1
|
Khattar G, Wei C, Davis A, Saliba F, Aoun L, Mourad O, Achkar MA, Rosenberg A, Grovu R, Bradu S, El-Sayegh S, Mustafa A. Systemic sclerosis and acute heart failure in prosthetic heart valve patients: A retrospective analysis. JOURNAL OF SCLERODERMA AND RELATED DISORDERS 2024:23971983241278853. [PMID: 39544905 PMCID: PMC11559524 DOI: 10.1177/23971983241278853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Accepted: 08/12/2024] [Indexed: 11/17/2024]
Abstract
Background Acute heart failure in patients with prosthetic heart valves is a complex problem with clinical and therapeutic challenges. Systemic sclerosis is a chronic autoimmune disease frequently associated with valvular abnormalities. The association between systemic sclerosis and acute heart failure in patients with prosthetic heart valves remains understudied. Methods Prosthetic valve patients were extracted from the National Inpatient Sample Database. Baseline patient demographics, comorbidities, and known acute heart failure risk factors were collected from the database using International Classification of Diseases, 10th Revision codes. Patients were subsequently stratified by the diagnosis of systemic sclerosis. The primary outcome was acute heart failure. while secondary outcome included pulmonary outcomes. Univariate and multivariate logistic regression analyses were performed. 1:1 matching was performed to verify our findings. Results Among 188,615 patients, 235 patients had systemic sclerosis. Systemic sclerosis patients had higher rates of acute heart failure relative to non-systemic sclerosis patients (28.5% vs 22.6%). On multivariate analysis, systemic sclerosis was associated with increased acute heart failure (adjusted OR: 1.38 (1.02-1.85), p = 0.036). After matching, systemic sclerosis was still associated with an increased incidence of acute heart failure (OR: 1.94 (1.25-3.03), p = 0.003). On subgroup analysis, patients with CREST syndrome did not show significantly increased acute heart failure (OR: 1.44 (0.84-2.47), p = 0.184). Patients with systemic sclerosis also showed a significantly higher rate of acute respiratory failure compared to non-systemic sclerosis patients (20.9% vs 13.7%, p = 0.001). Conclusion Systemic sclerosis may increase the risk for acute heart failure in patients with prosthetic valves. Closer monitoring for heart failure symptoms should be considered in systemic sclerosis patients with prosthetic valves.
Collapse
Affiliation(s)
- Georges Khattar
- Department of Medicine, Northwell Health, Staten Island, NY, USA
| | - Chapman Wei
- Department of Medicine, Northwell Health, Staten Island, NY, USA
| | - Alanna Davis
- Department of Medicine, Northwell Health, Staten Island, NY, USA
| | - Fares Saliba
- Department of Medicine, Northwell Health, Staten Island, NY, USA
| | - Laurence Aoun
- Department of Medicine, Northwell Health, Staten Island, NY, USA
| | - Omar Mourad
- Department of Medicine, Northwell Health, Staten Island, NY, USA
| | - Michel Al Achkar
- Department of Medicine, Northwell Health, Staten Island, NY, USA
| | - Angela Rosenberg
- Department of Medicine, Northwell Health, Staten Island, NY, USA
| | - Radu Grovu
- Department of Medicine, Northwell Health, Staten Island, NY, USA
| | - Stefan Bradu
- Department of Dermatology, Northwell Health, Staten Island, NY, USA
| | | | - Ahmad Mustafa
- Department of Cardiology, Northwell Health, Staten Island, NY, USA
| |
Collapse
|
2
|
Ren C, Carrillo ND, Cryns VL, Anderson RA, Chen M. Environmental pollutants and phosphoinositide signaling in autoimmunity. JOURNAL OF HAZARDOUS MATERIALS 2024; 465:133080. [PMID: 38091799 PMCID: PMC10923067 DOI: 10.1016/j.jhazmat.2023.133080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 11/16/2023] [Accepted: 11/22/2023] [Indexed: 02/08/2024]
Abstract
Environmental pollution stands as one of the most critical challenges affecting human health, with an estimated mortality rate linked to pollution-induced non-communicable diseases projected to range from 20% to 25%. These pollutants not only disrupt immune responses but can also trigger immunotoxicity. Phosphoinositide signaling, a pivotal regulator of immune responses, plays a central role in the development of autoimmune diseases and exhibits high sensitivity to environmental stressors. Among these stressors, environmental pollutants have become increasingly prevalent in our society, contributing to the initiation and exacerbation of autoimmune conditions. In this review, we summarize the intricate interplay between phosphoinositide signaling and autoimmune diseases within the context of environmental pollutants and contaminants. We provide an up-to-date overview of stress-induced phosphoinositide signaling, discuss 14 selected examples categorized into three groups of environmental pollutants and their connections to immune diseases, and shed light on the associated phosphoinositide signaling pathways. Through these discussions, this review advances our understanding of how phosphoinositide signaling influences the coordinated immune response to environmental stressors at a biological level. Furthermore, it offers valuable insights into potential research directions and therapeutic targets aimed at mitigating the impact of environmental pollutants on the pathogenesis of autoimmune diseases. SYNOPSIS: Phosphoinositide signaling at the intersection of environmental pollutants and autoimmunity provides novel insights for managing autoimmune diseases aggravated by pollutants.
Collapse
Affiliation(s)
- Chang Ren
- Department of Pharmacology, Joint Laboratory of Guangdong-Hong Kong Universities for Vascular Homeostasis and Diseases, School of Medicine, Southern University of Science and Technology, Shenzhen 518055, China
| | - Noah D Carrillo
- Department of Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Vincent L Cryns
- Department of Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA; University of Wisconsin Carbone Cancer Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Richard A Anderson
- University of Wisconsin Carbone Cancer Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Mo Chen
- Department of Pharmacology, Joint Laboratory of Guangdong-Hong Kong Universities for Vascular Homeostasis and Diseases, School of Medicine, Southern University of Science and Technology, Shenzhen 518055, China.
| |
Collapse
|
3
|
Pellicano C, Vantaggio L, Colalillo A, Pocino K, Basile V, Marino M, Basile U, Rosato E. Type 2 cytokines and scleroderma interstitial lung disease. Clin Exp Med 2023; 23:3517-3525. [PMID: 37392249 PMCID: PMC10618297 DOI: 10.1007/s10238-023-01125-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Accepted: 06/21/2023] [Indexed: 07/03/2023]
Abstract
Interstitial lung disease (ILD) is a life-threatening complication of systemic sclerosis (SSc). Type 2 (Th2) cytokines play a pivotal role in airway disease. Study aim was to evaluate serum level of Th2 interleukin (IL) and chemokine in SSc-ILD. Serum levels of IL-4, IL-5, IL-11, IL-13, IL-21, IL-31 and CXCL-13 were measured by Bio-Plex Multiplex Immunoassays in 60 SSc patients and 20 healthy controls (HC). Pulmonary function tests with diffusion lung capacity for carbon monoxide (DLco) and high resolution computed tomography (HRCT) were performed in SSc patients. ILD is defined as fibrotic changes (ground glass, reticular and honeycombing), assessed by Computer-Aided Lung Informatics for Pathology Evaluation and Ratings (CALIPER) software, affecting at least 10% of the lungs. Serum levels of Th2 cytokines were higher in SSc patients than HC. A linear correlation was observed between ground glass and IL-13 (r = 0.342, p < 0.01), IL-21 (r = 0.345, p < 0.01), IL-31 (r = 0.473, p < 0.001), IL-4 (r = 0.863, p < 0.001), IL-5 (r = 0.249, p < 0.05) and peripheral blood eosinophils (r = 0.463, p < 0.001). We found a negative correlation between DLco and IL-4 (r = - 0.511, p < 0.001) and peripheral blood eosinophils (r = - 0.446, p < 0.001). In the logistic regression analysis, IL-4 is associated with DLco ≤ 60% of the predicted [OR 1.039 (CI 95%: 1.015-1.064), p < 0.001], whilst mRSS [OR 1.138 (CI 95%: 1.023-1.266), p < 0.05] and IL-4 [OR 1.017 (CI 95%: 1-1.034), p < 0.05] were associated with ILD. Th2 inflammation could play a key role in early phase of SSc-ILD.
Collapse
Affiliation(s)
- Chiara Pellicano
- Department of Translational and Precision Medicine, Sapienza University of Rome, Viale Dell'Università 37, 00185, Rome, Italy
| | - Lorenzo Vantaggio
- Department of Translational and Precision Medicine, Sapienza University of Rome, Viale Dell'Università 37, 00185, Rome, Italy
| | - Amalia Colalillo
- Department of Translational and Precision Medicine, Sapienza University of Rome, Viale Dell'Università 37, 00185, Rome, Italy
| | - Krizia Pocino
- UOC of Clinical Pathology, General Hospital San Pietro Fatebenefratelli, 00189, Rome, Italy
| | - Valerio Basile
- Clinical Pathology Unit and Cancer Biobank, Department of Research and Advanced Technologies, IRCCS Regina Elena National Cancer Institute, 00144, Rome, Italy
| | - Mariapaola Marino
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Fondazione Policlinico A. Gemelli IRCCS, 00168, Rome, Italy
| | - Umberto Basile
- UOC Clinical Pathology DEA II Level, Hospital Santa Maria Goretti-ASL Latina, 04100, Latina, Italy
| | - Edoardo Rosato
- Department of Translational and Precision Medicine, Sapienza University of Rome, Viale Dell'Università 37, 00185, Rome, Italy.
| |
Collapse
|
4
|
Corano Scheri K, Liang X, Dalal V, Le Poole IC, Varga J, Hayashida T. SARA suppresses myofibroblast precursor transdifferentiation in fibrogenesis in a mouse model of scleroderma. JCI Insight 2022; 7:160977. [PMID: 36136606 PMCID: PMC9675568 DOI: 10.1172/jci.insight.160977] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 09/12/2022] [Indexed: 12/15/2022] Open
Abstract
We previously reported that Smad anchor for receptor activation (SARA) plays a critical role in maintaining epithelial cell phenotype. Here, we show that SARA suppressed myofibroblast precursor transdifferentiation in a mouse model of scleroderma. Mice overexpressing SARA specifically in PDGFR-β+ pericytes and pan-leukocytes (SARATg) developed significantly less skin fibrosis in response to bleomycin injection compared with wild-type littermates (SARAWT). Single-cell RNA-Seq analysis of skin PDGFR-β+ cells implicated pericyte subsets assuming myofibroblast characteristics under fibrotic stimuli, and SARA overexpression blocked the transition. In addition, a cluster that expresses molecules associated with Th2 cells and macrophage activation was enriched in SARAWT mice, but not in SARATg mice, after bleomycin treatment. Th2-specific Il-31 expression was increased in skin of the bleomycin-treated SARAWT mice and patients with scleroderma (or systemic sclerosis, SSc). Receptor-ligand analyses indicated that lymphocytes mediated pericyte transdifferentiation in SARAWT mice, while with SARA overexpression the myofibroblast activity of pericytes was suppressed. Together, these data suggest a potentially novel crosstalk between myofibroblast precursors and immune cells in the pathogenesis of SSc, in which SARA plays a critical role.
Collapse
Affiliation(s)
- Katia Corano Scheri
- Department of Pediatrics, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA.,Pediatric Nephrology, Ann and Robert H. Lurie Children’s Hospital of Chicago, Chicago, Illinois, USA
| | - Xiaoyan Liang
- Department of Pediatrics, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Vidhi Dalal
- Department of Pediatrics, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA.,Pediatric Nephrology, Ann and Robert H. Lurie Children’s Hospital of Chicago, Chicago, Illinois, USA
| | - I. Caroline Le Poole
- Departments of Dermatology and Microbiology and Immunology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - John Varga
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Tomoko Hayashida
- Department of Pediatrics, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA.,Pediatric Nephrology, Ann and Robert H. Lurie Children’s Hospital of Chicago, Chicago, Illinois, USA
| |
Collapse
|
5
|
Kendall RT, Renaud L, Baatz JE, Malaab M, Nguyen XX, Feghali-Bostwick CA. Systemic sclerosis biomarkers detection in the secretome of TGFβ1-activated primary human lung fibroblasts. J Proteomics 2021; 242:104243. [PMID: 33930553 DOI: 10.1016/j.jprot.2021.104243] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 02/16/2021] [Accepted: 04/20/2021] [Indexed: 12/17/2022]
Abstract
TGFβ1 is a profibrotic mediator that contributes to a broad spectrum of pathologies, including systemic sclerosis-associated pulmonary fibrosis (SSc-PF). However, the secretome of TGFβ1-stimulated primary human normal lung (NL) fibroblasts has not been well characterized. Using fluorescent 2-dimensional gel electrophoresis (2D-PAGE) and differential gel electrophoresis (DIGE) followed by Mass Spectrometry, we identified 37 differentially secreted proteins in the conditioned media of TGFβ1-activated NL fibroblasts and generated a protein-protein association network of the TGFβ1 secretome using STRING. Functional enrichment revealed that several biological processes and pathways characteristic of PF were enriched. Additionally, by comparing the TGFβ1 secretome of NL fibroblasts to proteomic biomarkers from biological fluids of systemic sclerosis (SSc) patients, we identified 11 overlapping proteins. Together our data validate the TGFβ1-induced secretome of NL fibroblasts as a valid in vitro model that reflects SSc biomarkers and identify potential therapeutic targets for SSc-PF. SIGNIFICANCE: All proteins secreted by fibroblasts into the extracellular space, representing the secretome, promote cell-to-cell communication as well as tissue homeostasis, immune mechanisms, developmental regulation, proteolysis, development of the extracellular matrix (ECM) and cell adhesion. Therefore, it is crucial to understand how TGFβ1, a well-known profibrotic cytokine, modulates the secretome of pulmonary fibroblasts, and how the TGFβ1-induced secretome resembles biomarkers in SSc. Using functional enrichment analysis, key pathways and hub proteins can be identified and studied as potential therapeutic targets for pulmonary fibrosis.
Collapse
Affiliation(s)
- Ryan T Kendall
- Department of Medicine, Rheumatology & Immunology, MUSC, Charleston, SC, United States of America
| | - Ludivine Renaud
- Department of Medicine, Rheumatology & Immunology, MUSC, Charleston, SC, United States of America.
| | - John E Baatz
- Department of Pediatrics, MUSC, Charleston, SC, United States of America.
| | - Maya Malaab
- Department of Medicine, Rheumatology & Immunology, MUSC, Charleston, SC, United States of America.
| | - Xinh-Xinh Nguyen
- Department of Medicine, Rheumatology & Immunology, MUSC, Charleston, SC, United States of America.
| | - Carol A Feghali-Bostwick
- Department of Medicine, Rheumatology & Immunology, MUSC, Charleston, SC, United States of America.
| |
Collapse
|
6
|
Implication of miR-126 and miR-139-5p in Plasmacytoid Dendritic Cell Dysregulation in Systemic Sclerosis. J Clin Med 2021; 10:jcm10030491. [PMID: 33573268 PMCID: PMC7866506 DOI: 10.3390/jcm10030491] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 01/19/2021] [Accepted: 01/26/2021] [Indexed: 12/25/2022] Open
Abstract
Compelling evidence shows the involvement of plasmacytoid dendritic cells (pDCs) in systemic sclerosis (SSc) pathogenesis. This study investigated whether microRNAs (miRNAs) are involved in the dysregulation of pDCs in SSc patients already at early stages. RNA from circulating pDCs was isolated from two independent cohorts of SSc patients with different disease phenotypes, and individuals with Raynaud’s phenomenon, for microRNA profiling and RNA-sequencing analysis. Proteomic analysis was exploited to identify novel direct miRNA targets at the protein level. Twelve and fifteen miRNAs were differentially expressed in at least one group of patients compared to healthy controls in discovery cohort I and II, respectively. Of note, miR-126 and miR-139-5p were upregulated in both preclinical and definite SSc patients and correlated with the expression of type I interferon (IFN)-responsive genes. Toll-like receptor 9 (TLR9) stimulation of healthy pDCs upregulated the expression of both miRNAs, similarly to what was observed in patients. The proteomic analysis identified USP24 as a novel target of miR-139-5p. The expression level of USP24 was inversely correlated with miR-139-5p expression in SSc patients and induced by TLR9 stimulation in healthy pDCs. These findings demonstrated that the miRNA profile is altered in pDCs of SSc patients already at early stages of the disease and indicate their potential contribution to pDC activation observed in patients.
Collapse
|
7
|
Paradowska-Gorycka A, Wajda A, Stypinska B, Walczuk E, Rzeszotarska E, Walczyk M, Haladyj E, Romanowska-Prochnicka K, Felis-Giemza A, Lewandowska A, Olesińska M. Variety of endosomal TLRs and Interferons (IFN-α, IFN-β, IFN-γ) expression profiles in patients with SLE, SSc and MCTD. Clin Exp Immunol 2021; 204:49-63. [PMID: 33336388 DOI: 10.1111/cei.13566] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 10/21/2020] [Accepted: 11/13/2020] [Indexed: 12/13/2022] Open
Abstract
We investigated Toll-like receptor (TLR)-3/-7/-8/-9 and interferon (IFN)-α/β/γ mRNA expression in whole blood and serum IFN-α/β/γ levels in patients with mixed connective tissue disease (MCTD), systemic lupus erythematosus (SLE) and systemic sclerosis (SSc) and in healthy subjects to assess the association between the TLR-IFN expression and severity of and susceptibility to diseases, and identify potential biomarkers. Expression of the IFN-γ, TLR-3 and TLR-8 was detected only in SLE patients. TLR-7, IFN-α and IFN-β expression was highest in SLE, while TLR-9 expression was highest in SSc patients. In SLE and MCTD patients a strong correlation was observed between TLR-7 and IFN-α expression and IFN-β and IFN-α expression. In MCTD patients, negative correlation between IFN-α and TLR-9 and TLR-7 and TLR-9 was revealed. TLR-9 expression in anti-U1-70k-negative, anti-C negative and anti-SmB-negative MCTD patients was higher than in MCTD-positive patients. We observed negative correlations between serum IFN-α levels and TLR-7 expression and C3 and C4 levels in SLE patients. In SLE patients we observed that with increased IFN-γ, TLR-3 and TLR-8 expression increased the value of C3 and C4. Our results confirmed that the endosomal TLR-IFN pathway seems to be more important in SLE than in MCTD or SSc, and that IFN-α and IFN-β may be possible biomarkers for SLE.
Collapse
Affiliation(s)
- A Paradowska-Gorycka
- Department of Molecular Biology, National Institute of Geriatrics, Rheumatology and Rehabilitation, Warsaw, Poland
| | - A Wajda
- Department of Molecular Biology, National Institute of Geriatrics, Rheumatology and Rehabilitation, Warsaw, Poland
| | - B Stypinska
- Department of Molecular Biology, National Institute of Geriatrics, Rheumatology and Rehabilitation, Warsaw, Poland
| | - E Walczuk
- Department of Molecular Biology, National Institute of Geriatrics, Rheumatology and Rehabilitation, Warsaw, Poland
| | - E Rzeszotarska
- Department of Molecular Biology, National Institute of Geriatrics, Rheumatology and Rehabilitation, Warsaw, Poland
| | - M Walczyk
- Department of Connective Tissue Diseases, National Institute of Geriatrics, Rheumatology and Rehabilitation, Warsaw, Poland
| | - E Haladyj
- Eli Lilly and Company, Indianapolis, Indiana, USA
| | - K Romanowska-Prochnicka
- Department of Connective Tissue Diseases, National Institute of Geriatrics, Rheumatology and Rehabilitation, Warsaw, Poland.,Department of General and Experimental Pathology with Centre for Preclinical Research and Technology (CEPT), Medical University of Warsaw, Warsaw, Poland
| | - A Felis-Giemza
- Department of Connective Tissue Diseases, National Institute of Geriatrics, Rheumatology and Rehabilitation, Warsaw, Poland
| | - A Lewandowska
- Department of Connective Tissue Diseases, National Institute of Geriatrics, Rheumatology and Rehabilitation, Warsaw, Poland
| | - M Olesińska
- Department of Connective Tissue Diseases, National Institute of Geriatrics, Rheumatology and Rehabilitation, Warsaw, Poland
| |
Collapse
|
8
|
Spiera R, Hummers L, Chung L, Frech TM, Domsic R, Hsu V, Furst DE, Gordon J, Mayes M, Simms R, Lafyatis R, Martyanov V, Wood T, Whitfield ML, Constantine S, Lee E, Dgetluck N, White B. Safety and Efficacy of Lenabasum in a Phase II, Randomized, Placebo-Controlled Trial in Adults With Systemic Sclerosis. Arthritis Rheumatol 2020; 72:1350-1360. [PMID: 32336038 PMCID: PMC7497006 DOI: 10.1002/art.41294] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Accepted: 04/21/2020] [Indexed: 12/31/2022]
Abstract
Objective To assess the safety and efficacy of lenabasum in diffuse cutaneous systemic sclerosis (dcSSc). Methods A randomized, double‐blind, placebo‐controlled, phase II study was conducted at 9 SSc clinics in the US. Adults with dcSSc of ≤6 years’ duration who were receiving stable standard‐of‐care treatment were randomized to receive lenabasum (n = 27) or placebo (n = 15). Lenabasum doses were 5 mg once daily, 20 mg once daily, or 20 mg twice daily for 4 weeks, followed by 20 mg twice daily for 8 weeks. Safety and efficacy were assessed at weeks 4, 8, 12, and 16. Results Adverse events (AEs) occurred in 63% of the lenabasum group and 60% of the placebo group, with no serious AEs related to lenabasum. Compared to placebo, lenabasum treatment was associated with greater improvement in the American College of Rheumatology Combined Response Index in diffuse cutaneous Systemic Sclerosis (CRISS) score and other efficacy outcome measures that assessed overall disease, skin involvement, and patient‐reported function. The median CRISS score increased in the lenabasum group during the study, reaching 0.33, versus 0.00 in the placebo group, at week 16 (P = 0.07 by 2‐sided mixed‐effects model repeated‐measures analysis). Gene expression in inflammation and fibrosis pathways was reduced, and inflammation and fibrosis were improved on histologic evaluation of skin biopsy specimens, in the lenabasum group compared to the placebo group (all P ≤ 0.05). Conclusion Despite a short trial duration in a small number of patients in this phase II study in dcSSc, our findings indicate that lenabasum improves efficacy outcomes and underlying disease pathology with a favorable safety profile.
Collapse
Affiliation(s)
| | - Laura Hummers
- Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Lorinda Chung
- Stanford University School of Medicine, Stanford, California, and Palo Alto VA Health Care System, Palo Alto, California
| | - Tracy M Frech
- University of Utah, and Salt Lake City VA Health Care System
| | - Robyn Domsic
- University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Vivien Hsu
- Rutgers Robert Wood Johnson Medical School, New Brunswick, New Jersey
| | - Daniel E Furst
- Arthritis Association of Southern California, Los Angeles, California
| | | | - Maureen Mayes
- University of Texas Health Science Center at Houston
| | - Robert Simms
- Boston University School of Medicine, Boston, Massachusetts
| | - Robert Lafyatis
- University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | | | - Tammara Wood
- Geisel School of Medicine at Dartmouth, Hanover, New Hampshire
| | | | | | | | | | | |
Collapse
|
9
|
Carvalheiro T, Affandi AJ, Malvar-Fernández B, Dullemond I, Cossu M, Ottria A, Mertens JS, Giovannone B, Bonte-Mineur F, Kok MR, Marut W, Reedquist KA, Radstake TR, García S. Induction of Inflammation and Fibrosis by Semaphorin 4A in Systemic Sclerosis. Arthritis Rheumatol 2019; 71:1711-1722. [PMID: 31012544 PMCID: PMC6790618 DOI: 10.1002/art.40915] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Accepted: 04/18/2019] [Indexed: 11/11/2022]
Abstract
OBJECTIVE To analyze the potential role of semaphorin 4A (Sema4A) in inflammatory and fibrotic processes involved in the pathology of systemic sclerosis (SSc). METHODS Sema4A levels in the plasma of healthy controls (n = 11) and SSc patients (n = 20) were determined by enzyme-linked immunosorbent assay (ELISA). The expression of Sema4A and its receptors in monocytes and CD4+ T cells from healthy controls and SSc patients (n = 6-7 per group) was determined by ELISA and flow cytometry. Th17 cytokine production by CD4+ T cells (n = 5-7) was analyzed by ELISA and flow cytometry. The production of inflammatory mediators and extracellular matrix (ECM) components by dermal fibroblast cells (n = 6) was analyzed by quantitative polymerase chain reaction, ELISA, Western blotting, confocal microscopy, and ECM deposition assay. RESULTS Plasma levels of Sema4A, and Sema4A expression by circulating monocytes and CD4+ T cells, were significantly higher in SSc patients than in healthy controls (P < 0.05). Inflammatory mediators significantly up-regulated the secretion of Sema4A by monocytes and CD4+ T cells from SSc patients (P < 0.05 versus unstimulated SSc cells). Functional assays showed that Sema4A significantly enhanced the expression of Th17 cytokines induced by CD3/CD28 in total CD4+ T cells as well in different CD4+ T cell subsets (P < 0.05 versus unstimulated SSc cells). Finally, Sema4A induced a profibrotic phenotype in dermal fibroblasts from both healthy controls and SSc patients, which was abrogated by blocking or silencing the expression of Sema4A receptors. CONCLUSION Our findings indicate that Sema4A plays direct and dual roles in promoting inflammation and fibrosis, 2 main features of SSc, suggesting that Sema4A might be a novel therapeutic target in SSc.
Collapse
Affiliation(s)
- Tiago Carvalheiro
- University Medical Center Utrecht, University of Utrecht, Utrecht, The Netherlands
| | - Alsya J Affandi
- University Medical Center Utrecht, University of Utrecht, Utrecht, The Netherlands
| | | | - Ilse Dullemond
- University Medical Center Utrecht, University of Utrecht, Utrecht, The Netherlands
| | - Marta Cossu
- University Medical Center Utrecht, University of Utrecht, Utrecht, The Netherlands
| | - Andrea Ottria
- University Medical Center Utrecht, University of Utrecht, Utrecht, The Netherlands
| | - Jorre S Mertens
- University Medical Center Utrecht, University of Utrecht, Utrecht, The Netherlands
| | - Barbara Giovannone
- University Medical Center Utrecht, University of Utrecht, Utrecht, The Netherlands
| | | | - Marc R Kok
- Maasstad Hospital Rotterdam, Rotterdam, The Netherlands
| | - Wioleta Marut
- University Medical Center Utrecht, University of Utrecht, Utrecht, The Netherlands
| | - Kris A Reedquist
- University Medical Center Utrecht, University of Utrecht, Utrecht, The Netherlands
| | - Timothy R Radstake
- University Medical Center Utrecht, University of Utrecht, Utrecht, The Netherlands
| | - Samuel García
- University Medical Center Utrecht, University of Utrecht, Utrecht, The Netherlands
| |
Collapse
|
10
|
Ramani K, Biswas PS. Interleukin-17: Friend or foe in organ fibrosis. Cytokine 2019; 120:282-288. [PMID: 30772195 DOI: 10.1016/j.cyto.2018.11.003] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Revised: 11/02/2018] [Accepted: 11/05/2018] [Indexed: 02/07/2023]
Abstract
Fibrosis affects all vital organs accounting for a staggering 45% of deaths worldwide and no effective therapies are currently available. Unresolved inflammation triggers downstream signaling events that lead to organ fibrosis. In recent years, proinflammatory cytokine Interleukin-17 (IL-17) has been implicated in several chronic inflammatory diseases that often culminate in organ damage followed by impaired wound healing and fibrosis. In this review, we outline the contribution of the IL-17 in mediating fibrotic diseases in various organs. A comprehensive understanding of the inflammatory events, and particularly the details of IL-17 signaling in vivo, could be beneficial in designing new therapeutic or preventive approaches to treat fibrosis. Additionally, understanding organ-specific differences in IL-17 activity could lead to targeted therapies and help spare other organs from unwanted side effects.
Collapse
Affiliation(s)
- Kritika Ramani
- Division of Rheumatology and Clinical Immunology, Department of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Partha S Biswas
- Division of Rheumatology and Clinical Immunology, Department of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA.
| |
Collapse
|
11
|
RNA Modifications Modulate Activation of Innate Toll-Like Receptors. Genes (Basel) 2019; 10:genes10020092. [PMID: 30699960 PMCID: PMC6410116 DOI: 10.3390/genes10020092] [Citation(s) in RCA: 77] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Revised: 01/23/2019] [Accepted: 01/25/2019] [Indexed: 12/13/2022] Open
Abstract
Self/foreign discrimination by the innate immune system depends on receptors that identify molecular patterns as associated to pathogens. Among others, this group includes endosomal Toll-like receptors, among which Toll-like receptors (TLR) 3, 7, 8, and 13 recognize and discriminate mammalian from microbial, potentially pathogen-associated, RNA. One of the discriminatory principles is the recognition of endogenous RNA modifications. Previous work has identified a couple of RNA modifications that impede activation of TLR signaling when incorporated in synthetic RNA molecules. Of note, work that is more recent has now shown that RNA modifications in their naturally occurring context can have immune-modulatory functions: Gm, a naturally occurring ribose-methylation within tRNA resulted in a lack of TLR7 stimulation and within a defined sequence context acted as antagonist. Additional RNA modifications with immune-modulatory functions have now been identified and recent work also indicates that RNA modifications within the context of whole prokaryotic or eukaryotic cells are indeed used for immune-modulation. This review will discuss new findings and developments in the field of immune-modulatory RNA modifications.
Collapse
|
12
|
Garcia S. Role of Semaphorins in Immunopathologies and Rheumatic Diseases. Int J Mol Sci 2019; 20:ijms20020374. [PMID: 30654587 PMCID: PMC6359241 DOI: 10.3390/ijms20020374] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2018] [Revised: 01/14/2019] [Accepted: 01/14/2019] [Indexed: 12/17/2022] Open
Abstract
Rheumatic diseases are disorders characterized by joint inflammation, in which other organs are also affected. There are more than two hundred rheumatic diseases, the most studied so far are rheumatoid arthritis, osteoarthritis, spondyloarthritis, systemic lupus erythematosus, and systemic sclerosis. The semaphorin family is a large group of proteins initially described as axon guidance molecules involved in nervous system development. Studies have demonstrated that semaphorins play a role in other processes such as the regulation of immunity, angiogenesis, bone remodeling, apoptosis, and cell migration and invasion. Moreover, semaphorins have been related to the pathogenesis of multiple sclerosis, asthma, Alzheimer, myocarditis, atherosclerosis, fibrotic diseases, osteopetrosis, and cancer. The aim of this review is to summarize current knowledge regarding the role of semaphorins in rheumatic diseases, and discuss their potential applications as therapeutic targets to treat these disorders.
Collapse
Affiliation(s)
- Samuel Garcia
- Department of Rheumatology and Clinical Immunology and Laboratory of Translational Immunology, University Medical Center Utrecht, University of Utrecht, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands.
| |
Collapse
|
13
|
van den Hombergh WMT, Kersten BE, Knaapen-Hans HKA, Thurlings RM, van der Kraan PM, van den Hoogen FHJ, Fransen J, Vonk MC. Hit hard and early: analysing the effects of high-dose methylprednisolone on nailfold capillary changes and biomarkers in very early systemic sclerosis: study protocol for a 12-week randomised controlled trial. Trials 2018; 19:449. [PMID: 30134971 PMCID: PMC6104002 DOI: 10.1186/s13063-018-2798-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Accepted: 07/09/2018] [Indexed: 01/08/2023] Open
Abstract
Background Mounting evidence indicates that inflammatory mechanisms drive systemic sclerosis (SSc) vasculopathy and fibrosis, especially early in the disease. Therefore, patients with very early SSc could benefit from early treatments targeting inflammation. Glucocorticoids are among the most potent anti-inflammatory and immunosuppressive agents. Several studies have demonstrated a mixed response to treatment with glucocorticoids in SSc, probably because it is seldom initiated at very early stages of the disease. We hypothesise that by inhibiting the inflammatory process driving SSc disease progression, glucocorticoid treatments will induce remission in patients with very early SSc. Methods/design This study is a 12-week, randomised, double-blind, placebo-controlled trial analysing the effects of high-dose intravenous methylprednisolone in very early SSc. Thirty patients who fulfil the criteria for very early SSc will be randomly assigned in a 2:1 ratio to receive either intravenous methylprednisolone or a placebo on three consecutive days over three consecutive months. In this study, the primary endpoint will be the change in capillary density between the baseline and after 12 weeks of treatment. The secondary outcomes of this study are a change in selected biomarkers, other changes in the nailfold capillaries, signs of established SSc and changes in physical function, general health and utilities, as reported through questionnaires. Discussion This trial is the first aiming to treat very early SSc and is promising because it targets the very early stages of the disease process by using an inexpensive and relatively safe treatment known to be highly effective against inflammation. The use of vasculopathy and inflammatory biomarkers as well as clinical signs and symptoms as the endpoints in our study enables us to meet the patient need for markers of disease activity. If it is possible to prevent clinically significant disease in patients with very early SSc by using a safe treatment, this will cause a paradigm shift in scleroderma care and research. Trial registration ClinicalTrials.gov Identifier: NCT03059979. Registered on 20 February 2017. Electronic supplementary material The online version of this article (10.1186/s13063-018-2798-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Wieneke M T van den Hombergh
- Department of Rheumatology, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525, GA, Nijmegen, The Netherlands.
| | - Brigit E Kersten
- Department of Rheumatology, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525, GA, Nijmegen, The Netherlands
| | - Hanneke K A Knaapen-Hans
- Department of Rheumatology, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525, GA, Nijmegen, The Netherlands
| | - Rogier M Thurlings
- Department of Rheumatology, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525, GA, Nijmegen, The Netherlands
| | - Peter M van der Kraan
- Experimental Rheumatology, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525, GA, Nijmegen, The Netherlands
| | - Frank H J van den Hoogen
- Department of Rheumatology, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525, GA, Nijmegen, The Netherlands
| | - Jaap Fransen
- Department of Rheumatology, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525, GA, Nijmegen, The Netherlands
| | - Madelon C Vonk
- Department of Rheumatology, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525, GA, Nijmegen, The Netherlands
| |
Collapse
|
14
|
Meiners S, Evankovich J, Mallampalli RK. The ubiquitin proteasome system as a potential therapeutic target for systemic sclerosis. Transl Res 2018; 198:17-28. [PMID: 29702079 DOI: 10.1016/j.trsl.2018.03.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Revised: 03/20/2018] [Accepted: 03/24/2018] [Indexed: 01/16/2023]
Abstract
The present review aims to summarize available knowledge on the role of the ubiquitin-proteasome system (UPS) in the pathogenesis of scleroderma and scleroderma-related disease mechanisms. This will provide the reader with a more mechanistic understanding of disease pathogenesis and help to identify putative novel targets within the UPS for potential therapeutic intervention. Because of the heterogenous manifestations of scleroderma, we will primarily focus on conserved mechanisms that are involved in the development of lung scleroderma phenotypes.
Collapse
Affiliation(s)
- Silke Meiners
- Comprehensive Pneumology Center (CPC), University Hospital, Ludwig Maximilians University, Helmholtz Zentrum München, Germany; Comprehensive Pneumology Center, Munich (CPC-M), Germany; Member of the German Center for Lung Research (DZL), Munich, Germany.
| | - John Evankovich
- Pulmonary, Allergy, and Critical Care Medicine, Acute Lung Injury Center of Excellence, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Rama K Mallampalli
- Pulmonary, Allergy, and Critical Care Medicine, Acute Lung Injury Center of Excellence, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA; Department of Cell Biology and Physiology, University of Pittsburgh, Pittsburgh, PA, USA; Medical Specialty Service Line, Veterans Affairs Pittsburgh Healthcare System, Pittsburgh, PA, USA.
| |
Collapse
|
15
|
Taniguchi T, Miyagawa T, Tamaki Z, Nakamura K, Yamashita T, Saigusa R, Takahashi T, Toyama T, Ichimura Y, Yoshizaki A, Tada Y, Sugaya M, Kadono T, Sato S, Asano Y. A possible implication of reduced levels of LIF, LIFR, and gp130 in vasculopathy related to systemic sclerosis. Arch Dermatol Res 2017; 309:833-842. [DOI: 10.1007/s00403-017-1786-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Revised: 09/10/2017] [Accepted: 10/04/2017] [Indexed: 12/18/2022]
|
16
|
Krasimirova E, Velikova T, Ivanova-Todorova E, Tumangelova-Yuzeir K, Kalinova D, Boyadzhieva V, Stoilov N, Yoneva T, Rashkov R, Kyurkchiev D. Treg/Th17 cell balance and phytohaemagglutinin activation of T lymphocytes in peripheral blood of systemic sclerosis patients. World J Exp Med 2017; 7:84-96. [PMID: 28890870 PMCID: PMC5571452 DOI: 10.5493/wjem.v7.i3.84] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Revised: 05/26/2017] [Accepted: 07/03/2017] [Indexed: 02/06/2023] Open
Abstract
AIM To investigate T-cell activation, the percentage of peripheral T regulatory cells (Tregs), Th17 cells and the circulating cytokine profile in systemic sclerosis (SSc).
METHODS We enrolled a total of 24 SSc patients and 16 healthy controls in the study and divided the patients as having diffuse cutaneous SSc (dcSSc, n = 13) or limited cutaneous SSc (lcSSc, n = 11). We performed a further subdivision of the patients regarding the stage of the disease - early, intermediate or late. Peripheral venous blood samples were collected from all subjects. We performed flow cytometric analysis of the activation capacity of T-lymphocytes upon stimulation with PHA-M and of the percentage of peripheral Tregs and Th17 cells in both patients and healthy controls. We used ELISA to quantitate serum levels of human interleukin (IL)-6, IL-10, tissue growth factor-β1 (TGF-β1), and IL-17A.
RESULTS We identified a decreased percentage of CD3+CD69+ cells in PHA-stimulated samples from SSc patients in comparison with healthy controls (13.35% ± 2.90% vs 37.03% ± 2.33%, P < 0.001). However, we did not establish a correlation between the down-regulated CD3+CD69+ cells and the clinical subset, nor regarding the stage of the disease. The activated CD4+CD25+ peripheral lymphocytes were represented in decreased percentage in patients when compared to controls (6.30% ± 0.68% vs 9.36% ± 1.08%, P = 0.016). Regarding the forms of the disease, dcSSc patients demonstrated lower frequency of CD4+CD25+ T cells against healthy subjects (5.95% ± 0.89% vs 9.36% ± 1.08%, P = 0.025). With regard to Th17 cells, our patients demonstrated increased percentage in comparison with controls (18.13% ± 1.55% vs 13.73% ± 1.21%, P = 0.031). We detected up-regulated Th17 cells within the lcSSc subset against controls (20.46% ± 2.41% vs 13.73% ± 1.21%, P = 0.025), nevertheless no difference was found between dcSSc and lcSSc patients. Flow cytometric analysis revealed an increased percentage of CD4+CD25-Foxp3+ in dcSSc patients compared to controls (10.94% ± 1.65% vs 6.88% ± 0.91, P = 0.032). Regarding the peripheral cytokine profile, we detected raised levels of IL-6 [2.10 (1.05-4.60) pg/mL vs 0.00 pg/mL, P < 0.001], TGF-β1 (19.94 ± 3.35 ng/mL vs 10.03 ± 2.25 ng/mL, P = 0.02), IL-10 (2.83 ± 0.44 pg/mL vs 0.68 ± 0.51 pg/mL, P = 0.008), and IL-17A [6.30 (2.50-15.60) pg/mL vs 0 (0.00-0.05) pg/mL, P < 0.001] in patients when compared to healthy controls. Furthermore, we found increased circulating IL-10, TGF-β, IL-6 and IL-17A in the lcSSc subset vs control subjects, as it follows: IL-10 (3.32 ± 0.59 pg/mL vs 0.68 ± 0.51 pg/mL, P = 0.003), TGF-β1 (22.82 ± 4.99 ng/mL vs 10.03 ± 2.25 ng/mL, P = 0.031), IL-6 [2.08 (1.51-4.69) pg/mL vs 0.00 pg/mL, P < 0.001], and IL-17A [14.50 (8.55-41.65) pg/mL vs 0.00 (0.00-0.05) pg/mL, P < 0.001]. Furthermore, circulating IL-17A was higher in lcSSc as opposed to dcSSc subset (31.99 ± 13.29 pg/mL vs 7.14 ± 3.01 pg/mL, P = 0.008). Within the dcSSc subset, raised levels of IL-17A and IL-6 were detected vs healthy controls: IL-17A [2.60 (0.45-9.80) pg/mL vs 0.00 (0.00-0.05) pg/mL, P < 0.001], IL-6 [2.80 (1.03-7.23) pg/mL vs 0.00 pg/mL, P < 0.001]. Regarding the stages of the disease, TGF-β1 serum levels were increased in early stage against late stage, independently from the SSc phenotype (30.03 ± 4.59 ng/mL vs 13.08 ± 4.50 ng/mL, P = 0.017).
CONCLUSION It is likely that the altered percentage of Th17 and CD4+CD25-FoxP3+ cells along with the peripheral cytokine profile in patients with SSc may play a key role in the pathogenesis of the disease.
Collapse
|
17
|
Sirt1 ameliorates systemic sclerosis by targeting the mTOR pathway. J Dermatol Sci 2017; 87:149-158. [PMID: 28532580 DOI: 10.1016/j.jdermsci.2017.04.013] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2016] [Revised: 03/26/2017] [Accepted: 04/25/2017] [Indexed: 01/17/2023]
Abstract
BACKGROUND Systemic sclerosis (SSc) is a chronic autoimmune disease characterized by inflammation and fibrosis. Our previous research has indicated that Sirtuin1 (Sirt1) plays a role in the regulation of TNF-α-induced inflammation; however, whether Sirt1 may inhibit the progress of SSc by blocking inflammation remains unknown. OBJECTIVE We aimed to investigate the function of Sirt1 in SSc. METHODS The function and its mechanism of Sirt1 were evaluated in fibroblasts or scleroderma mice. The expression of Sirt1 and cytokines was analyzed using real-time PCR, western blot, ELISA and immunohistochemistry. RESULTS We determined that fibroblasts of SSc patients were activated to exhibit inflammation. Sirt1, activated by resveratrol (Res), ameliorated cutaneous inflammation and fibrosis in bleomycin (BLM)-induced scleroderma mice. An improvement in mammalian target of rapamycin (mTOR) was identified in the fibroblasts of SSc patients and the skin lesions of BLM mice. Rapamycin, an mTOR specific inhibitor, substantially inhibited the induced inflammation and fibrosis. The enhancement of mTOR expression in the skin lesions of the BLM-treated mice was significantly inhibited by Sirt1 activation. However, in both the BLM-treated cells and mice, Res exerted an inhibitory function on the expression of inflammatory factors, and collagen was diminished following mTOR knockdown. These findings suggest that Res may inhibit inflammation and fibrosis via mTOR. CONCLUSION The modulation of Sirt1 activity may represent a potential therapeutic method for SSc. The mechanism may involve the inhibition of mTOR phosphorylation, whereas mTOR activity was shown to be a pathogenic culprit of SSc.
Collapse
|
18
|
Fu B, Liu ZL, Zhang H, Gu F. Interleukin-13 and age-related macular degeneration. Int J Ophthalmol 2017; 10:535-540. [PMID: 28503424 DOI: 10.18240/ijo.2017.04.06] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2015] [Accepted: 12/03/2016] [Indexed: 02/07/2023] Open
Abstract
AIM o identify the effects of interleukin (IL)-13 on retinal pigment epithelial (RPE) cells and the IL-13 level in aqueous humor of age-related macular degeneration (AMD) patients. METHODS IL-13 levels in aqueous humor specimens from AMD patients were detected with enzyme-linked immunosorbent assay (ELISA). ARPE-19 cells were treated with 10 ng/mL IL-13 for 12, 24, and 48h. The cell proliferaton was evaluated by the MTS method. The mRNA and protein levels of α-SMA and ZO-1 were evaluated with quantitative real-time polymerase chain reaction (qRT-PCR) and Western blot respectively. The expression of tumor necrosis factor-α (TNF-α), transforming growth factor-β (TGF-β) and vascular endothelial growth factor (VEGF) were assessed by ELISA. RESULTS IL-13 levels in the aqueous humor of patients with AMD were significantly higher than those in the control (167.33±17.64 vs 27.12±5.65 pg/mL; P<0.01). In vitro, IL-13 of high concentrations (10, 15, and 20 ng/mL) inhibited ARPE-19 cell proliferation. α-SMA mRNA in ARPE-19 cell were increased (1.017±0.112 vs 1.476±0.168; P<0.001) and ZO-1 decreased (1.051±0.136 vs 0.702±0.069; P<0.001) after treated with 10 ng/mL IL-13 for 48h. The protein expression of α-SMA and ZO-1 also showed the same tendency (α-SMA: P=0.038; ZO-1: P=0.008). IL-13 significantly reduced the level of TNF-α (44.70±1.67 vs 31.79±3.53 pg/mL; P=0.005) at 48h, but the level of TGF-β2 was significantly increased from 34.44±2.92 to 57.61±6.31 pg/mL at 24h (P=0.004) and from 61.26±1.11 to 86.91±3.59 pg/mL at 48h (P<0.001). While expressions of VEGF didn't change after IL-13 treatment. CONCLUSION IL-13 in vitro inhibit ARPE-19 cell proliferation and expression in the aqueous may be associated with AMD.
Collapse
Affiliation(s)
- Bo Fu
- China Medical University, Shenyang 110001, Liaoning Province, China
| | - Zhe-Li Liu
- China Medical University, Shenyang 110001, Liaoning Province, China
| | - Han Zhang
- Department of Ophthalmology, the First Hospital of China Medical University, Shenyang 110001, Liaoning Province, China
| | - Feng Gu
- Department of Ophthalmology, the First Hospital of China Medical University, Shenyang 110001, Liaoning Province, China
| |
Collapse
|
19
|
Hossen MJ, Yang WS, Kim D, Aravinthan A, Kim JH, Cho JY. Thymoquinone: An IRAK1 inhibitor with in vivo and in vitro anti-inflammatory activities. Sci Rep 2017; 7:42995. [PMID: 28216638 PMCID: PMC5316937 DOI: 10.1038/srep42995] [Citation(s) in RCA: 100] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Accepted: 01/18/2017] [Indexed: 12/14/2022] Open
Abstract
Thymoquinone (TQ) is a bioactive component of black seed (Nigella sativa) volatile oil and has been shown to have anti-oxidative, anti-inflammatory, and anti-cancer properties. In the present study, we explored the molecular mechanisms that underlie the anti-inflammatory effect of TQ and its target proteins using lipopolysaccharide (LPS)-stimulated murine macrophage-like RAW264.7 and human monocyte-like U937 cells, together with LPS/D-galactosamine (GalN)-induced acute hepatitis and HCl/EtOH-induced gastritis mouse models. TQ strongly inhibited the production of nitric oxide (NO) and repressed NO synthase (iNOS), tumor necrosis factor (TNF)-α, cyclooxygenase (COX)−2, interleukin (IL)−6, and IL-1β expression in LPS-activated RAW264.7 cells. Treatment of LPS/D-GalN–induced hepatitis and EtOH/HCl–induced gastritis mouse models with TQ significantly ameliorated disease symptoms. Using luciferase reporter gene assays, we also showed that the nuclear levels of transcription factors and phosphorylation patterns of signaling proteins, activator protein (AP)−1, and nuclear factor (NF)-κB pathways were all affected by TQ treatment. Finally, we used additional kinase and luciferase validation assays with interleukin-1 receptor-associated kinase 1 (IRAK1) to show that IRAK1 is directly suppressed by TQ treatment. Together, these findings strongly suggest that the anti-inflammatory actions of TQ are caused by suppression of IRAK-linked AP-1/NF-κB pathways.
Collapse
Affiliation(s)
- Muhammad Jahangir Hossen
- Department of Genetic Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea.,Department of Animal Science, Patuakhali Science and Technology University, Dumki, Patuakhali 8602, Bangladesh
| | - Woo Seok Yang
- Department of Genetic Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Daewon Kim
- Laboratory of Bio-informatics, Department of Multimedia Engineering, Dankook University, Cheonan 31116, Republic of Korea
| | - Adithan Aravinthan
- Department of Physiology, College of Veterinary Medicine, Chonbuk National University, Iksan 54596, Republic of Korea
| | - Jong-Hoon Kim
- Department of Physiology, College of Veterinary Medicine, Chonbuk National University, Iksan 54596, Republic of Korea
| | - Jae Youl Cho
- Department of Genetic Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea
| |
Collapse
|
20
|
Mohammad Hosseini A, Majidi J, Baradaran B, Yousefi M. Toll-Like Receptors in the Pathogenesis of Autoimmune Diseases. Adv Pharm Bull 2015; 5:605-14. [PMID: 26793605 DOI: 10.15171/apb.2015.082] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2014] [Revised: 07/02/2014] [Accepted: 07/21/2014] [Indexed: 12/15/2022] Open
Abstract
Human Toll-like receptors (TLRs) are a family of transmembrane receptors, which play a key role in both innate and adaptive immune responses. Beside of recognizing specific molecular patterns that associated with different types of pathogens, TLRs may also detect a number of self-proteins and endogenous nucleic acids. Activating TLRs lead to the heightened expression of various inflammatory genes, which have a protective role against infection. Data rising predominantly from human patients and animal models of autoimmune disease indicate that, inappropriate triggering of TLR pathways by exogenous or endogenous ligands may cause the initiation and/or perpetuation of autoimmune reactions and tissue damage. Given their important role in infectious and non-infectious disease process, TLRs and its signaling pathways emerge as appealing targets for therapeutics. In this review, we demonstrate how TLRs pathways could be involved in autoimmune disorders and their therapeutic application.
Collapse
Affiliation(s)
| | - Jafar Majidi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mehdi Yousefi
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran. ; Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
21
|
Takahashi T, Asano Y, Noda S, Aozasa N, Akamata K, Taniguchi T, Ichimura Y, Toyama T, Sumida H, Kuwano Y, Tada Y, Sugaya M, Kadono T, Sato S. A possible contribution of lipocalin-2 to the development of dermal fibrosis, pulmonary vascular involvement and renal dysfunction in systemic sclerosis. Br J Dermatol 2015; 173:681-9. [PMID: 25781362 DOI: 10.1111/bjd.13779] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/09/2015] [Indexed: 12/16/2022]
Abstract
BACKGROUND Lipocalin-2 is an adipocytokine implicated in apoptosis, innate immunity, angiogenesis, and the development of chronic kidney disease. OBJECTIVES To investigate the role of lipocalin-2 in systemic sclerosis (SSc). MATERIALS AND METHODS Serum lipocalin-2 levels were determined by enzyme-linked immunosorbent assay in 50 patients with SSc and 19 healthy subjects. Lipocalin-2 expression was evaluated in the skin of patients with SSc and bleomycin (BLM)-treated mice and in Fli1-deficient endothelial cells by reverse transcriptase-real time polymerase chain reaction, immunoblotting and/or immunohistochemistry. RESULTS Although serum lipocalin-2 levels were comparable between patients with SSc and healthy controls, the prevalence of scleroderma renal crisis was significantly higher in patients with SSc with elevated serum lipocalin-2 levels than in those with normal levels. Furthermore, serum lipocalin-2 levels inversely correlated with estimated glomerular filtration rate in patients with SSc with renal dysfunction. Among patients with SSc with normal renal function, serum lipocalin-2 levels positively correlated with skin score in patients with diffuse cutaneous SSc with disease duration of < 3 years and inversely correlated with estimated right ventricular systolic pressure in total patients with SSc. Importantly, in SSc lesional skin, lipocalin-2 expression was increased in dermal fibroblasts and endothelial cells. In BLM-treated mice, lipocalin-2 was highly expressed in dermal fibroblasts, but not in endothelial cells. On the other hand, the deficiency of transcription factor Fli1, which is implicated in SSc vasculopathy, induced lipocalin-2 expression in cultivated endothelial cells. CONCLUSIONS Lipocalin-2 may be involved in renal dysfunction and dermal fibrosis of SSc. Dysregulated matrix metalloproteinase-9/lipocalin-2-dependent angiogenesis due to Fli1 deficiency may contribute to the development of pulmonary arterial hypertension associated with SSc.
Collapse
Affiliation(s)
- T Takahashi
- Department of Dermatology, University of Tokyo Graduate School of Medicine, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Y Asano
- Department of Dermatology, University of Tokyo Graduate School of Medicine, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - S Noda
- Department of Dermatology, University of Tokyo Graduate School of Medicine, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - N Aozasa
- Department of Dermatology, University of Tokyo Graduate School of Medicine, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - K Akamata
- Department of Dermatology, University of Tokyo Graduate School of Medicine, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - T Taniguchi
- Department of Dermatology, University of Tokyo Graduate School of Medicine, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Y Ichimura
- Department of Dermatology, University of Tokyo Graduate School of Medicine, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - T Toyama
- Department of Dermatology, University of Tokyo Graduate School of Medicine, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - H Sumida
- Department of Dermatology, University of Tokyo Graduate School of Medicine, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Y Kuwano
- Department of Dermatology, University of Tokyo Graduate School of Medicine, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Y Tada
- Department of Dermatology, University of Tokyo Graduate School of Medicine, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - M Sugaya
- Department of Dermatology, University of Tokyo Graduate School of Medicine, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - T Kadono
- Department of Dermatology, University of Tokyo Graduate School of Medicine, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - S Sato
- Department of Dermatology, University of Tokyo Graduate School of Medicine, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| |
Collapse
|
22
|
Coelho Horimoto AM, da Costa IP. Autoanticorpos em esclerose sistêmica e sua correlação com as manifestações clínicas da doença em pacientes do Centro-Oeste do Brasil. REVISTA BRASILEIRA DE REUMATOLOGIA 2015; 55:229-39. [DOI: 10.1016/j.rbr.2014.09.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2014] [Revised: 09/02/2014] [Accepted: 09/21/2014] [Indexed: 02/03/2023] Open
|
23
|
Arismendi M, Giraud M, Ruzehaji N, Dieudé P, Koumakis E, Ruiz B, Airo P, Cusi D, Matucci-Cerinic M, Salvi E, Cuomo G, Hachulla E, Diot E, Caramaschi P, Riccieri V, Avouac J, Kayser C, Allanore Y. Identification of NF-κB and PLCL2 as new susceptibility genes and highlights on a potential role of IRF8 through interferon signature modulation in systemic sclerosis. Arthritis Res Ther 2015; 17:71. [PMID: 25880423 PMCID: PMC4422604 DOI: 10.1186/s13075-015-0572-y] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2014] [Accepted: 02/20/2015] [Indexed: 12/15/2022] Open
Abstract
INTRODUCTION Systemic sclerosis (SSc) and primary biliary cirrhosis (PBC) are rare polygenic autoimmune diseases (AIDs) characterized by fibroblast dysfunction. Furthermore, both diseases share some genetic bases with other AIDs, as evidenced by autoimmune gene pleiotropism. The present study was undertaken to investigate whether single-nucleotide polymorphisms (SNPs) identified by a large genome-wide association study (GWAS) in PBC might contribute to SSc susceptibility. METHODS Sixteen PBC susceptibility SNPs were genotyped in a total of 1,616 patients with SSc and 3,621 healthy controls from two European populations (France and Italy). RESULTS We observed an association between PLCL2 rs1372072 (odds ratio (OR) = 1.22, 95% confidence interval (CI) 1.12 to 1.33, P adj = 7.22 × 10(-5)), nuclear factor-kappa-B (NF-κB) rs7665090 (OR = 1.15, 95% CI 1.06 to 1.25, P adj = 0.01), and IRF8 rs11117432 (OR = 0.75, 95% CI 0.67 to 0.86, P adj = 2.49 × 10(-4)) with SSc susceptibility. Furthermore, phenotype stratification showed an association between rs1372072 and rs11117432 with the limited cutaneous subgroup (lcSSc) (P adj = 4.45 × 10(-4) and P adj = 0.001), whereas rs7665090 was associated with the diffuse cutaneous subtype (dcSSc) (P adj = 0.003). Genotype-mRNA expression correlation analysis revealed that the IRF8 protective allele was associated with increased interferon-gamma (IFN-γ) expression (P = 0.03) in patients with SSc but decreased type I IFN (IFIT1) expression in patients and controls (P = 0.02). In addition, we found an epistatic interaction between NF-κB and IRF8 (OR = 0.56, 95% CI 0.00 to 0.74, P = 4 × 10(-4)) which in turn revealed that the IRF8 protective effect is dependent on the presence of the NF-κB susceptibility allele. CONCLUSIONS An analysis of pleiotropic genes identified two new susceptibility genes for SSc (NF-κB and PLCL2) and confirmed the IRF8 locus. Furthermore, the IRF8 variant influenced the IFN signature, and we found an interaction between IRF8 and NF-κB gene variants that might play a role in SSc susceptibility.
Collapse
Affiliation(s)
- Maria Arismendi
- Paris Descartes University, INSERM U1016, Institut Cochin, Sorbonne Paris Cité, Paris, France. .,CAPES Foundation, Ministry of Education of Brazil, Brasília, DF, 70040-020, Brazil.
| | - Matthieu Giraud
- Paris Descartes University, INSERM U1016, Institut Cochin, Sorbonne Paris Cité, Paris, France.
| | - Nadira Ruzehaji
- Paris Descartes University, INSERM U1016, Institut Cochin, Sorbonne Paris Cité, Paris, France.
| | - Philippe Dieudé
- Paris Diderot University, Rheumatology Department, Hôpital Bichat Claude Bernard, APHP, Paris, France. .,Paris Diderot University, INSERM U699, Hôpital Bichat Claude Bernard, Paris, France.
| | - Eugenie Koumakis
- Paris Descartes University, INSERM U1016, Institut Cochin, Sorbonne Paris Cité, Paris, France.
| | - Barbara Ruiz
- Paris Descartes University, INSERM U1016, Institut Cochin, Sorbonne Paris Cité, Paris, France.
| | - Paolo Airo
- Rheumatology and Clinical Immunology, Spedali Civili, Brescia, Italy.
| | - Daniele Cusi
- University of Milano, Department of Medicine, Surgery and Dentistry San Paolo & Genomics and Bioinformatics Platform, Fondazione Filarete, Milan, Italy.
| | - Marco Matucci-Cerinic
- Department of Biomedicine & Division of Rheumatology AOUC, Department of Rheumatology AVC, Department of Medicine & Denothe Centre, University of Florence, Florence, Italy.
| | - Erika Salvi
- University of Milano, Department of Medicine, Surgery and Dentistry San Paolo & Genomics and Bioinformatics Platform, Fondazione Filarete, Milan, Italy.
| | - Giovanna Cuomo
- Department of Clinical and Experimental Medicine, Rheumatology Unit, Second University of Naples, Naples, Italy.
| | - Eric Hachulla
- Université Lille II, Médecine Interne, Lille, France.
| | | | - Paola Caramaschi
- Rheumatology Unit, Azienda Ospedaliera Universitaria Integrata di Verona, Verona, Italy.
| | - Valeria Riccieri
- Division of Rheumatology, Department of Medical Clinic and Therapy, University "Sapienza" of Rome, Rome, Italy.
| | - Jérôme Avouac
- Paris Descartes University, INSERM U1016, Institut Cochin, Sorbonne Paris Cité, Paris, France. .,Paris Descartes University, Rheumatology A Department, Cochin Hospital, APHP, Paris, France.
| | - Cristiane Kayser
- Department of Rheumatology, Federal University of São Paulo, São Paulo, Brazil.
| | - Yannick Allanore
- Paris Descartes University, INSERM U1016, Institut Cochin, Sorbonne Paris Cité, Paris, France. .,Paris Descartes University, Rheumatology A Department, Cochin Hospital, APHP, Paris, France.
| |
Collapse
|
24
|
Pérez-Frías A, González-Tajuelo R, Núñez-Andrade N, Tejedor R, García-Blanco MJ, Vicente-Rabaneda E, Castañeda S, Gamallo C, Silván J, Esteban-Villafruela A, Cubero-Rueda L, García-García C, Muñoz-Calleja C, García-Diez A, Urzainqui A. Development of an autoimmune syndrome affecting the skin and internal organs in P-selectin glycoprotein ligand 1 leukocyte receptor-deficient mice. Arthritis Rheumatol 2015; 66:3178-89. [PMID: 25132671 DOI: 10.1002/art.38808] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2014] [Accepted: 07/29/2014] [Indexed: 12/20/2022]
Abstract
OBJECTIVE To define and characterize the progression of the spontaneous autoimmune disease that develops in mice in the absence of the leukocyte adhesion receptor P-selectin glycoprotein ligand 1 (PSGL-1). METHODS Skin-resident immune cells from PSGL-1-deficient mice and C57BL/6 control mice of different ages were isolated and analyzed by flow cytometry. Biochemical parameters were analyzed in mouse serum and urine, and the presence of serum autoantibodies was investigated. Skin and internal organs were extracted, and their structure was analyzed histologically. RESULTS Skin-resident innate and adaptive immune cells from PSGL-1(-/-) mice had a proinflammatory phenotype with an imbalanced T effector cell:Treg cell ratio. Sera from PSGL-1(-/-) mice had circulating autoantibodies commonly detected in connective tissue-related human autoimmune diseases. Biochemical and histologic analysis of skin and internal organs revealed skin fibrosis and structural and functional abnormalities in the lungs and kidneys. Furthermore, PSGL-1(-/-) mice exhibited vascular alterations, showing loss of dermal vessels, small vessel medial layer remodeling in the lungs and kidneys, and ischemic processes in the kidney that promote renal infarcts. CONCLUSION Our study demonstrates that immune system overactivation due to PSGL-1 deficiency triggers an autoimmune syndrome with characteristics similar to systemic sclerosis, including skin fibrosis, vascular alterations, and systemic organ involvement. These results suggest that PSGL-1 expression contributes to the maintenance of the homeostasis of the immune system and could act as a barrier for autoimmunity in mice.
Collapse
Affiliation(s)
- A Pérez-Frías
- Fundación de Investigación Biomédica, Instituto de Investigación Sanitaria-Princesa, and Hospital de la Princesa, Madrid, Spain
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Kowal-Bielecka O, Bielecki M, Guiducci S, Trzcinska-Butkiewicz B, Michalska-Jakubus M, Matucci-Cerinic M, Brzosko M, Krasowska D, Chyczewski L, Kowal K. High serum sCD163/sTWEAK ratio is associated with lower risk of digital ulcers but more severe skin disease in patients with systemic sclerosis. Arthritis Res Ther 2014; 15:R69. [PMID: 23800379 PMCID: PMC4060194 DOI: 10.1186/ar4246] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2012] [Revised: 02/21/2013] [Accepted: 06/24/2013] [Indexed: 11/10/2022] Open
Abstract
INTRODUCTION Systemic sclerosis (SSc) is an autoimmune disease characterized by chronic inflammation, vascular injury and excessive fibrosis. CD163 is a scavenger receptor which affects inflammatory response and may contribute to connective tissue remodelling. It has recently been demonstrated that CD163 can bind and neutralize the TNF-like weak inducer of apoptosis (TWEAK), a multifunctional cytokine which regulates inflammation, angiogenesis and tissue remodelling. We aimed to investigate the relationships between serum levels of soluble CD163 (sCD163) and soluble TWEAK (sTWEAK) in relation to disease manifestations in SSc patients. METHODS This study included 89 patients with SSc who had not received immunosuppressive drugs or steroids for at least 6 months and 48 age- and sex-matched healthy controls (HC) from four European centres. Serum concentrations of sTWEAK and sCD163 were measured using commercially available ELISA kits. RESULTS The mean serum concentrations of sTWEAK were comparable between SSc patients (mean +/- SD: 270 +/- 171 pg/mL) and HC (294 +/- 147pg/mL, P >0.05). Concentration of sCD163 and sCD163/sTWEAK ratio were significantly greater in SSc patients (984 +/- 420 ng/mL and 4837 +/- 3103, respectively) as compared to HC (823 +/- 331 ng/mL and 3115 +/- 1346 respectively, P <0.05 for both). High sCD163 levels and a high sCD163/sTWEAK ratio (defined as > mean +2SD of HC) were both associated with a lower risk of digital ulcers in SSc patients (OR, 95%CI: 0.09; 0.01, 0.71, and 0.17; 0.06, 0.51, respectively). Accordingly, patients without digital ulcers had a significantly higher sCD163 concentration and sCD163/sTWEAK ratio as compared to SSc patients with digital ulcers (P <0.01 for both) and HC (P <0.05 for both). A high sCD163/sTWEAK ratio, but not high sCD163 levels, was associated with greater skin involvement. CONCLUSIONS The results of our study indicate that CD163-TWEAK interactions might play a role in the pathogenesis of SSc and that CD163 may protect against the development of digital ulcers in SSc. Further studies are required to reveal whether targeting of the CD163-TWEAK pathway might be a potential strategy for treating vascular disease and/or skin fibrosis in SSc.
Collapse
|
26
|
Li H, Nagai T, Hasui K, Matsuyama T. Depletion of folate receptor β-expressing macrophages alleviates bleomycin-induced experimental skin fibrosis. Mod Rheumatol 2014; 24:816-22. [PMID: 24498991 DOI: 10.3109/14397595.2013.879415] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
OBJECTIVES Folate receptor β (FRβ)-expressing macrophages have been identified as activated macrophages. Here, we investigated the infiltration of FRβ-expressing macrophages in a murine model of bleomycin (BLM)-induced skin fibrosis and assessed the antifibrotic effects of depletion of FRβ-expressing macrophages in this model using a recombinant immunotoxin to FRβ. METHODS A recombinant immunotoxin (anti-FRβ-PE38) was prepared by conjugating the Fv portion of the anti-mouse FRβ heavy chain with truncated Pseudomonas exotoxin A (VH-PE38) and the Fv portion of the anti-mouse FRβ light chain. BLM-induced skin fibrosis mice were intravenously treated with either anti-FRβ-PE38 or VH-PE38 as a control protein. Skin fibrosis was evaluated by the change of skin thickness and hydroxyproline content on Day 29. The TGFβ1 mRNA levels in the treated skin were assessed by quantitative real-time RT-PCR on Day 9. RESULTS Numbers of FRβ-expressing macrophages increased in BLM-injected skin. Anti-FRβ-PE38 treatment led to a dramatic reduction in the number of FRβ-expressing macrophages. Additionally, skin thickness and hydroxyproline content, were markedly reduced. TGFβ1 mRNA levels were also down-regulated after the treatment. TGFβ1 expression was enriched in FRβ-expressing macrophages compared with FRβ-negative macrophages. CONCLUSION These results indicated that anti-FRβ-PE38 treatment efficiently depleted FRβ-expressing macrophages and consequently alleviated BLM-induced skin fibrosis.
Collapse
Affiliation(s)
- Hua Li
- Department of Immunology, Graduate School of Medical and Dental Sciences, Kagoshima University , Kagoshima , Japan
| | | | | | | |
Collapse
|
27
|
TNF-α promoter single nucleotide polymorphisms and haplotypes associate with susceptibility of immune thrombocytopenia in Chinese adults. Hum Immunol 2014; 75:980-5. [PMID: 25158149 DOI: 10.1016/j.humimm.2014.08.197] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2014] [Revised: 06/12/2014] [Accepted: 08/14/2014] [Indexed: 10/25/2022]
Abstract
OBJECTIVE Tumor necrosis factor-alpha (TNF-α) participates as a candidate susceptibility factor for immune thrombocytopenia (ITP). This study attempted to investigate the association between five single nucleotide polymorphisms (SNPs) spanning the TNF-α promoter and the susceptibility of primary ITP in Chinese Han adults. METHODS In 215 adult primary ITP patients and 206 healthy controls, SNPs were detected by PCR-RFLP and PCR-SSP. The χ(2) test or fisher's exact test was used to compare frequencies of genotypes and alleles between patients and controls. Haplotypes were analyzed with the SHEsis online program. TNF-α, IFN-γ and Galectin-9 mRNA of 35 newly diagnosed adult ITP patients and 35 healthy controls were detected by qRT-PCR. RESULTS The haplotype GGC (-238G/-308G/-857C) of TNF-α promoter was significantly associated with a decreased susceptibility of primary ITP, especially in males. The relative levels of mRNA expression of TNF-α, IFN-γ and Gal-9 in adult active primary ITP patients was significantly up-regulated compared with patients in remission and controls. CONCLUSIONS This study represented the first report that the haplotype GGC of TNF-α was differentially associated with the susceptibility of primary ITP in Chinese Han adults. The up-regulation of TNF-α, IFN-γ and Galectin-9 was significantly correlated with active primary ITP in adult patients.
Collapse
|
28
|
Pope J, Walker KM, de Leon F, Vanderhoek L, Seney S, Summers KL. Correlations between changes in cytokines and clinical outcomes for early phase (proof of concept) trials in active diffuse systemic sclerosis using data from an imatinib study. Rheumatology (Oxford) 2014; 53:1830-4. [PMID: 24850877 DOI: 10.1093/rheumatology/keu216] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
OBJECTIVE Data from a small study testing imatinib to treat SSc were used to determine if cytokine changes were related to differences in clinical parameters to model future early phase trials pairing cytokine changes and clinical parameters. METHODS Plasma and punch skin biopsy specimens collected at baseline and 6 months were analysed for levels of 26 fibrotic and inflammatory cytokines using multiplexed immunoassays and ELISA. Seven of nine patients on active treatment had paired data. Biopsies were biopulverized and standardized to protein levels in the tissue homogenate. Plasma was frozen at -80°C and analysed using multiplexed immunoassays or ELISAs standardized to CRP. Correlations between fold changes in cytokines and differences in clinical parameters (skin score, physician and patient global assessments and HAQ) were performed. P < 0.01 was considered significant. RESULTS After 6 months of imatinib treatment, plasma levels of soluble vascular cell adhesion molecule 1 decreased significantly (P < 0.001), while tissue levels of soluble intercellular adhesion molecule 1 increased (P < 0.01). Some significant correlations between fold changes in certain plasma fibrotic and inflammatory cytokines and changes in clinical parameters after 6 months of treatment were found: patient global scores and IL-13 (r = 0.964, P < 0.0001); ESR and IL-12p70 (r = -0.903, P < 0.01); in tissue samples, patient global score and soluble E-selectin (r = 0.913, P < 0.01); and physician global score with sCD40L (r = -0.883, P < 0.01). CONCLUSION Some serum and tissue cytokines may have a role in early phase clinical trials of SSc, correlating with changes in clinical parameters. Serum and tissue samples could be analysed in early phase trials to determine whether they support the clinical observations. TRIAL REGISTRATION http://clinicaltrials.gov/show/NCT01545427.
Collapse
Affiliation(s)
- Janet Pope
- Division of Rheumatology, Department of Medicine, Schulich School of Medicine & Dentistry, University of Western Ontario, London, ON, Canada, Medical School, Trinity College Dublin, Dublin, Ireland, Department of Mathematics/Statistics, McMaster University, Hamilton, Lawson Health Research Institute and Department of Microbiology & Immunology, Schulich School of Medicine & Dentistry, University of Western Ontario, London, ON, Canada
| | - Kyle M Walker
- Division of Rheumatology, Department of Medicine, Schulich School of Medicine & Dentistry, University of Western Ontario, London, ON, Canada, Medical School, Trinity College Dublin, Dublin, Ireland, Department of Mathematics/Statistics, McMaster University, Hamilton, Lawson Health Research Institute and Department of Microbiology & Immunology, Schulich School of Medicine & Dentistry, University of Western Ontario, London, ON, Canada
| | - Faye de Leon
- Division of Rheumatology, Department of Medicine, Schulich School of Medicine & Dentistry, University of Western Ontario, London, ON, Canada, Medical School, Trinity College Dublin, Dublin, Ireland, Department of Mathematics/Statistics, McMaster University, Hamilton, Lawson Health Research Institute and Department of Microbiology & Immunology, Schulich School of Medicine & Dentistry, University of Western Ontario, London, ON, Canada
| | - Louise Vanderhoek
- Division of Rheumatology, Department of Medicine, Schulich School of Medicine & Dentistry, University of Western Ontario, London, ON, Canada, Medical School, Trinity College Dublin, Dublin, Ireland, Department of Mathematics/Statistics, McMaster University, Hamilton, Lawson Health Research Institute and Department of Microbiology & Immunology, Schulich School of Medicine & Dentistry, University of Western Ontario, London, ON, Canada
| | - Shannon Seney
- Division of Rheumatology, Department of Medicine, Schulich School of Medicine & Dentistry, University of Western Ontario, London, ON, Canada, Medical School, Trinity College Dublin, Dublin, Ireland, Department of Mathematics/Statistics, McMaster University, Hamilton, Lawson Health Research Institute and Department of Microbiology & Immunology, Schulich School of Medicine & Dentistry, University of Western Ontario, London, ON, Canada
| | - Kelly L Summers
- Division of Rheumatology, Department of Medicine, Schulich School of Medicine & Dentistry, University of Western Ontario, London, ON, Canada, Medical School, Trinity College Dublin, Dublin, Ireland, Department of Mathematics/Statistics, McMaster University, Hamilton, Lawson Health Research Institute and Department of Microbiology & Immunology, Schulich School of Medicine & Dentistry, University of Western Ontario, London, ON, Canada Division of Rheumatology, Department of Medicine, Schulich School of Medicine & Dentistry, University of Western Ontario, London, ON, Canada, Medical School, Trinity College Dublin, Dublin, Ireland, Department of Mathematics/Statistics, McMaster University, Hamilton, Lawson Health Research Institute and Department of Microbiology & Immunology, Schulich School of Medicine & Dentistry, University of Western Ontario, London, ON, Canada
| |
Collapse
|
29
|
Ohira H, Watanabe H. Pathophysiology and recent findings of primary biliary cirrhosis complicated by systemic sclerosis. Hepatol Res 2014; 44:377-83. [PMID: 24308674 DOI: 10.1111/hepr.12285] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2013] [Revised: 12/02/2013] [Accepted: 12/02/2013] [Indexed: 02/08/2023]
Abstract
Primary biliary cirrhosis (PBC) can be complicated by systemic sclerosis (SSc) and, more specifically, limited cutaneous SSc (lcSSc), which was previously called CREST syndrome. Moreover, combined PBC and SSc has been described in many case reports. Although neither the etiology of PBC nor that of SSc has been elucidated, some genetic and immunological factors are known to be shared. Both disorders are autoimmune fibrotic diseases characterized by increased levels of profibrotic cytokines transforming growth factor β (TGFβ) and interleukin-6, which have recently been suggested to influence T-helper 17 cells and regulatory T cells involved in acquired immunity. lcSSc is accompanied by CREST symptoms, although complete CREST cases are rare, with relatively high prevalence of Raynaud's phenomenon, sclerodactyly and telangiectasia, and lower prevalence of calcinosis and esophageal dysmotility. Because patients with anticentromere antibody positive PBC-SSc are at a high risk of developing portal hypertension, particular attention should be paid to the management of gastroesophageal varices. In addition, the management of SSc-related non-hepatic disorders, such as pulmonary fibrosis, pulmonary hypertension, heart disorder, infection and malignancy, is also important for improved outcomes. Because PBC is often complicated by rheumatic disease, hepatologists should keep the possibility of systemic disorder in mind when examining PBC patients.
Collapse
Affiliation(s)
- Hiromasa Ohira
- Department of Gastroenterology and Rheumatology, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Hiroshi Watanabe
- Department of Gastroenterology and Rheumatology, Fukushima Medical University School of Medicine, Fukushima, Japan
| |
Collapse
|
30
|
van Bon L, Affandi AJ, Broen J, Christmann RB, Marijnissen RJ, Stawski L, Farina GA, Stifano G, Mathes AL, Cossu M, York M, Collins C, Wenink M, Huijbens R, Hesselstrand R, Saxne T, DiMarzio M, Wuttge D, Agarwal SK, Reveille JD, Assassi S, Mayes M, Deng Y, Drenth JPH, de Graaf J, den Heijer M, Kallenberg CGM, Bijl M, Loof A, van den Berg WB, Joosten LAB, Smith V, de Keyser F, Scorza R, Lunardi C, van Riel PLCM, Vonk M, van Heerde W, Meller S, Homey B, Beretta L, Roest M, Trojanowska M, Lafyatis R, Radstake TRDJ. Proteome-wide analysis and CXCL4 as a biomarker in systemic sclerosis. N Engl J Med 2014; 370:433-43. [PMID: 24350901 PMCID: PMC4040466 DOI: 10.1056/nejmoa1114576] [Citation(s) in RCA: 339] [Impact Index Per Article: 30.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
BACKGROUND Plasmacytoid dendritic cells have been implicated in the pathogenesis of systemic sclerosis through mechanisms beyond the previously suggested production of type I interferon. METHODS We isolated plasmacytoid dendritic cells from healthy persons and from patients with systemic sclerosis who had distinct clinical phenotypes. We then performed proteome-wide analysis and validated these observations in five large cohorts of patients with systemic sclerosis. Next, we compared the results with those in patients with systemic lupus erythematosus, ankylosing spondylitis, and hepatic fibrosis. We correlated plasma levels of CXCL4 protein with features of systemic sclerosis and studied the direct effects of CXCL4 in vitro and in vivo. RESULTS Proteome-wide analysis and validation showed that CXCL4 is the predominant protein secreted by plasmacytoid dendritic cells in systemic sclerosis, both in circulation and in skin. The mean (±SD) level of CXCL4 in patients with systemic sclerosis was 25,624±2652 pg per milliliter, which was significantly higher than the level in controls (92.5±77.9 pg per milliliter) and than the level in patients with systemic lupus erythematosus (1346±1011 pg per milliliter), ankylosing spondylitis (1368±1162 pg per milliliter), or liver fibrosis (1668±1263 pg per milliliter). CXCL4 levels correlated with skin and lung fibrosis and with pulmonary arterial hypertension. Among chemokines, only CXCL4 predicted the risk and progression of systemic sclerosis. In vitro, CXCL4 down-regulated expression of transcription factor FLI1, induced markers of endothelial-cell activation, and potentiated responses of toll-like receptors. In vivo, CXCL4 induced the influx of inflammatory cells and skin transcriptome changes, as in systemic sclerosis. CONCLUSIONS Levels of CXCL4 were elevated in patients with systemic sclerosis and correlated with the presence and progression of complications, such as lung fibrosis and pulmonary arterial hypertension. (Funded by the Dutch Arthritis Association and others.).
Collapse
Affiliation(s)
- Lenny van Bon
- The authors' affiliations are listed in the Appendix
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Aydoğdu E, Pamuk ÖN, Dönmez S, Pamuk GE. Decreased interleukin-20 level in patients with systemic sclerosis: are they related with angiogenesis? Clin Rheumatol 2013; 32:1599-603. [PMID: 23812620 DOI: 10.1007/s10067-013-2317-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2012] [Revised: 02/17/2013] [Accepted: 06/09/2013] [Indexed: 11/28/2022]
Abstract
In this study, we aimed to evaluate the relation between angiogenesis indicators and T helper 17 cytokine group in patients with systemic sclerosis (SSc) which is a disease characterized by impaired angiogenesis and autoimmune response. In our study, patients with SSc are compared with patients with primary Raynaud's phenomenon (RP) and healthy controls. Forty SSc patients, 18 primary RP cases, and 20 healthy controls were included in our study. The demographic and clinical features of patients with SSc were recorded. The serum levels of vascular endothelial growth factor (VEGF), vascular endothelial (VE)-cadherin, interleukin (IL)-20, IL-22, and IL-23 were assessed. In the SSc group, IL-20 level was significantly lower than in both primary RP group and controls (p values <0.001). VE-cadherin level in SSc was significantly higher than in primary RP (p = 0.016). The IL-22 and IL-23 and VEGF levels of SSc, primary RP, and control groups were similar (p values >0.05). In SSc patients, IL-23 correlated negatively with VEGF (r = -0.36, p = 0.025) and positively with VE-cadherin (r = 0.55, p < 0.001). IL-20 levels in SSc patients correlated with disease duration (r = 0.32, p = 0.044). SSc patients with limited involvement had significantly higher VE-cadherin levels than SSc patients with diffuse involvement (p = 0.044). We observed that IL-20 which is an IL-10 group angiogenesis indicator was observed to be suppressed in SSc, suggesting abnormal angiogenesis.
Collapse
Affiliation(s)
- Erkan Aydoğdu
- Department of Rheumatology, Trakya University Medical Faculty, Edirne, Turkey
| | | | | | | |
Collapse
|
32
|
Marut W, Jamier V, Kavian N, Servettaz A, Winyard PG, Eggleton P, Anwar A, Nicco C, Jacob C, Chéreau C, Weill B, Batteux F. The natural organosulfur compound dipropyltetrasulfide prevents HOCl-induced systemic sclerosis in the mouse. Arthritis Res Ther 2013; 15:R167. [PMID: 24286210 PMCID: PMC3979139 DOI: 10.1186/ar4351] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2013] [Accepted: 09/26/2013] [Indexed: 11/24/2022] Open
Abstract
Introduction The aim of this study was to test the naturally occurring organosulfur compound dipropyltetrasulfide (DPTTS), found in plants, which has antibiotic and anticancer properties, as a treatment for HOCl-induced systemic sclerosis in the mouse. Methods The prooxidative, antiproliferative, and cytotoxic effects of DPTTS were evaluated ex vivo on fibroblasts from normal and HOCl mice. In vivo, the antifibrotic and immunomodulating properties of DPTTS were evaluated in the skin and lungs of HOCl mice. Results H2O2 production was higher in fibroblasts derived from HOCl mice than in normal fibroblasts (P < 0.05). DPTTS did not increase H2O2 production in normal fibroblasts, but DPTTS dose-dependently increased H2O2 production in HOCl fibroblasts (P < 0.001 with 40 μM DPTTS). Because H2O2 reached a lethal threshold in cells from HOCl mice, the antiproliferative, cytotoxic, and proapoptotic effects of DPTTS were significantly higher in HOCl fibroblasts than for normal fibroblasts. In vivo, DPTTS decreased dermal thickness (P < 0.001), collagen content in skin (P < 0.01) and lungs (P < 0.05), αSMA (P < 0.01) and pSMAD2/3 (P < 0.01) expression in skin, formation of advanced oxidation protein products and anti-DNA topoisomerase-1 antibodies in serum (P < 0.05) versus untreated HOCl mice. Moreover, in HOCl mice, DPTTS reduced splenic B-cell counts (P < 0.01), the proliferative rates of B-splenocytes stimulated by lipopolysaccharide (P < 0.05), and T-splenocytes stimulated by anti-CD3/CD28 mAb (P < 0.001). Ex vivo, it also reduced the production of IL-4 and IL-13 by activated T cells (P < 0.05 in both cases). Conclusions The natural organosulfur compound DPTTS prevents skin and lung fibrosis in the mouse through the selective killing of diseased fibroblasts and its immunomodulating properties. DPTTS may be a potential treatment for systemic sclerosis.
Collapse
|
33
|
Abstract
Accumulative evidence demonstrates the crucial role of evolutionary conserved Toll-like receptors (TLRs) in identifying microbial or viral compounds. TLRs are also able to recognise endogenous molecules which are released upon cell damage or stress and have been shown to play a key role in numerous autoimmune diseases including systemic sclerosis (SSc). A classic feature of SSc, is vascular injury manifested as Raynaud's phenomenon and ischaemia of the skin, resulting in the release of endogenous TLR ligands during inflammation and local tissue damage. These locally released TLR ligands bind TLRs possibly complexed to autoantibodies, and initiate intracellular signalling pathways and may be one of the mechanisms that initiate and drive autoimmunity and subsequent fibrosis. Activation of the immune system results in interferon (IFN) sensitive gene transcription. There is also an IFN gene signature in SSc peripheral blood. TLRs may represent the link between immune activation, common in SSc, and tissue fibrosis. Therefore, a better understanding of the mechanisms of TLR-mediated pathogenesis and therapies targeting individual TLRs, may provide a more specific approach of treating multi-systemic autoimmune diseases. This review aims to integrate the current knowledge of TLR function in the autoimmune disorders with particular emphasis on SSc. We suggest the TLR system as a new therapeutic target.
Collapse
|
34
|
Viswanath V, Phiske MM, Gopalani VV. Systemic sclerosis: current concepts in pathogenesis and therapeutic aspects of dermatological manifestations. Indian J Dermatol 2013; 58:255-68. [PMID: 23918994 PMCID: PMC3726870 DOI: 10.4103/0019-5154.113930] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Systemic sclerosis (SSc) is a chronic, multisystem connective tissue disease with protean clinical manifestations. Recent advances in understanding the pathogenic mechanisms have led to development of target-oriented and vasomodulatory drugs which play a pivotal role in treating various dermatological manifestations. An exhaustive literature search was done using Medline, Embase, and Cochrane library to review the recent concepts regarding pathogenesis and evidence-based treatment of salient dermatological manifestations. The concept of shared genetic risk factors for the development of autoimmune diseases is seen in SSc. It is divided into fibroproliferative and inflammatory groups based on genome-wide molecular profiling. Genetic, infectious, and environmental factors play a key role; vascular injury, fibrosis, and immune activation are the chief pathogenic factors. Vitamin D deficiency has been documented in SSc and correlates with the severity of skin involvement. Skin sclerosis, Raynaud's phenomenon (RP) with digital vasculopathies, pigmentation, calcinosis, and leg ulcers affect the patient's quality of life. Immunosuppressives, biologicals, and hematopoietic stem cell transplantation are efficacious in skin sclerosis. Endothelin A receptor antagonists, calcium-channel blockers, angiotensin receptor inhibitors, prostacyclin analogs, and phosphodiesterase type 5 (PDE-5) inhibitors are the mainstay in RP and digital vasculopathies. Pigmentation in SSc has been attributed to melanogenic potential of endothelin-1 (ET-1); the role of ET 1 antagonists and vitamin D analogs needs to be investigated. Sexual dysfunction in both male and female patients has been attributed to vasculopathy and fibrosis, wherein PDE-5 inhibitors are found to be useful. The future concepts of treating SSc may be based on the gene expression signature.
Collapse
Affiliation(s)
- Vishalakshi Viswanath
- Department of Dermatology, Rajiv Gandhi Medical College and CSM Hospital, Kalwa, Thane Municipal Corporation, India
| | | | | |
Collapse
|
35
|
Abstract
The connective tissue disorders comprise a number of related conditions that include systemic lupus erythematosus (SLE) and the antiphospholipid (Hughes) syndrome, scleroderma, myositis and Sjögren's syndrome. They are characterized by autoantibody production and other immune-mediated dysfunction. There are common clinical and serological features with some patients having multiple overlapping connective tissue disorders. The latest advances include new approaches to therapy, including more focused utilization of existing therapies and the introduction of biological therapies in SLE, more precise protocols for assessment of severe disease manifestations such as in interstitial lung disease and pulmonary artery hypertension in scleroderma, new antibodies for disease characterization in myositis and new approaches to patient assessment in Sjögren's syndrome. B cells have a critical role in most, if not all of these disorders such that B-cell depletion or suppression of B-cell activating cytokines improves disease in many patients. In particular, the introduction of rituximab, a monoclonal antibody targeting the CD20 molecule on B cells, into clinical practice for rheumatoid arthritis and B-cell lymphoma has been a key driver of experimental approaches to therapy in connective tissue disorders. Genetic studies also suggest a role for the innate immune system in disease pathogenesis, suggesting further future targets for biological therapies over the next few years.
Collapse
Affiliation(s)
- Vijay Rao
- Rheumatology Department, Queen Elizabeth Hospital Birmingham, Birmingham, UK
| | | |
Collapse
|
36
|
Chrobak I, Lenna S, Stawski L, Trojanowska M. Interferon-γ promotes vascular remodeling in human microvascular endothelial cells by upregulating endothelin (ET)-1 and transforming growth factor (TGF) β2. J Cell Physiol 2013; 228:1774-83. [PMID: 23359533 DOI: 10.1002/jcp.24337] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2012] [Accepted: 01/18/2013] [Indexed: 12/17/2022]
Abstract
Systemic sclerosis (SSc) is a complex disease characterized by vascular alterations, activation of the immune system and tissue fibrosis. Previous studies have implicated activation of the interferon pathways in the pathogenesis of SSc. The goal of this study was to determine whether interferon type I and/or type II could play a pathogenic role in SSc vasculopathy. Human dermal microvascular endothelial cells (HDMVECs) and fibroblasts were obtained from foreskins of healthy newborns. The RT Profiler PCR Array System was utilized to screen for EndoMT genes. Treatment with IFN-α or IFN-γ downregulated Fli1 and VE-cadherin. In contrast, IFN-α and IFN-γ exerted opposite effects on the expression of α-SMA, CTGF, ET-1, and TGFβ2, with IFN-α downregulating and IFN-γ upregulating this set of genes. Blockade of TGFβ signaling normalized IFN-γ-mediated changes in Fli1, VE-cadherin, CTGF, and ET-1 levels, whereas upregulation of α-SMA and TGFβ2 was not affected. Bosentan treatment was more effective than TGFβ blockade in reversing the actions of IFN-γ, including downregulation of α-SMA and TGFβ2, suggesting that activation of the ET-1 pathway plays a main role in the IFN-γ responses in HDMECs. IFN-γ induced expression of selected genes related to endothelial-to-mesenchymal transition (EndoMT), including Snail1, FN1, PAI1, TWIST1, STAT3, RGS2, and components of the WNT pathway. The effect of IFN-γ on EndoMT was mediated via TGFβ2 and ET-1 signaling pathways. This study demonstrates distinct effects of IFN-α and IFN-γ on the biology of vascular endothelial cells. IFN-γ may contribute to abnormal vascular remodeling and fibrogenesis in SSc, partially via induction of EndoMT.
Collapse
Affiliation(s)
- Izabela Chrobak
- Boston University School of Medicine, Arthritis Center, Boston, Massachusetts 02118, USA
| | | | | | | |
Collapse
|
37
|
Potential role of human-specific genes, human-specific microRNAs and human-specific non-coding regulatory RNAs in the pathogenesis of systemic sclerosis and Sjögren's syndrome. Autoimmun Rev 2013; 12:1046-51. [PMID: 23684698 DOI: 10.1016/j.autrev.2013.04.004] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2013] [Accepted: 04/24/2013] [Indexed: 12/20/2022]
Abstract
The etiology and pathogenesis of human autoimmune diseases remain unknown despite intensive investigations. Although remarkable progress has been accomplished through genome wide association studies in the identification of genetic factors that may predispose to their occurrence or modify their clinical presentation to date no specific gene abnormalities have been conclusively demonstrated to be responsible for these diseases. The completion of the human and chimpanzee genome sequencing has opened up novel opportunities to examine the possible contribution of human specific genes and other regulatory elements unique to the human genome, such as microRNAs and non-coding RNAs, towards the pathogenesis of a variety of human disorders. Thus, it is likely that these human specific genes and non-coding regulatory elements may be involved in the development or the pathogenesis of various disorders that do not occur in non-human primates including certain autoimmune diseases such as Systemic Sclerosis and Primary Sjögren's Syndrome. Here, we discuss recent evidence supporting the notion that human specific genes or human specific microRNA and other non-coding RNA regulatory elements unique to the human genome may participate in the development or in the pathogenesis of Systemic Sclerosis and Primary Sjögren's Syndrome.
Collapse
|
38
|
Li J, Wang X, Zhang F, Yin H. Toll-like receptors as therapeutic targets for autoimmune connective tissue diseases. Pharmacol Ther 2013; 138:441-51. [PMID: 23531543 DOI: 10.1016/j.pharmthera.2013.03.003] [Citation(s) in RCA: 89] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2013] [Accepted: 02/19/2013] [Indexed: 02/06/2023]
Abstract
Autoimmune connective tissue diseases (ACTDs) are a family of consistent systemic autoimmune inflammatory disorders, including systemic lupus erythematosus (SLE), rheumatoid arthritis (RA), systemic sclerosis (SSc) and Sjögren's syndrome (SS). IL-1R-like receptors (TLRs) are located on various cellular membranes and sense exogenous and endogenous danger-associated molecular patterns (DAMPs) and pathogen-associated molecular patterns (PAMPs), playing a critical role in innate immune responses. During the past decade, the investigation of TLRs in inflammatory autoimmune diseases has been fruitful. In this report, we review the significant biochemical, physiological and pathological studies of the key functions of TLRs in ACTDs. Several proteins in the TLR signaling pathways (e.g., IKK-2 and MyD88) have been identified as potential therapeutic targets for the treatment of ACTDs. Antibodies, oligodeoxyribonucleotides (ODNs) and small molecular inhibitors (SMIs) have been tested to modulate TLR signaling. Some drug-like SMIs of TLR signaling, such as RDP58, ST2825, ML120B and PHA-408, have demonstrated remarkable potential, with promising safety and efficacy profiles, which should warrant further clinical investigation. Nonetheless, one should bear in mind that all TLRs exert both protective and pathogenic functions; the function of TLR4 in inflammatory bowel disease represents such an example. Therefore, an important aspect of TLR modulator development involves the identification of a balance between the suppression of disease-inducing inflammation, while retaining the beneficiary host immune response.
Collapse
Affiliation(s)
- Jing Li
- Department of Chemistry and Biochemistry and Biofrontiers Institute, University of Colorado at Boulder, Boulder, CO 80309-0596, USA
| | | | | | | |
Collapse
|
39
|
Peripheral blood mononuclear cells from patients with systemic sclerosis spontaneously secrete increased amounts of vascular endothelial growth factor (VEGF) already in the early stage of the disease. Adv Med Sci 2012; 56:255-63. [PMID: 21983449 DOI: 10.2478/v10039-011-0025-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
PURPOSE To investigate the capacity of the peripheral blood mononuclear cells (PBMC) from patients with systemic sclerosis (SSc) to produce vascular endothelial growth factor (VEGF), and to identify clinical associations of altered production of VEGF by PBMC in SSc. In addition, correlation with another pro-angiogenic cytokine, TNF-related weak inducer of apoptosis (TWEAK), was evaluated. METHODS PBMC were isolated from 25 patients with SSc and 17 healthy controls (HC). VEGF and TWEAK were measured in the supernatants of cultured PBMC using commercially available ELISA kits. RESULTS PBMC from SSc patients spontaneously released significantly greater amounts of VEGF as compared with HC. Production of VEGF was comparable between patients with early SSc and those with longer disease duration, and in both SSc groups higher than in HC. Patients without active digital ulcers produced significantly greater amounts of VEGF as compared with HC, while there was no significant difference in the production of VEGF between SSc patients with active digital ulcers and HC. VEGF/TWEAK ratio was significantly higher in PBMC from SSc patients than in HC indicating that high production of VEGF is not paralleled by increased release of TWEAK in SSc. CONCLUSIONS PBMC form SSc patients produce increased amounts of VEGF already in the early stage of disease. There is an imbalance in the profile of pro-angiogenic mediators produced by PBMC in SSc which might contribute to the pathogenesis of SSc. Further studies should address clinical significance of our findings.
Collapse
|
40
|
Ramirez GA, Franchini S, Rovere-Querini P, Sabbadini MG, Manfredi AA, Maugeri N. The role of platelets in the pathogenesis of systemic sclerosis. Front Immunol 2012; 3:160. [PMID: 22719739 PMCID: PMC3376452 DOI: 10.3389/fimmu.2012.00160] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2012] [Accepted: 05/29/2012] [Indexed: 12/15/2022] Open
Abstract
Systemic sclerosis (SSc) is an inflammatory disease of unknown etiology characterized by widespread organ dysfunction due to fibrosis and ischemia. Its nebulous pathogenic background and the consequent absence of an etiological therapy prevent the adoption of satisfying treatment strategies, able to improve patients’ quality of life and survival and stimulate researchers to identify a unifying pathogenic target. Platelets show a unique biological behavior, lying at the crossroads between vascular function, innate and adaptive immunity, and regulation of cell proliferation. Consequently they are also emerging players in the pathogenesis of many inflammatory diseases, including SSc. In the setting of SSc platelets are detectable in a persistent activated state, which is intimately linked to the concomitant presence of an injured endothelium and to the widespread activation of the innate and adaptive immune system. As a consistent circulating source of bioactive compounds platelets contribute to the development of many characteristic phenomena of SSc, such as fibrosis and impaired vascular tone.
Collapse
Affiliation(s)
- Giuseppe A Ramirez
- Department of Immunology, San Raffaele Scientific Institute, Università Vita-Salute San Raffaele, Milano, Italy
| | | | | | | | | | | |
Collapse
|
41
|
Wermuth PJ, Jimenez SA. Gadolinium compounds signaling through TLR4 and TLR7 in normal human macrophages: establishment of a proinflammatory phenotype and implications for the pathogenesis of nephrogenic systemic fibrosis. THE JOURNAL OF IMMUNOLOGY 2012; 189:318-27. [PMID: 22649203 DOI: 10.4049/jimmunol.1103099] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Nephrogenic systemic sibrosis is a progressive disorder occurring in some renal insufficiency patients exposed to gadolinium-based contrast agents (GdBCA). Previous studies demonstrated that the GdBCA Omniscan upregulated several innate immunity pathways in normal differentiated human macrophages, induced rapid nuclear localization of the transcription factor NF-κB, and increased the expression and production of numerous profibrotic/proinflammatory cytokines, chemokines, and growth factors. To further examine GdBCA stimulation of the innate immune system, cultured human embryonic kidney 293 cells expressing one of seven different human TLRs or one of two human nucleotide-binding oligomerization domain-like receptors were exposed in vitro for 24 h to various GdBCA. The signaling activity of each compound was evaluated by its ability to activate an NF-κB-inducible reporter gene. Omniscan and gadodiamide induced strong TLR4- and TLR7-mediated reporter gene activation. The other Gd compounds examined failed to induce reporter gene activation. TLR pathway inhibition using chloroquine or an inhibitor of IL-1R-associated kinases 1 and 4 in normal differentiated human macrophages abrogated Omniscan-induced gene expression. Omniscan and gadodiamide signaling via TLRs 4 and 7 resulted in increased production and expression of numerous proinflammatory/profibrotic cytokines, chemokines, and growth factors, including CXCL10, CCL2, CCL8, CXCL12, IL-4, IL-6, TGF-β, and vascular endothelial growth factor. These observations suggest that TLR activation by environmental stimuli may participate in the pathogenesis of nephrogenic systemic fibrosis and of other fibrotic disorders including systemic sclerosis.
Collapse
Affiliation(s)
- Peter J Wermuth
- Jefferson Institute of Molecular Medicine, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | | |
Collapse
|
42
|
Abstract
Dendritic cells are professional antigen-presenting cells that are most studied for their function in mediating T-cell tolerance and T-cell activation. In addition, recent evidence indicates that dendritic cells can regulate the vasculature and function of fibroblast-type cells. The potential contribution of dendritic cells to scleroderma and fibrosis is not well-understood. In this article, we review recent studies as well as describe our own ongoing work that points toward a role for dendritic cells in scleroderma and fibrosis.
Collapse
|
43
|
Emerging roles for TNIP1 in regulating post-receptor signaling. Cytokine Growth Factor Rev 2012; 23:109-18. [PMID: 22542476 DOI: 10.1016/j.cytogfr.2012.04.002] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2011] [Revised: 03/29/2012] [Accepted: 04/06/2012] [Indexed: 02/08/2023]
Abstract
A vast number of cellular processes and signaling pathways are regulated by various receptors, ranging from transmembrane to nuclear receptors. These receptor-mediated processes are modulated by a diverse set of regulatory proteins. TNFα-induced protein 3-interacting protein 1 is such a protein that inhibits both transduction by transmembrane receptors, such as TNFα-receptor, EGF-R, and TLR, and nuclear receptors' PPAR and RAR activity. These receptors play key roles in regulating inflammation and inflammatory diseases. A growing number of references have implicated TNIP1 through GWAS and expression studies in chronic inflammatory diseases such as psoriasis and rheumatoid arthritis, although TNIP1s exact role has yet been determined. In this review, we aim to integrate the current knowledge of TNIP1s functions with the diseases in which it has been associated to potentially elucidate the role this regulator has in promoting or alleviating these inflammatory diseases.
Collapse
|
44
|
Abstract
PURPOSE OF REVIEW This review aims to provide an overview of the recent data that emerged, further substantiating the critical role of innate immunity in systemic sclerosis (SSc). RECENT FINDINGS Driven by the evidence that newly identified SSc susceptibility genes are predominantly involved in immune regulation, we discuss the aberrant antigen presenting cell (APC) activation observed in the course of disease. In particular, we report the alternate activation of 'M1' and 'M2' macrophages reflecting different clinical phenotypes and the aberrant Toll-like receptor (TLR) response, whose effect on cytokine production is mostly evident in the early phases of disease; we especially highlight the increasing importance attributed to TLR3-mediated fibrosis. We next discuss the potential role for interferon (IFN) - producing plasmacytoid dendritic cells (pDCs) in triggering or perpetuating the inflammatory loop caused by TLR hyperactivation, possibly resulting in inflammasome-derived IL-1β-mediated fibrosis and IL-17 producing T helper cells (Th17) skewing. SUMMARY We propose to approach SSc as a multistep immune-mediated disease that is in need of a therapeutic strategy designed to interfere with one or more of these aberrant molecular pathways. Targeting of DCs could be such a target by which dampening the immune system could modify the course of SSc.
Collapse
|
45
|
Bhattacharyya S, Wei J, Varga J. Understanding fibrosis in systemic sclerosis: shifting paradigms, emerging opportunities. Nat Rev Rheumatol 2011; 8:42-54. [PMID: 22025123 PMCID: PMC3954787 DOI: 10.1038/nrrheum.2011.149] [Citation(s) in RCA: 289] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Fibrosis in multiple organs is a prominent pathological finding and distinguishing hallmark of systemic sclerosis (SSc). Findings during the past 5 years have contributed to a more complete understanding of the complex cellular and molecular underpinning of fibrosis in SSc. Fibroblasts, the principal effector cells, are activated in the profibrotic cellular milieu by cytokines and growth factors, developmental pathways, endothelin 1 and thrombin. Innate immune signaling via Toll-like receptors, matrix-generated biomechanical stress signaling via integrins, hypoxia and oxidative stress seem to be implicated in perpetuating the process. Beyond chronic fibroblast activation, fibrosis represents a failure to terminate tissue repair, coupled with an expanded population of mesenchymal cells originating from bone marrow and transdifferentiation of epithelial cells, endothelial cells and pericytes. In addition, studies have identified intrinsic alterations in SSc fibroblasts resulting from epigenetic changes, as well as altered microRNA expression that might underlie the cell-autonomous, persistent activation phenotype of these cells. Precise characterization of the deregulated extracellular and intracellular signaling pathways, mediators and cellular differentiation programs that contribute to fibrosis in SSc will facilitate the development of selective, targeted therapeutic strategies. Effective antifibrotic therapy will ultimately involve novel compounds and repurposing of drugs that are already approved for other indications.
Collapse
Affiliation(s)
- Swati Bhattacharyya
- Division of Rheumatology, Department of Medicine, Feinberg School of Medicine, Northwestern University, McGaw M300, 240 East Huron Street, Chicago, IL 60611, USA
| | | | | |
Collapse
|
46
|
Bongi SM, Del Rosso A, Passalacqua M, Miccio S, Cerinic MM. Manual lymph drainage improving upper extremity edema and hand function in patients with systemic sclerosis in edematous phase. Arthritis Care Res (Hoboken) 2011; 63:1134-41. [DOI: 10.1002/acr.20487] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
|
47
|
Pattanaik D, Brown M, Postlethwaite AE. Vascular involvement in systemic sclerosis (scleroderma). J Inflamm Res 2011; 4:105-25. [PMID: 22096374 PMCID: PMC3218751 DOI: 10.2147/jir.s18145] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Systemic sclerosis (SSc) is an acquired multiorgan connective tissue disease with variable mortality and morbidity dictated by clinical subset type. The etiology of the basic disease and pathogenesis of the systemic autoimmunity, fibrosis, and fibroproliferative vasculopathy are unknown and debated. In this review, the spectrum of vascular abnormalities and the options currently available to treat the vascular manifestations of SSc are discussed. Also discussed is how the hallmark pathologies (ie, how autoimmunity, vasculopathy, and fibrosis of the disease) might be effected and interconnected with modulatory input from lysophospholipids, sphingosine 1-phosphate, and lysophosphatidic acid.
Collapse
Affiliation(s)
- Debendra Pattanaik
- Division of Connective Tissue Diseases, Department of Medicine, University of Tennessee Health Science Center, Memphis, TN, USA
| | | | | |
Collapse
|
48
|
Allanore Y, Saad M, Dieudé P, Avouac J, Distler JHW, Amouyel P, Matucci-Cerinic M, Riemekasten G, Airo P, Melchers I, Hachulla E, Cusi D, Wichmann HE, Wipff J, Lambert JC, Hunzelmann N, Tiev K, Caramaschi P, Diot E, Kowal-Bielecka O, Valentini G, Mouthon L, Czirják L, Damjanov N, Salvi E, Conti C, Müller M, Müller-Ladner U, Riccieri V, Ruiz B, Cracowski JL, Letenneur L, Dupuy AM, Meyer O, Kahan A, Munnich A, Boileau C, Martinez M. Genome-wide scan identifies TNIP1, PSORS1C1, and RHOB as novel risk loci for systemic sclerosis. PLoS Genet 2011; 7:e1002091. [PMID: 21750679 PMCID: PMC3131285 DOI: 10.1371/journal.pgen.1002091] [Citation(s) in RCA: 179] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2010] [Accepted: 04/05/2011] [Indexed: 12/18/2022] Open
Abstract
Systemic sclerosis (SSc) is an orphan, complex, inflammatory disease affecting the immune system and connective tissue. SSc stands out as a severely incapacitating and life-threatening inflammatory rheumatic disease, with a largely unknown pathogenesis. We have designed a two-stage genome-wide association study of SSc using case-control samples from France, Italy, Germany, and Northern Europe. The initial genome-wide scan was conducted in a French post quality-control sample of 564 cases and 1,776 controls, using almost 500 K SNPs. Two SNPs from the MHC region, together with the 6 loci outside MHC having at least one SNP with a P<10(-5) were selected for follow-up analysis. These markers were genotyped in a post-QC replication sample of 1,682 SSc cases and 3,926 controls. The three top SNPs are in strong linkage disequilibrium and located on 6p21, in the HLA-DQB1 gene: rs9275224, P = 9.18×10(-8), OR = 0.69, 95% CI [0.60-0.79]; rs6457617, P = 1.14×10(-7) and rs9275245, P = 1.39×10(-7). Within the MHC region, the next most associated SNP (rs3130573, P = 1.86×10(-5), OR = 1.36 [1.18-1.56]) is located in the PSORS1C1 gene. Outside the MHC region, our GWAS analysis revealed 7 top SNPs (P<10(-5)) that spanned 6 independent genomic regions. Follow-up of the 17 top SNPs in an independent sample of 1,682 SSc and 3,926 controls showed associations at PSORS1C1 (overall P = 5.70×10(-10), OR:1.25), TNIP1 (P = 4.68×10(-9), OR:1.31), and RHOB loci (P = 3.17×10(-6), OR:1.21). Because of its biological relevance, and previous reports of genetic association at this locus with connective tissue disorders, we investigated TNIP1 expression. A markedly reduced expression of the TNIP1 gene and also its protein product were observed both in lesional skin tissue and in cultured dermal fibroblasts from SSc patients. Furthermore, TNIP1 showed in vitro inhibitory effects on inflammatory cytokine-induced collagen production. The genetic signal of association with TNIP1 variants, together with tissular and cellular investigations, suggests that this pathway has a critical role in regulating autoimmunity and SSc pathogenesis.
Collapse
Affiliation(s)
- Yannick Allanore
- Université Paris Descartes, Rhumatologie A, INSERM, U1016, Hôpital Cochin, APHP, Paris, France.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Santegoets KCM, van Bon L, van den Berg WB, Wenink MH, Radstake TRDJ. Toll-like receptors in rheumatic diseases: are we paying a high price for our defense against bugs? FEBS Lett 2011; 585:3660-6. [PMID: 21513712 DOI: 10.1016/j.febslet.2011.04.028] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2011] [Revised: 04/11/2011] [Accepted: 04/12/2011] [Indexed: 12/31/2022]
Abstract
In the last decade Toll-like receptor (TLR) research has led to new insights in the pathogenesis of many rheumatic diseases. In autoimmune diseases like systemic lupus erythematosus, rheumatoid arthritis and systemic sclerosis TLR signaling is likely to be involved in tolerance breakthrough and chronic inflammation via combined Fc gamma receptors and TLR recognition of immune complexes. Furthermore, inflammatory diseases like psoriatic arthritis and gout also show more and more evidence for TLR involvement. In this review we will discuss the involvement of TLR signaling in several rheumatic diseases and stress their similarities and differences based on recent findings.
Collapse
Affiliation(s)
- K C M Santegoets
- Department of Rheumatology, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands
| | | | | | | | | |
Collapse
|
50
|
Atamas SP, Luzina IG, Ingels J, Choi J, Wong WK, Furst DE, Clements PJ, Postlethwaite AE. Stimulation with type I collagen induces changes in gene expression in peripheral blood mononuclear cells from patients with diffuse cutaneous systemic sclerosis (scleroderma). Clin Exp Immunol 2011; 161:426-35. [PMID: 20529088 DOI: 10.1111/j.1365-2249.2010.04189.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
An autoantigenic role for collagen type I (CI) has been suggested previously in diffuse cutaneous systemic sclerosis (dcSSc). Whether CI is indeed capable of affecting the immune system in dcSSc is not known. Patients with early (3 years or less) or late (>3 years) dcSSc and healthy controls donated blood. Peripheral blood mononuclear cells (PBMC) were cultured with or without CI, and expression of genes known for their involvement in autoimmune and inflammatory processes was assessed using cDNA arrays; results were confirmed by real-time polymerase chain reaction and enzyme-linked immunosorbent assay for selected genes. Patients with early and late dcSSc were similarly different from healthy controls in basal gene expression. When cultured with CI, PBMC from patients with early dcSSc differed from healthy controls in expression of 34 genes, whereas PBMC from patients with late dcSSc differed from healthy controls in expression of only 29 genes. Direct comparisons of matched PBMC samples cultured with and without CI revealed differences in expression of eight genes in healthy controls, of five genes in patients with early dcSSc, and no differences in patients with late dcSSc. Thus, PBMC from patients with dcSSc respond differently than do PBMC from healthy controls when cultured with CI. Exposure to CI in culture of PBMC from patients in the early stage of dcSSc in contrast to PBMC from patients with late-stage dcSSc evokes a greater degree of activation of immune-related genes, suggesting that CI is more dominant as an autoantigen in early versus late dcSSc.
Collapse
Affiliation(s)
- S P Atamas
- The University of Maryland School of Medicine and Baltimore VA Medical Center, Baltimore, MD 21201, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|