 and endothelial cell injury, and that CD226 may be a useful target in the treatment of SSc.
Collapse
Affiliation(s)
- Masahiro Ayano
- Department of Medicine and Biosystemic Science, Kyushu University Graduate School of Medical Sciences, Fukuoka 812-8582, Japan
| | - Hiroshi Tsukamoto
- Department of Medicine and Biosystemic Science, Kyushu University Graduate School of Medical Sciences, Fukuoka 812-8582, Japan;
| | - Kentaro Kohno
- Department of Hematology, National Kyushu Medical Center, Fukuoka 810-8563, Japan
| | - Naoyasu Ueda
- Department of Internal Medicine, Miyazaki Prefectural Miyazaki Hospital, Miyazaki 880-8510, Japan
| | - Atsushi Tanaka
- Department of Medicine and Biosystemic Science, Kyushu University Graduate School of Medical Sciences, Fukuoka 812-8582, Japan
| | - Hiroki Mitoma
- Department of Medicine and Biosystemic Science, Kyushu University Graduate School of Medical Sciences, Fukuoka 812-8582, Japan
| | - Mitsuteru Akahoshi
- Department of Medicine and Biosystemic Science, Kyushu University Graduate School of Medical Sciences, Fukuoka 812-8582, Japan
| | - Yojiro Arinobu
- Department of Medicine and Biosystemic Science, Kyushu University Graduate School of Medical Sciences, Fukuoka 812-8582, Japan
| | - Hiroaki Niiro
- Clinical Education Center, Kyushu University Hospital, Fukuoka 812-8582, Japan; and
| | - Takahiko Horiuchi
- Department of Internal Medicine, Kyushu University Beppu Hospital, Beppu 874-0836, Japan
| | - Koichi Akashi
- Department of Medicine and Biosystemic Science, Kyushu University Graduate School of Medical Sciences, Fukuoka 812-8582, Japan
| |
Collapse
|
27
|
Huang XL, Wang YJ, Yan JW, Wan YN, Chen B, Li BZ, Yang GJ, Wang J. Role of anti-inflammatory cytokines IL-4 and IL-13 in systemic sclerosis. Inflamm Res 2015; 64:151-9. [PMID: 25725697 DOI: 10.1007/s00011-015-0806-0] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2015] [Accepted: 02/16/2015] [Indexed: 02/06/2023] Open
Abstract
OBJECTIVE The aim of this paper is to review the anti-inflammatory cytokines IL-4 and IL-13 and their receptor signals; we discuss new insight into their possible roles in systemic sclerosis (SSc) and their overlapping function in SSc. INTRODUCTION SSc is a connective tissue disease characterized by fibrosis. The exact etiology of SSc is unknown, and no therapy has been proved effective in modifying its course. Recently the roles of IL-4 and IL-13 in the development of SSc have been extensively considered. The possible roles of IL-4 and IL-13, especially their overlapping function, in SSc are not well documented. METHODS A literature survey was performed using a PubMed database search to gather complete information regarding IL-4 and IL-13 and their role in inflammation. RESULTS AND CONCLUSIONS The participation of complex pathways of IL-4 and IL-13 in the process of inflammation and fibrosis action in SSc is still not very clear, and some pathogenesis of regulation found in vitro needs to be further proved. There is still more work which could be done to achieve useful developments with therapeutic benefit in SSc.
Collapse
Affiliation(s)
- Xiao-Lei Huang
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, No. 81, Meishan Road, 230032, Hefei, Anhui, People's Republic of China
| | | | | | | | | | | | | | | |
Collapse
|
28
|
Kendall RT, Feghali-Bostwick CA. Fibroblasts in fibrosis: novel roles and mediators. Front Pharmacol 2014; 5:123. [PMID: 24904424 PMCID: PMC4034148 DOI: 10.3389/fphar.2014.00123] [Citation(s) in RCA: 701] [Impact Index Per Article: 63.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2014] [Accepted: 05/09/2014] [Indexed: 12/23/2022] Open
Abstract
Fibroblasts are the most common cell type of the connective tissues found throughout the body and the principal source of the extensive extracellular matrix (ECM) characteristic of these tissues. They are also the central mediators of the pathological fibrotic accumulation of ECM and the cellular proliferation and differentiation that occurs in response to prolonged tissue injury and chronic inflammation. The transformation of the fibroblast cell lineage involves classical developmental signaling programs and includes a surprisingly diverse range of precursor cell types—most notably, myofibroblasts that are the apex of the fibrotic phenotype. Myofibroblasts display exaggerated ECM production; constitutively secrete and are hypersensitive to chemical signals such as cytokines, chemokines, and growth factors; and are endowed with a contractile apparatus allowing them to manipulate the ECM fibers physically to close open wounds. In addition to ECM production, fibroblasts have multiple concomitant biological roles, such as in wound healing, inflammation, and angiogenesis, which are each interwoven with the process of fibrosis. We now recognize many common fibroblast-related features across various physiological and pathological protracted processes. Indeed, a new appreciation has emerged for the role of non-cancerous fibroblast interactions with tumors in cancer progression. Although the predominant current clinical treatments of fibrosis involve non-specific immunosuppressive and anti-proliferative drugs, a variety of potential therapies under investigation specifically target fibroblast biology.
Collapse
Affiliation(s)
- Ryan T Kendall
- Division of Rheumatology and Immunology, Department of Medicine, Medical University of South Carolina Charleston, SC, USA
| | - Carol A Feghali-Bostwick
- Division of Rheumatology and Immunology, Department of Medicine, Medical University of South Carolina Charleston, SC, USA
| |
Collapse
|
29
|
Early Systemic Sclerosis: Serum Profiling of Factors Involved in Endothelial, T-cell, and Fibroblast Interplay is Marked by Elevated Interleukin-33 Levels. J Clin Immunol 2014; 34:663-8. [DOI: 10.1007/s10875-014-0037-0] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2013] [Accepted: 04/02/2014] [Indexed: 02/06/2023]
|
30
|
Ho YY, Lagares D, Tager AM, Kapoor M. Fibrosis--a lethal component of systemic sclerosis. Nat Rev Rheumatol 2014; 10:390-402. [PMID: 24752182 DOI: 10.1038/nrrheum.2014.53] [Citation(s) in RCA: 233] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Fibrosis is a pathological process characterized by excessive accumulation of connective tissue components in an organ or tissue. Fibrosis is produced by deregulated wound healing in response to chronic tissue injury or chronic inflammation, the hallmarks of rheumatic diseases. Progressive fibrosis, which distorts tissue architecture and results in progressive loss of organ function, is now recognized to be one of the major causes of morbidity and mortality in individuals with one of the most lethal rheumatic disease, systemic sclerosis (SSc). In this Review, we discuss the pathological role of fibrosis in SSc. We discuss the involvement of endothelium and pericyte activation, aberrant immune responses, endoplasmic reticulum stress and chronic tissue injury in the initiation of fibrosis in SSc. We then discuss fibroblast activation and myofibroblast differentiation that occurs in response to these initiating processes and is responsible for excessive accumulation of extracellular matrix. Finally, we discuss the chemical and mechanical signals that drive fibroblast activation and myofibroblast differentiation, which could serve as targets for new therapies for fibrosis in SSc.
Collapse
Affiliation(s)
- Yuen Yee Ho
- Shriners Hospital for Children, Division of Surgical Research, McGill University, 1529 Cedar Avenue, Montreal, QC H3G1A6, Canada
| | - David Lagares
- Pulmonary and Critical Care Unit and Centre for Immunology and Inflammatory Diseases, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, 149 13th Street, Charlestown, MA 02129, USA
| | - Andrew M Tager
- Pulmonary and Critical Care Unit and Centre for Immunology and Inflammatory Diseases, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, 149 13th Street, Charlestown, MA 02129, USA
| | - Mohit Kapoor
- The Toronto Western Research Institute, Division of Orthopaedics, Toronto Western Hospital, The University Health Network, 60 Leonard Avenue, Toronto, ON M5T 2S8, Canada
| |
Collapse
|
31
|
O'Reilly S. Role of interleukin-13 in fibrosis, particularly systemic sclerosis. Biofactors 2013; 39:593-6. [PMID: 23893513 DOI: 10.1002/biof.1117] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2013] [Revised: 04/25/2013] [Accepted: 05/07/2013] [Indexed: 11/07/2022]
Abstract
Chronic inflammation can lead to altered extracellular matrix deposition and ultimately fibrosis. Interleukin-13 (IL-13) is a cytokine that was found to promote IgE class switching and inhibit proinflammatory cytokines. However, it is now recognized as an important mediator in allergy and most importantly fibrosis. Indeed, animal studies with genetically deleted mice have demonstrated its critical role in fibrosis and although it shares over lapping functions with IL-4 it is the dominant cytokine in fibrosis. Systemic sclerosis is an autoimmune disease in which there is chronic inflammation and fibrosis. The disease is associated with a Th2 polarization and IL-13 levels are elevated both in the blood and in the skin of patients. This review will examine the role of IL-13 in driving fibrosis with a particular emphasis on systemic sclerosis as a prototypical fibrotic disease. It will highlight recent research into the role of IL-13 and how this cytokine may be targeted in systemic sclerosis.
Collapse
Affiliation(s)
- Steven O'Reilly
- Musculoskeletal Research Group, Institute of Cellular Medicine, Faculty of Medicine, Newcastle University, Newcastle upon Tyne, UK
| |
Collapse
|
32
|
Datta A, Alexander R, Sulikowski MG, Nicholson AG, Maher TM, Scotton CJ, Chambers RC. Evidence for a functional thymic stromal lymphopoietin signaling axis in fibrotic lung disease. THE JOURNAL OF IMMUNOLOGY 2013; 191:4867-79. [PMID: 24081992 DOI: 10.4049/jimmunol.1300588] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Thymic stromal lymphopoietin (TSLP) recently has emerged as a key cytokine in the development of type 2 immune responses. Although traditionally associated with allergic inflammation, type 2 responses are also recognized to contribute to the pathogenesis of tissue fibrosis. However, the role of TSLP in the development of non-allergen-driven diseases, characterized by profibrotic type 2 immune phenotypes and excessive fibroblast activation, remains underexplored. Fibroblasts represent the key effector cells responsible for extracellular matrix production but additionally play important immunoregulatory roles, including choreographing immune cell recruitment through chemokine regulation. The aim of this study was to examine whether TSLP may be involved in the pathogenesis of a proto-typical fibrotic disease, idiopathic pulmonary fibrosis (IPF). We combined the immunohistochemical analysis of human IPF biopsy material with signaling studies by using cultured primary human lung fibroblasts and report for the first time, to our knowledge, that TSLP and its receptor (TSLPR) are highly upregulated in IPF. We further show that lung fibroblasts represent both a novel cellular source and target of TSLP and that TSLP induces fibroblast CCL2 release (via STAT3) and subsequent monocyte chemotaxis. These studies extend our understanding of TSLP as a master regulator of type 2 immune responses beyond that of allergic inflammatory conditions and suggest a novel role for TSLP in the context of chronic fibrotic lung disease.
Collapse
Affiliation(s)
- Arnab Datta
- Centre for Inflammation and Tissue Repair, University College London, London WC1E 6JF, United Kingdom
| | | | | | | | | | | | | |
Collapse
|
33
|
An update on stem cell transplantation in autoimmune rheumatologic disorders. Curr Allergy Asthma Rep 2013; 12:530-40. [PMID: 22956390 DOI: 10.1007/s11882-012-0298-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Stem cell transplant (SCT) has long been the standard of care for several hematologic, immunodeficient, and oncologic disorders. Recently, SCT has become an increasingly utilized therapy for refractory autoimmune rheumatologic disorders (ARDs). The efficacy of SCT in ARDs has been attributed to resetting an aberrant immune system either through direct immune replacement with hematopoietic stem cells or through immunomodulation with mesenchymal stem cells. Among ARDs, refractory systemic sclerosis (SSc) and systemic lupus erythematosus (SLE) are the most common indications for SCT. SCT has also been used in refractory rheumatoid arthritis, inflammatory myopathies, antiphospholipid syndrome, granulomatosis with polyangiitis, and pediatric ARDs. Complete responses have been reported in approximately 30 % of patients in all disease categories. Transplant-related mortality, however, remains a concern. Future large multi-center prospective randomized clinical trials will help to better define the specific role of SCT in the treatment of patients with ARDs.
Collapse
|
34
|
Strategies for anti-fibrotic therapies. Biochim Biophys Acta Mol Basis Dis 2012; 1832:1088-103. [PMID: 23266403 DOI: 10.1016/j.bbadis.2012.12.007] [Citation(s) in RCA: 125] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2012] [Revised: 12/07/2012] [Accepted: 12/08/2012] [Indexed: 02/07/2023]
Abstract
The fibrotic diseases encompass a wide spectrum of entities including such multisystemic diseases as systemic sclerosis, nephrogenic systemic fibrosis and sclerodermatous graft versus host disease, as well as organ-specific disorders such as pulmonary, liver, and kidney fibrosis. Collectively, given the wide variety of affected organs, the chronic nature of the fibrotic processes, and the large number of individuals suffering their devastating effects, these diseases pose one of the most serious health problems in current medicine and a serious economic burden to society. Despite these considerations there is currently no accepted effective treatment. However, remarkable progress has been achieved in the elucidation of their pathogenesis including the identification of the critical role of myofibroblasts and the determination of molecular mechanisms that result in the transcriptional activation of the genes responsible for the fibrotic process. Here we review the origin of the myofibroblast and discuss the crucial regulatory pathways involving multiple growth factors and cytokines that participate in the pathogenesis of the fibrotic process. Potentially effective therapeutic strategies based upon this new information are considered in detail and the major challenges that remain and their possible solutions are presented. It is expected that translational efforts devoted to convert this new knowledge into novel and effective anti-fibrotic drugs will be forthcoming in the near future. This article is part of a Special Issue entitled: Fibrosis: Translation of basic research to human disease.
Collapse
|
35
|
Duffield JS, Lupher M, Thannickal VJ, Wynn TA. Host responses in tissue repair and fibrosis. ANNUAL REVIEW OF PATHOLOGY-MECHANISMS OF DISEASE 2012; 8:241-76. [PMID: 23092186 DOI: 10.1146/annurev-pathol-020712-163930] [Citation(s) in RCA: 451] [Impact Index Per Article: 34.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Myofibroblasts accumulate in the spaces between organ structures and produce extracellular matrix (ECM) proteins, including collagen I. They are the primary "effector" cells in tissue remodeling and fibrosis. Previously, leukocyte progenitors termed fibrocytes and myofibroblasts generated from epithelial cells through epithelial-to-mesenchymal transition (EMT) were considered the primary sources of ECM-producing myofibroblasts in injured tissues. However, genetic fate mapping experiments suggest that mesenchyme-derived cells, known as resident fibroblasts, and pericytes are the primary precursors of scar-forming myofibroblasts, whereas epithelial cells, endothelial cells, and myeloid leukocytes contribute to fibrogenesis predominantly by producing key fibrogenic cytokines and by promoting cell-to-cell communication. Numerous cytokines derived from T cells, macrophages, and other myeloid cell populations are important drivers of myofibroblast differentiation. Monocyte-derived cell populations are key regulators of the fibrotic process: They act as a brake on the processes driving fibrogenesis, and they dismantle and degrade established fibrosis. We discuss the origins, modes of activation, and fate of myofibroblasts in various important fibrotic diseases and describe how manipulation of macrophage activation could help ameliorate fibrosis.
Collapse
Affiliation(s)
- Jeremy S Duffield
- Division of Nephrology, Center for Lung Biology, and the Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98019, USA
| | | | | | | |
Collapse
|
36
|
Todd NW, Luzina IG, Atamas SP. Molecular and cellular mechanisms of pulmonary fibrosis. FIBROGENESIS & TISSUE REPAIR 2012; 5:11. [PMID: 22824096 PMCID: PMC3443459 DOI: 10.1186/1755-1536-5-11] [Citation(s) in RCA: 305] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/02/2012] [Accepted: 06/28/2012] [Indexed: 12/22/2022]
Abstract
Pulmonary fibrosis is a chronic lung disease characterized by excessive accumulation of extracellular matrix (ECM) and remodeling of the lung architecture. Idiopathic pulmonary fibrosis is considered the most common and severe form of the disease, with a median survival of approximately three years and no proven effective therapy. Despite the fact that effective treatments are absent and the precise mechanisms that drive fibrosis in most patients remain incompletely understood, an extensive body of scientific literature regarding pulmonary fibrosis has accumulated over the past 35 years. In this review, we discuss three broad areas which have been explored that may be responsible for the combination of altered lung fibroblasts, loss of alveolar epithelial cells, and excessive accumulation of ECM: inflammation and immune mechanisms, oxidative stress and oxidative signaling, and procoagulant mechanisms. We discuss each of these processes separately to facilitate clarity, but certainly significant interplay will occur amongst these pathways in patients with this disease.
Collapse
Affiliation(s)
- Nevins W Todd
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA.
| | | | | |
Collapse
|
37
|
Abstract
Fibrosis is a pathological feature of most chronic inflammatory diseases. Fibrosis, or scarring, is defined by the accumulation of excess extracellular matrix components. If highly progressive, the fibrotic process eventually leads to organ malfunction and death. Fibrosis affects nearly every tissue in the body. Here we discuss how key components of the innate and adaptive immune response contribute to the pathogenesis of fibrosis. We also describe how cell-intrinsic changes in important structural cells can perpetuate the fibrotic response by regulating the differentiation, recruitment, proliferation and activation of extracellular matrix-producing myofibroblasts. Finally, we highlight some of the key mechanisms and pathways of fibrosis that are being targeted as potential therapies for a variety of important human diseases.
Collapse
|
38
|
Abstract
Fibrosis is a pathological feature of most chronic inflammatory diseases. Fibrosis, or scarring, is defined by the accumulation of excess extracellular matrix components. If highly progressive, the fibrotic process eventually leads to organ malfunction and death. Fibrosis affects nearly every tissue in the body. Here we discuss how key components of the innate and adaptive immune response contribute to the pathogenesis of fibrosis. We also describe how cell-intrinsic changes in important structural cells can perpetuate the fibrotic response by regulating the differentiation, recruitment, proliferation and activation of extracellular matrix-producing myofibroblasts. Finally, we highlight some of the key mechanisms and pathways of fibrosis that are being targeted as potential therapies for a variety of important human diseases.
Collapse
Affiliation(s)
- Thomas A Wynn
- Immunopathogenesis Section, Program in Barrier Immunity and Repair, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA.
| | | |
Collapse
|