1
|
Lee K, Gharaie S, Kurzhagen JT, Newman-Rivera AM, Arend LJ, Noel S, Rabb H. Double-negative T cells have a reparative role after experimental severe ischemic acute kidney injury. Am J Physiol Renal Physiol 2024; 326:F942-F956. [PMID: 38634135 PMCID: PMC11386976 DOI: 10.1152/ajprenal.00376.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 03/25/2024] [Accepted: 04/15/2024] [Indexed: 04/19/2024] Open
Abstract
T cells mediate organ injury and repair. A proportion of unconventional kidney T cells called double-negative (DN) T cells (TCR+ CD4- CD8-), with anti-inflammatory properties, were previously demonstrated to protect from early injury in moderate experimental acute kidney injury (AKI). However, their role in repair after AKI has not been studied. We hypothesized that DN T cells mediate repair after severe AKI. C57B6 mice underwent severe (40 min) unilateral ischemia-reperfusion injury (IRI). Kidney DN T cells were studied by flow cytometry and compared with gold-standard anti-inflammatory CD4+ regulatory T cells (Tregs). In vitro effects of DN T cells and Tregs on renal tubular epithelial cell (RTEC) repair after injury were quantified with live-cell analysis. DN T cells, Tregs, CD4, or vehicle were adoptively transferred after severe AKI. Glomerular filtration rate (GFR) was measured using fluorescein isothiocyanate (FITC)-sinistrin. Fibrosis was assessed with Masson's trichrome staining. Profibrotic genes were measured with qRT-PCR. Percentages and the numbers of DN T cells substantially decreased during repair phase after severe AKI, as well as their activation and proliferation. Both DN T cells and Tregs accelerated RTEC cell repair in vitro. Post-AKI transfer of DN T cells reduced kidney fibrosis and improved GFR, as did Treg transfer. DN T cell transfer lowered transforming growth factor (TGF)β1 and α-smooth muscle actin (αSMA) expression. DN T cells reduced effector-memory CD4+ T cells and IL-17 expression. DN T cells undergo quantitative and phenotypical changes after severe AKI, accelerate RTEC repair in vitro as well as improve GFR and renal fibrosis in vivo. DN T cells have potential as immunotherapy to accelerate repair after AKI.NEW & NOTEWORTHY Double-negative (DN) T cells (CD4- CD8-) are unconventional kidney T cells with regulatory abilities. Their role in repair from acute kidney injury (AKI) is unknown. Kidney DN T cell population decreased during repair after ischemic AKI, in contrast to regulatory T cells (Tregs) which increased. DN T cell administration accelerated tubular repair in vitro, while after severe in vivo ischemic injury reduced kidney fibrosis and increased glomerular filtration rate (GFR). DN T cell infusion is a potential therapeutic agent to improve outcome from severe AKI.
Collapse
Affiliation(s)
- Kyungho Lee
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
- Division of Nephrology, Department of Medicine, Cell and Gene Therapy Institute, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Sepideh Gharaie
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| | - Johanna T Kurzhagen
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| | - Andrea M Newman-Rivera
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| | - Lois J Arend
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| | - Sanjeev Noel
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| | - Hamid Rabb
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| |
Collapse
|
2
|
Wu J, Zhang X, Lin S, Wei Q, Lin Z, Jin O, Gu J. Alterations in peripheral T- and B-cell subsets in patients with systemic sclerosis. Int J Rheum Dis 2024; 27:e15145. [PMID: 38661314 DOI: 10.1111/1756-185x.15145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Revised: 03/22/2024] [Accepted: 03/27/2024] [Indexed: 04/26/2024]
Abstract
OBJECTIVES To determine the alteration of peripheral T and B cell subsets in patients with systemic sclerosis (SSc) and to evaluate their correlation with the progression of SSc. METHODS We recruited 47 SSc patients and 45 healthy controls (HCs) in this study. Demographic and clinical data were then collected. Flow cytometry was used to detect the proportions of 44 different T and B cell subsets in circulating blood. RESULTS The proportion of total B cells (p = .043) decreased in SSc patients, together with similar frequencies of total T cells, CD4+ T cells, and CD8+ T cells in both groups. Several subsets of T and B cells differed significantly between these two groups. Follicular helper T cells-1 (Tfh1) (p < .001), helper T cells-1 (Th1) (p = .001), regulatory T cells (Treg) (p = .004), effector memory CD8+ T cells (p = .041), and cytotoxic T cells-17 (Tc17) (p = .01) were decreased in SSc patients. Follicular helper T cells-2 (Tfh2) (p = .001) and, helper T cells-2 (Th2) (p = .001) levels increased in the SSc group. Regulatory B cells (Breg) (p = .015) were lower in the SSc group, together with marginal zone (MZ) B cells (p < .001), memory B cells (p = .001), and non-switched B cells (p = .005). The modified Rodnan skin score (mRSS) correlated with helper T cells-17 (Th17) (r = -.410, p = .004), Tfh1 (r = -.321, p = .028), peripheral helper T cells (Tph) (r = -.364, p = .012) and plasma cells (r = -.312, p = .033). CONCLUSIONS The alterations in T and B cells implied immune dysfunction, which may play an essential role in systemic sclerosis.
Collapse
Affiliation(s)
- Jialing Wu
- Department of Rheumatology and Immunology, Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Xi Zhang
- Department of Rheumatology and Immunology, Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Shen Lin
- Department of Rheumatology and Immunology, Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Qiujing Wei
- Department of Rheumatology and Immunology, Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Zhiming Lin
- Department of Rheumatology and Immunology, Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Ou Jin
- Department of Rheumatology and Immunology, Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Jieruo Gu
- Department of Rheumatology and Immunology, Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
| |
Collapse
|
3
|
Liakouli V, Ciancio A, Del Galdo F, Giacomelli R, Ciccia F. Systemic sclerosis interstitial lung disease: unmet needs and potential solutions. Nat Rev Rheumatol 2024; 20:21-32. [PMID: 37923862 DOI: 10.1038/s41584-023-01044-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/06/2023] [Indexed: 11/06/2023]
Abstract
Systemic sclerosis (SSc), or scleroderma, is a rare, complex, systemic autoimmune disease of unknown aetiology, characterized by high morbidity and mortality often resulting from cardiopulmonary complications such as interstitial lung disease and pulmonary arterial hypertension. Despite substantial progress in unravelling the pathways involved in the pathogenesis of SSc and the increasing number of therapeutic targets tested in clinical trials, there is still no cure for this disease, although several proposed treatments might limit the involvement of specific organs, thereby slowing the natural history of the disease. A specific focus of recent research has been to address the plethora of unmet needs regarding the global management of SSc-related interstitial lung disease, including its pathogenesis, early diagnosis, risk stratification of patients, appropriate treatment regimens and monitoring of treatment response, as well as the definition of progression and predictors of progression and mortality. More refined stratification of patients on the basis of clinical features, molecular signatures, identification of subpopulations with distinct clinical trajectories and implementation of outcome measures for future clinical trials could also improve therapeutic management strategies, helping to avoid poor outcomes related to lung involvement.
Collapse
Affiliation(s)
- Vasiliki Liakouli
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy.
| | - Antonio Ciancio
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Francesco Del Galdo
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, UK
- Scleroderma Programme, NIHR Biomedical Research Centre, Leeds Teaching Hospital Trusts, Leeds, UK
| | - Roberto Giacomelli
- Rheumatology and Immunology Unit, Department of Medicine, University of Rome Campus Biomedico, Rome, Italy
| | - Francesco Ciccia
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
| |
Collapse
|
4
|
Zhao K, Kong C, Shi N, Jiang J, Li P. Potential angiogenic, immunomodulatory, and antifibrotic effects of mesenchymal stem cell-derived extracellular vesicles in systemic sclerosis. Front Immunol 2023; 14:1125257. [PMID: 37251412 PMCID: PMC10213547 DOI: 10.3389/fimmu.2023.1125257] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 04/24/2023] [Indexed: 05/31/2023] Open
Abstract
Systemic sclerosis (SSc) is an intricate systemic autoimmune disease with pathological features such as vascular injury, immune dysregulation, and extensive fibrosis of the skin and multiple organs. Treatment options are limited; however, recently, mesenchymal stem cell-derived extracellular vesicles (MSC-EVs) have been acknowledged in preclinical and clinical trials as being useful in treating autoimmune diseases and are likely superior to MSCs alone. Recent research has also shown that MSC-EVs can ameliorate SSc and the pathological changes in vasculopathy, immune dysfunction, and fibrosis. This review summarizes the therapeutic effects of MSC-EVs on SSc and the mechanisms that have been discovered to provide a theoretical basis for future studies on the role of MSC-EVs in treating SSc.
Collapse
Affiliation(s)
- Kelin Zhao
- Department of Rheumatology and Immunology, China-Japan Union Hospital, Jilin University, Changchun, China
| | - Chenfei Kong
- Scientific Research Center, China-Japan Union Hospital, Jilin University, Changchun, China
| | - Naixu Shi
- Department of Stomatology, China-Japan Union Hospital, Jilin University, Changchun, China
| | - Jinlan Jiang
- Scientific Research Center, China-Japan Union Hospital, Jilin University, Changchun, China
| | - Ping Li
- Department of Rheumatology and Immunology, China-Japan Union Hospital, Jilin University, Changchun, China
| |
Collapse
|
5
|
Ciurea A, Rednic NV, Soancă A, Micu IC, Stanomir A, Oneț D, Șurlin P, Filipescu I, Roman A, Stratul ȘI, Pamfil C. Current Perspectives on Periodontitis in Systemic Sclerosis: Associative Relationships, Pathogenic Links, and Best Practices. Diagnostics (Basel) 2023; 13:diagnostics13050841. [PMID: 36899985 PMCID: PMC10000920 DOI: 10.3390/diagnostics13050841] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 02/15/2023] [Accepted: 02/20/2023] [Indexed: 02/25/2023] Open
Abstract
Systemic sclerosis is a chronic, autoimmune, multisystemic disease characterized by aberrant extracellular matrix protein deposition and extreme progressive microvasculopathy. These processes lead to damage within the skin, lungs, or gastrointestinal tract, but also to facial changes with physiognomic and functional alterations, and dental and periodontal lesions. Orofacial manifestations are common in SSc but are frequently overshadowed by systemic complications. In clinical practice, oral manifestations of SSc are suboptimally addressed, while their management is not included in the general treatment recommendations. Periodontitis is associated with autoimmune-mediated systemic diseases, including systemic sclerosis. In periodontitis, the microbial subgingival biofilm induces host-mediated inflammation with subsequent tissue damage, periodontal attachment, and bone loss. When these diseases coexist, patients experience additive damage, increasing malnutrition, and morbidity. The present review discusses the links between SSc and periodontitis, and provides a clinical guide for preventive and therapeutical approaches in the management of these patients.
Collapse
Affiliation(s)
- Andreea Ciurea
- Department of Periodontology, Faculty of Dental Medicine, Iuliu Hațieganu University of Medicine and Pharmacy Cluj-Napoca, Victor Babeș St., No. 15, 400012 Cluj-Napoca, Romania
| | - Nicolae Voicu Rednic
- Department of Gastroenterology, Iuliu Hațieganu University of Medicine and Pharmacy Cluj-Napoca, “Prof. Dr. Octavian Fodor” Regional Institute of Gastroenterology and Hepatology, Croitorilor St., No. 19, 400394 Cluj-Napoca, Romania
| | - Andrada Soancă
- Department of Periodontology, Faculty of Dental Medicine, Iuliu Hațieganu University of Medicine and Pharmacy Cluj-Napoca, Victor Babeș St., No. 15, 400012 Cluj-Napoca, Romania
| | - Iulia Cristina Micu
- Department of Periodontology, Faculty of Dental Medicine, Iuliu Hațieganu University of Medicine and Pharmacy Cluj-Napoca, Victor Babeș St., No. 15, 400012 Cluj-Napoca, Romania
| | - Alina Stanomir
- Department of Periodontology, Faculty of Dental Medicine, Iuliu Hațieganu University of Medicine and Pharmacy Cluj-Napoca, Victor Babeș St., No. 15, 400012 Cluj-Napoca, Romania
| | - Diana Oneț
- Department of Periodontology, Faculty of Dental Medicine, Iuliu Hațieganu University of Medicine and Pharmacy Cluj-Napoca, Victor Babeș St., No. 15, 400012 Cluj-Napoca, Romania
| | - Petra Șurlin
- Department of Periodontology, University of Medicine and Pharmacy Craiova, Petru Rareș St., No. 2, 200349 Craiova, Romania
| | - Ileana Filipescu
- Department of Rheumatology, Iuliu Hațieganu University of Medicine and Pharmacy Cluj-Napoca, Clinicilor St., No. 2, 400000 Cluj-Napoca, Romania
| | - Alexandra Roman
- Department of Periodontology, Faculty of Dental Medicine, Iuliu Hațieganu University of Medicine and Pharmacy Cluj-Napoca, Victor Babeș St., No. 15, 400012 Cluj-Napoca, Romania
- Correspondence: ; Tel.: +40-722-627-488
| | - Ștefan Ioan Stratul
- Department of Periodontology, Faculty of Dental Medicine, Anton Sculean Research Center for Periodontal and Peri-Implant Diseases, Victor Babeș University of Medicine and Pharmacy Timișoara, Revoluției from 1989 St., No. 9, 300041 Timișoara, Romania
| | - Cristina Pamfil
- Department of Rheumatology, Iuliu Hațieganu University of Medicine and Pharmacy Cluj-Napoca, Clinicilor St., No. 2, 400000 Cluj-Napoca, Romania
| |
Collapse
|
6
|
Udagawa C, Nakano MH, Yoshida T, Ohe Y, Kato K, Mushiroda T, Zembutsu H. Association between genetic variants and the risk of nivolumab-induced immune-related adverse events. Pharmacogenomics 2022; 23:887-901. [PMID: 36268685 DOI: 10.2217/pgs-2022-0113] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Aim: We sought to identify the variants that could predict the risk of nivolumab-induced immune-related adverse events (irAEs) in patients with cancer. Patients & methods: We enrolled 622 Japanese patients and carried out a genome-wide association study. The associations for 507 single nucleotide polymorphisms (SNPs) showing p < 0.001 were further investigated using an independent cohort. Results: In the combined analysis, possible associations were found for a total of 90 SNPs. Although no SNPs were identified to be significantly associated with nivolumab-induced irAEs, the SNP most strongly associated with nivolumab-induced irAEs was rs469490. Conclusion: This study is an important hypothesis-generating study to guide future studies in larger and/or other ethnic cohorts.
Collapse
Affiliation(s)
- Chihiro Udagawa
- Department of Genetics Medicine & services, National Cancer Center Hospital, Tokyo, 104-0045, Japan
| | - Mari Hara Nakano
- Division of Breast & Endocrine Surgery, Department of Surgery, St. Marianna University School of Medicine, Kawasaki, 216-8511, Japan
| | - Teruhiko Yoshida
- Department of Genetics Medicine & services, National Cancer Center Hospital, Tokyo, 104-0045, Japan
| | - Yuichiro Ohe
- Department of Thoracic Oncology, National Cancer Center Hospital, Tokyo, 104-0045, Japan
| | - Ken Kato
- Department of Gastrointestinal Medical Oncology, National Cancer Center Hospital, Tokyo, 104-0045, Japan
| | - Taisei Mushiroda
- Laboratory for Pharmacogenomics, RIKEN Center for Integrative Medical Science, Yokohama, 230-0045, Japan
| | - Hitoshi Zembutsu
- Department of Clinical Genomics, National Cancer Center Research Institute, Tokyo, 104-0045, Japan
| |
Collapse
|
7
|
Radziszewska A, Moulder Z, Jury EC, Ciurtin C. CD8 + T Cell Phenotype and Function in Childhood and Adult-Onset Connective Tissue Disease. Int J Mol Sci 2022; 23:11431. [PMID: 36232733 PMCID: PMC9569696 DOI: 10.3390/ijms231911431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 09/14/2022] [Accepted: 09/19/2022] [Indexed: 11/21/2022] Open
Abstract
CD8+ T cells are cytotoxic lymphocytes that destroy pathogen infected and malignant cells through release of cytolytic molecules and proinflammatory cytokines. Although the role of CD8+ T cells in connective tissue diseases (CTDs) has not been explored as thoroughly as that of other immune cells, research focusing on this key component of the immune system has recently gained momentum. Aberrations in cytotoxic cell function may have implications in triggering autoimmunity and may promote tissue damage leading to exacerbation of disease. In this comprehensive review of current literature, we examine the role of CD8+ T cells in systemic lupus erythematosus, Sjögren's syndrome, systemic sclerosis, polymyositis, and dermatomyositis with specific focus on comparing what is known about CD8+ T cell peripheral blood phenotypes, CD8+ T cell function, and CD8+ T cell organ-specific profiles in adult and juvenile forms of these disorders. Although, the precise role of CD8+ T cells in the initiation of autoimmunity and disease progression remains to be elucidated, increasing evidence indicates that CD8+ T cells are emerging as an attractive target for therapy in CTDs.
Collapse
Affiliation(s)
- Anna Radziszewska
- Centre for Adolescent Rheumatology Versus Arthritis at University College London (UCL), University College London Hospital (UCLH), Great Ormond Street Hospital (GOSH), London WC1E 6JF, UK
- Centre for Rheumatology Research, Division of Medicine, University College London, London WC1E 6JF, UK
| | - Zachary Moulder
- University College London Medical School, University College London, London WC1E 6DE, UK
| | - Elizabeth C. Jury
- Centre for Rheumatology Research, Division of Medicine, University College London, London WC1E 6JF, UK
| | - Coziana Ciurtin
- Centre for Adolescent Rheumatology Versus Arthritis at University College London (UCL), University College London Hospital (UCLH), Great Ormond Street Hospital (GOSH), London WC1E 6JF, UK
- Centre for Rheumatology Research, Division of Medicine, University College London, London WC1E 6JF, UK
| |
Collapse
|
8
|
Ware CF, Croft M, Neil GA. Realigning the LIGHT signaling network to control dysregulated inflammation. J Exp Med 2022; 219:213236. [PMID: 35604387 PMCID: PMC9130030 DOI: 10.1084/jem.20220236] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 04/29/2022] [Accepted: 05/03/2022] [Indexed: 11/10/2022] Open
Abstract
Advances in understanding the physiologic functions of the tumor necrosis factor superfamily (TNFSF) of ligands, receptors, and signaling networks are providing deeper insight into pathogenesis of infectious and autoimmune diseases and cancer. LIGHT (TNFSF14) has emerged as an important modulator of critical innate and adaptive immune responses. LIGHT and its signaling receptors, herpesvirus entry mediator (TNFRSF14), and lymphotoxin β receptor, form an immune regulatory network with two co-receptors of herpesvirus entry mediator, checkpoint inhibitor B and T lymphocyte attenuator, and CD160. Deciphering the fundamental features of this network reveals new understanding to guide therapeutic development. Accumulating evidence from infectious diseases points to the dysregulation of the LIGHT network as a disease-driving mechanism in autoimmune and inflammatory reactions in barrier organs, including coronavirus disease 2019 pneumonia and inflammatory bowel diseases. Recent clinical results warrant further investigation of the LIGHT regulatory network and application of target-modifying therapeutics for disease intervention.
Collapse
Affiliation(s)
- Carl F Ware
- Infectious and Inflammatory Diseases Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA
| | - Michael Croft
- Division of Immune Regulation, La Jolla Institute for Immunology, La Jolla, CA
| | | |
Collapse
|
9
|
Traber KE, Dimbo EL, Shenoy AT, Symer EM, Allen E, Mizgerd JP, Quinton LJ. Neutrophil-Derived Oncostatin M Triggers Diverse Signaling Pathways during Pneumonia. Infect Immun 2021; 89:e00655-20. [PMID: 33526570 PMCID: PMC8090961 DOI: 10.1128/iai.00655-20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Accepted: 01/11/2021] [Indexed: 11/20/2022] Open
Abstract
Pneumonia is a major public health concern, causing significant morbidity and mortality annually despite the broad use of antimicrobial agents. Underlying many of the severe sequelae of acute lung infections is dysfunction of the immune response, which remains incompletely understood yet is an attractive target of adjunct therapy in pneumonia. Here, we investigate the role of oncostatin M (OSM), a pleiotropic cytokine of the interleukin-6 (IL-6) family, and how its signaling modulates multiple innate immune pathways during pneumonia. Previously, we showed that OSM is necessary for neutrophil recruitment to the lungs during pneumonia by stimulating STAT3-driven CXCL5 expression. In this study, transcriptional profiling of whole-lung pneumonia with OSM neutralization revealed 241 differentially expressed genes following only 6 h of infection. Many downregulated genes are associated with STAT1, STAT3, and interferon signaling, suggesting these pathways are induced by OSM early in pneumonia. Interestingly, STAT1 and STAT3 activation was subsequently upregulated with OSM neutralization by 24 h, suggesting that OSM interruption dysregulates these central signaling pathways. When we investigated the source of OSM in pneumonia, neutrophils and, to a lesser extent, macrophages appear to be primary sources, suggesting a positive feedback loop of OSM production by neutrophils. From these studies, we conclude that OSM produced by recruited neutrophils tunes early innate immune signaling pathways, improving pneumonia outcomes.
Collapse
Affiliation(s)
- Katrina E Traber
- Pulmonary Center, Boston University School of Medicine, Boston, Massachusetts, USA
- Department of Medicine, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Ernest L Dimbo
- Pulmonary Center, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Anukul T Shenoy
- Pulmonary Center, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Elise M Symer
- Pulmonary Center, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Eri Allen
- Pulmonary Center, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Joseph P Mizgerd
- Pulmonary Center, Boston University School of Medicine, Boston, Massachusetts, USA
- Department of Medicine, Boston University School of Medicine, Boston, Massachusetts, USA
- Department of Microbiology, Boston University School of Medicine, Boston, Massachusetts, USA
- Department of Biochemistry, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Lee J Quinton
- Pulmonary Center, Boston University School of Medicine, Boston, Massachusetts, USA
- Department of Medicine, Boston University School of Medicine, Boston, Massachusetts, USA
- Department of Microbiology, Boston University School of Medicine, Boston, Massachusetts, USA
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, Massachusetts, USA
| |
Collapse
|
10
|
Mesenchymal stromal cells for systemic sclerosis treatment. Autoimmun Rev 2021; 20:102755. [PMID: 33476823 DOI: 10.1016/j.autrev.2021.102755] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 12/10/2020] [Indexed: 02/06/2023]
Abstract
Systemic sclerosis (SSc) is a rare chronic autoimmune disease characterized by vasculopathy, dysregulation of innate and adaptive immune responses, and progressive fibrosis. SSc remains an orphan disease, with high morbity and mortality in SSc patients. The mesenchymal stromal cells (MSC) demonstrate in vitro and in vivo pro-angiogenic, immuno-suppressive, and anti-fibrotic properties and appear as a promising stem cell therapy type, that may target the key pathological features of SSc disease. This review aims to summarize acquired knowledge in the field of :1) MSC definition and in vitro and in vivo functional properties, which vary according to the donor type (allogeneic or autologous), the tissue sources (bone marrow, adipose tissue or umbilical cord) or inflammatory micro-environment in the recipient; 2) preclinical studies in various SSc animal models , which showed reduction in skin and lung fibrosis after MSC infusion; 3) first clinical trials in human, with safety and early efficacy results reported in SSc patients or currently tested in several ongoing clinical trials.
Collapse
|
11
|
Gapud EJ, Trejo-Zambrano MI, Gomez-Banuelos E, Tiniakou E, Antiochos B, Granville DJ, Andrade F, Casciola-Rosen L, Rosen A. Granzyme B Induces IRF-3 Phosphorylation through a Perforin-Independent Proteolysis-Dependent Signaling Cascade without Inducing Cell Death. THE JOURNAL OF IMMUNOLOGY 2020; 206:335-344. [PMID: 33288544 DOI: 10.4049/jimmunol.2000546] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Accepted: 11/11/2020] [Indexed: 11/19/2022]
Abstract
Granzyme B (GrB) is an immune protease implicated in the pathogenesis of several human diseases. In the current model of GrB activity, perforin determines whether the downstream actions of GrB occur intracellularly or extracellularly, producing apoptotic cytotoxicity or nonapoptotic effects, respectively. In the current study, we demonstrate the existence of a broad range of GrB-dependent signaling activities that 1) do not require perforin, 2) occur intracellularly, and 3) for which cell death is not the dominant outcome. In the absence of perforin, we show that GrB enzymatic activity still induces substoichiometric activation of caspases, which through nonlethal DNA damage response signals then leads to activity-associated phosphorylation of IFN regulatory factor-3. These findings illustrate an unexpected potential interface between GrB and innate immunity separate from the traditional role of GrB in perforin-dependent GrB-mediated apoptosis that could have mechanistic implications for human disease.
Collapse
Affiliation(s)
- Eric J Gapud
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21224
| | | | - Eduardo Gomez-Banuelos
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21224
| | - Eleni Tiniakou
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21224
| | - Brendan Antiochos
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21224
| | - David J Granville
- International Collaboration on Repair Discoveries Centre, Vancouver Coastal Health Research Institute, University of British Columbia, Vancouver, British Columbia V5Z 1M9, Canada.,Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia V5Z 1M9, Canada
| | - Felipe Andrade
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21224
| | - Livia Casciola-Rosen
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21224
| | - Antony Rosen
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21224; .,Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21224; and.,Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD 21224
| |
Collapse
|
12
|
Alzghoul BN, Hamburger R, Lewandowski T, Janssen B, Grey D, Xue W, Ataya A, Alnuaimat H, Gomez-Manjarres DC, Patel D, Reddy R. Pulmonary hypertension in patients with interstitial pneumonia with autoimmune features. Pulm Circ 2020; 10:2045894020944117. [PMID: 33343878 DOI: 10.1177/2045894020944117] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Accepted: 06/29/2020] [Indexed: 11/16/2022] Open
Abstract
Pulmonary hypertension in interstitial lung diseases is associated with increased mortality and hospitalizations and reduced exercise capacity. Interstitial pneumonia with autoimmune features (IPAF) is a recently described interstitial lung disease. The characteristics of pulmonary hypertension in IPAF patients are unknown. We sought to characterize patients with IPAF based on their echocardiographic probability of pulmonary hypertension and compare patients with and without pulmonary hypertension identified by right heart catheterization. We conducted a retrospective study of patients seen in the interstitial lung disease clinic from 2015 to 2018. Forty-seven patients with IPAF were identified. Patients were classified into low, intermediate and high echocardiographic pulmonary hypertension probabilities. A sub-group analysis of patients with pulmonary hypertension and without pulmonary hypertension (IPAF-PH vs. IPAF-no PH) identified by right heart catheterization was also performed. Linear regression analysis was performed to study the association between 6-min-walk-distance (6MWD) and pulmonary vascular resistance (PVR) while adjusting for age and body mass index. Right ventricular hypertrophy (>5 mm), right ventricular enlargement (>41 mm) and right ventricular systolic dysfunction defined as fractional area change% ≤35 was present in 76%, 24%, and 39% of patients, respectively. Pulmonary hypertension was identified in 12.7% of patients. IPAF-PH patients had higher mean pulmonary artery pressure and lower cardiac output compared to the IPAF-no PH group (34 mmHg vs. 19 mmHg, p = 0.002 and 4.0 vs. 5.7 L/min, p = 0.023, respectively). Lower 6MWD was associated with higher PVR on regression analysis (p = 0.002). Pulmonologists should be aware that a significant number of IPAF patients may develop pulmonary hypertension. Reduced 6MWD may suggest the presence of pulmonary hypertension in IPAF patients.
Collapse
Affiliation(s)
- Bashar N Alzghoul
- Division of Pulmonary, Critical Care and Sleep Medicine, University of Florida, Gainesville, FL, USA
| | - Robert Hamburger
- Division of Cardiovascular Medicine, University of Florida, Gainesville, FL, USA
| | - Thomas Lewandowski
- Division of Cardiovascular Medicine, University of Florida, Gainesville, FL, USA
| | - Brandon Janssen
- Department of Internal Medicine, University of Florida, Gainesville, FL, USA
| | - Daniel Grey
- Department of Internal Medicine, University of Florida, Gainesville, FL, USA
| | - Wei Xue
- Department of Biostatistics, College of Public Health and Health Professions and College of Medicine, University of Florida, Gainesville, FL, USA
| | - Ali Ataya
- Division of Pulmonary, Critical Care and Sleep Medicine, University of Florida, Gainesville, FL, USA
| | - Hassan Alnuaimat
- Division of Pulmonary, Critical Care and Sleep Medicine, University of Florida, Gainesville, FL, USA
| | - Diana C Gomez-Manjarres
- Division of Pulmonary, Critical Care and Sleep Medicine, University of Florida, Gainesville, FL, USA
| | - Divya Patel
- Division of Pulmonary, Critical Care and Sleep Medicine, University of Florida, Gainesville, FL, USA
| | - Raju Reddy
- Division of Pulmonary, Critical Care and Sleep Medicine, University of Florida, Gainesville, FL, USA
| |
Collapse
|
13
|
Nihtyanova SI, Denton CP. Pathogenesis of systemic sclerosis associated interstitial lung disease. JOURNAL OF SCLERODERMA AND RELATED DISORDERS 2020; 5:6-16. [PMID: 35382227 PMCID: PMC8922569 DOI: 10.1177/2397198320903867] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Accepted: 12/24/2019] [Indexed: 12/12/2022]
Abstract
Systemic sclerosis is an autoimmune disease leading to vasculopathy and fibrosis
of skin and internal organs. Despite likely shared pathogenic mechanisms, the
patterns of skin and lung fibrosis differ. Pathogenesis of interstitial lung
disease, a major cause of death in systemic sclerosis, reflects the intrinsic
disease pathobiology and is associated with distinct clinical phenotypes and
laboratory characteristics. The commonest histological pattern of systemic
sclerosis–interstitial lung disease is non-specific interstitial pneumonia.
Systemic sclerosis–interstitial lung disease pathogenesis involves multiple
components, including susceptibility and triggering factors, which could be
genetic or environmental. The process is amplified likely through ongoing
inflammation and the link between inflammatory activity and fibrosis with IL6
emerging as a key mediator. The disease is driven by epithelial injury,
reflected by markers in the serum, such as surfactant proteins and KL-6. In
addition, mediators that are produced by epithelial cells and that regulate
inflammatory cell trafficking may be important, especially CCL2. Other factors,
such as CXCL4 and CCL18, point towards immune-mediated damage or injury
response. Monocytes and alternatively activated macrophages appear to be
important. Transforming growth factor beta appears central to pathogenesis and
regulates epithelial repair and fibroblast activation. Understanding
pathogenesis may help to unravel the stages of systemic sclerosis–interstitial
lung disease, risks of progression and determinants of outcome. With this
article, we set out to review the multiple factors, including genetic,
environmental, cellular and molecular, that may be involved in the pathogenesis
of systemic sclerosis–interstitial lung disease and the mechanisms leading to
sustained fibrosis. We propose a model for the pathogenesis of systemic
sclerosis–interstitial lung disease, based on the available literature.
Collapse
Affiliation(s)
- Svetlana I Nihtyanova
- Centre for Rheumatology and Connective Tissue Diseases, University College London, London, UK
| | - Christopher P Denton
- Centre for Rheumatology and Connective Tissue Diseases, University College London, London, UK
| |
Collapse
|
14
|
Richards CD, Botelho F. Oncostatin M in the Regulation of Connective Tissue Cells and Macrophages in Pulmonary Disease. Biomedicines 2019; 7:E95. [PMID: 31817403 PMCID: PMC6966661 DOI: 10.3390/biomedicines7040095] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 11/23/2019] [Accepted: 11/26/2019] [Indexed: 12/16/2022] Open
Abstract
Oncostatin M (OSM), as one of the gp130/IL-6 family of cytokines, interacts with receptor complexes that include the gp130 signaling molecule and OSM receptor β OSMRβ chain subunits. OSMRβ chains are expressed relatively highly across a broad array of connective tissue (CT) cells of the lung, such as fibroblasts, smooth muscle cells, and epithelial cells, thus enabling robust responses to OSM, compared to other gp130 cytokines, in the regulation of extracellular matrix (ECM) remodeling and inflammation. OSMRβ chain expression in lung monocyte/macrophage populations is low, whereas other receptor subunits, such as that for IL-6, are present, enabling responses to IL-6. OSM is produced by macrophages and neutrophils, but not CT cells, indicating a dichotomy of OSM roles in macrophage verses CT cells in lung inflammatory disease. ECM remodeling and inflammation are components of a number of chronic lung diseases that show elevated levels of OSM. OSM-induced products of CT cells, such as MCP-1, IL-6, and PGE2 can modulate macrophage function, including the expression of OSM itself, indicating feedback loops that characterize Macrophage and CT cell interaction.
Collapse
Affiliation(s)
- Carl D. Richards
- McMaster Immunology Research Centre, Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON L8S 3Z5, Canada;
| | | |
Collapse
|
15
|
Wu B, Rockel JS, Lagares D, Kapoor M. Ephrins and Eph Receptor Signaling in Tissue Repair and Fibrosis. Curr Rheumatol Rep 2019; 21:23. [PMID: 30980212 DOI: 10.1007/s11926-019-0825-x] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
PURPOSE OF REVIEW Fibrosis is a pathological feature of many human diseases that affect multiple organs. The development of anti-fibrotic therapies has been a difficult endeavor due to the complexity of signaling pathways associated with fibrogenic processes, complicating the identification and modulation of specific targets. Evidence suggests that ephrin ligands and Eph receptors are crucial signaling molecules that contribute to physiological wound repair and the development of tissue fibrosis. Here, we discuss recent advances in the understanding of ephrin and Eph signaling in tissue repair and fibrosis. RECENT FINDINGS Ephrin-B2 is implicated in fibrosis of multiple organs. Intercepting its signaling may help counteract fibrosis. Ephrins and Eph receptors are candidate mediators of fibrosis. Ephrin-B2, in particular, promotes fibrogenic processes in multiple organs. Thus, therapeutic strategies targeting Ephrin-B2 signaling could yield new ways to treat organ fibrosis.
Collapse
Affiliation(s)
- Brian Wu
- The Arthritis Program, Krembil Research Institute, University Health Network, Toronto, Ontario, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Jason S Rockel
- The Arthritis Program, Krembil Research Institute, University Health Network, Toronto, Ontario, Canada
| | - David Lagares
- Center for Immunology and Inflammatory Diseases, Division of Rheumatology, Allergy and Immunology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA. .,Department of Medicine, Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA. .,Fibrosis Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| | - Mohit Kapoor
- The Arthritis Program, Krembil Research Institute, University Health Network, Toronto, Ontario, Canada. .,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada. .,Department of Surgery, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
16
|
Abstract
Oncostain M, a member of the IL-6 family of cytokines, is produced by immune cells in response to infections and tissue injury. OSM has a broad, often context-dependent effect on various cellular processes including differentiation, hematopoiesis, cell proliferation, and cell survival. OSM signaling is initiated by binding to type I (LIFRβ/gp130) or type II (OSMRβ/gp130) receptor complexes and involves activation of Janus kinase/signal transducer and activator of transcription, mitogen-activated protein kinase, and phosphatidylinositol-3-kinase. High levels of OSM have been detected in many chronic inflammatory conditions characterized by fibrosis, giving a rationale to target OSM for the treatment of these diseases. Here we discuss the current knowledge on the role of OSM in various stages of the fibrotic process including inflammation, vascular dysfunction, and activation of fibroblasts.
Collapse
Affiliation(s)
| | - Maria Trojanowska
- Corresponding Author: Maria Trojanowska, Boston University School of Medicine, 72 East Concord St, E-5, Boston, MA 02118, Tel.: 617-638-4318; Fax: 617-638-5226
| |
Collapse
|
17
|
Brown M, O'Reilly S. The immunopathogenesis of fibrosis in systemic sclerosis. Clin Exp Immunol 2018; 195:310-321. [PMID: 30430560 DOI: 10.1111/cei.13238] [Citation(s) in RCA: 119] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/07/2018] [Indexed: 02/07/2023] Open
Abstract
Systemic sclerosis (SSc) is an idiopathic systemic autoimmune disease. It is characterized by a triad of hallmarks: immune dysfunction, fibrosis and vasculopathy. Immune dysfunction in SSc is characterized by the activation and recruitment of immune cells and the production of autoantibodies and cytokines. How immune abnormalities link the fibrosis and vasculopathy in SSc is poorly understood. A plethora of immune cell types are implicated in the immunopathogenesis of SSc, including T cells, B cells, dendritic cells, mast cells and macrophages. How these different cell types interact to contribute to SSc is complicated, and can involve cell-to-cell interactions and communication via cytokines, including transforming growth factor (TGF)-β, interleukin (IL)-6 and IL-4. We will attempt to review significant and recent research demonstrating the importance of immune cell regulation in the immunopathogenesis of SSc with a particular focus on fibrosis.
Collapse
Affiliation(s)
- M Brown
- Faculty of Health and Life Sciences, Northumbria University, Newcastle upon Tyne, UK
| | - S O'Reilly
- Faculty of Health and Life Sciences, Northumbria University, Newcastle upon Tyne, UK
| |
Collapse
|
18
|
West NR, Owens BMJ, Hegazy AN. The oncostatin M-stromal cell axis in health and disease. Scand J Immunol 2018; 88:e12694. [DOI: 10.1111/sji.12694] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2018] [Accepted: 06/15/2018] [Indexed: 12/17/2022]
Affiliation(s)
- Nathaniel R. West
- Department of Cancer Immunology; Genentech; South San Francisco California
| | - Benjamin M. J. Owens
- Somerville College; University of Oxford; Oxford UK
- EUSA Pharma; Hemel Hempstead UK
| | - Ahmed N. Hegazy
- Division of Gastroenterology, Infectiology, and Rheumatology; Charité Universitätsmedizin; Berlin Germany
- Deutsches Rheuma-Forschungszentrum; ein Institut der Leibniz-Gemeinschaft; Berlin Germany
| |
Collapse
|
19
|
Atzeni F, Gerardi MC, Barilaro G, Masala IF, Benucci M, Sarzi-Puttini P. Interstitial lung disease in systemic autoimmune rheumatic diseases: a comprehensive review. Expert Rev Clin Immunol 2017; 14:69-82. [DOI: 10.1080/1744666x.2018.1411190] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Affiliation(s)
- Fabiola Atzeni
- Maurizio Benucci, Rheumatology Unit, San Giovanni Di Dio Hospital, Florence, Italy
| | - Maria Chiara Gerardi
- Rheumatology Unit, Department of Internal Medicine, ASST-Fatebenefratelli L. Sacco University Hospital, Milan, Italy
| | | | | | - Maurizio Benucci
- Rheumatology Unit, Department of Internal Medicine, ASST-Fatebenefratelli L. Sacco University Hospital, Milan, Italy
| | - Piercarlo Sarzi-Puttini
- Rheumatology Unit, Department of Internal Medicine, ASST-Fatebenefratelli L. Sacco University Hospital, Milan, Italy
| |
Collapse
|
20
|
Komori T, Morikawa Y. Oncostatin M in the development of metabolic syndrome and its potential as a novel therapeutic target. Anat Sci Int 2017; 93:169-176. [PMID: 29103176 DOI: 10.1007/s12565-017-0421-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2017] [Accepted: 10/28/2017] [Indexed: 01/01/2023]
Abstract
Oncostatin M (OSM), a member of the IL-6 family of cytokines, plays an important role in various biologic actions, including cell growth, neuronal development, and inflammatory responses. Recently, we demonstrated the unique relationship between OSM and metabolic syndrome in mice. Mice lacking OSM receptor β subunit (OSMRβ-/- mice) exhibited late-onset obesity. Before the onset of obesity, adipose tissue inflammation and insulin resistance were observed in OSMRβ-/- mice. In addition, high-fat diet-induced metabolic disorders, including obesity, adipose tissue inflammation, insulin resistance, and hepatic steatosis, were aggravated in OSMRβ-/- mice compared to those in wild-type mice. Consistent with these findings, OSM treatment dramatically improved these metabolic disorders in the mouse model of metabolic syndrome. Interestingly, OSM directly changed the phenotypes of adipose tissue macrophages toward anti-inflammatory M2 type. Furthermore, fatty acid content in the hepatocytes was decreased by OSM through expression regulation of several key enzymes of hepatic lipid metabolism. These findings suggest that OSM is a novel therapeutic target for metabolic syndrome.
Collapse
Affiliation(s)
- Tadasuke Komori
- Department of Anatomy and Neurobiology, Wakayama Medical University, 811-1 Kimiidera, Wakayama, 641-8509, Japan.
| | - Yoshihiro Morikawa
- Department of Anatomy and Neurobiology, Wakayama Medical University, 811-1 Kimiidera, Wakayama, 641-8509, Japan
| |
Collapse
|
21
|
Fuschiotti P. Current perspectives on the role of CD8+ T cells in systemic sclerosis. Immunol Lett 2017; 195:55-60. [PMID: 28987475 DOI: 10.1016/j.imlet.2017.10.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2017] [Revised: 10/03/2017] [Accepted: 10/04/2017] [Indexed: 02/06/2023]
Abstract
Despite long-standing recognition of the importance of T cells in systemic sclerosis (SSc; scleroderma), the role of CD8+ T cells in disease pathogenesis has not been well studied. Our work has shown that over-production of the pro-fibrotic cytokine IL-13 by peripheral blood effector/memory CD8+ T cells is critical for predisposing patients to more severe forms of cutaneous fibrosis. Moreover, IL-13-producing CD8+ T cells induce a pro-fibrotic phenotype in normal and SSc dermal fibroblasts, and exhibit a strong cytotoxic activity ex vivo. We also found that CD8+ T cells are predominantly abundant in the skin lesions of patients in the early stages of diffuse cutaneous (dc)SSc compare to late-stage disease patients. Isolation of CD8+ T cells from the lesional skin of early active dcSSc patients, established that they are skin-resident, express cytolytic molecules and co-express extremely high levels of IL-13 and IFNγ. Other recent studies corroborate these findings and together strongly suggest that CD8+ T cells contribute to SSc pathogenesis through the production of high levels of cytokines with pro-inflammatory and pro-fibrotic function as well as by exhibiting a cytotoxic activity.
Collapse
Affiliation(s)
- Patrizia Fuschiotti
- Department of Medicine, Division of Rheumatology and Clinical Immunology, University of Pittsburgh School of Medicine, S709 BST, 200 Lothrop Street, Pittsburgh, PA 15261, USA.
| |
Collapse
|
22
|
Interleukin-6 and Related Proteins as Biomarkers in Systemic Sclerosis. JOURNAL OF SCLERODERMA AND RELATED DISORDERS 2017. [DOI: 10.5301/jsrd.5000266] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
The search for biomarkers in systemic sclerosis (SSc) is driven by a goal to stratify patients, identify potential subgroups for treatment, and help assess response to therapy. Emerging evidence indicates that interleukin-6 (IL-6) and some family members are key biomarkers involved in SSc pathogenesis and therefore suitable targets for therapy. Recent studies evaluating IL-6 and its canonical Janus kinase/signal transducers and activators of transcription downstream pathways in modulating fibrotic response and immune cell function suggest a pivotal role for IL-6 in SSc pathogenesis. Although the significance and effect of local tissue expression of IL-6 and its family members are less well established, high levels of circulating IL-6 may identify subgroups of patients with early-stage disease, particularly those at risk for progressive lung fibrosis. In addition, higher disease activity may portend poor prognostic outcome in terms of survival and skin disease. Longitudinal assessment of serum levels of IL-6 and its signaling associates may prove valuable in monitoring response to treatment. As an IL-6–dependent surrogate marker, C-reactive protein may assist cohort enrichment if targeted treatment for IL-6 demonstrates efficacy, especially in subgroups with high IL-6 levels. Although IL-6 appears to be a key factor in the hierarchy of the complex network of disease-associated molecules, the systemic or autocrine/paracrine manner in which IL-6 asserts its profibrotic effects—particularly its interaction with other key pathogenic factors in SSc—is unknown. Ongoing clinical trials will help to delineate the mechanisms of IL-6 in SSc pathogenesis and inform on the role of these biomarkers.
Collapse
|
23
|
Zuo X, Zhang L, Luo H, Li Y, Zhu H. Systematic approach to understanding the pathogenesis of systemic sclerosis. Clin Genet 2017; 92:365-371. [PMID: 27918067 DOI: 10.1111/cge.12946] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Revised: 11/24/2016] [Accepted: 11/27/2016] [Indexed: 12/13/2022]
Affiliation(s)
- Xiaoxia Zuo
- Department of Rheumatology, Xiangya Hospital; Central South University; Changsha People's Republic of China
| | - Lihua Zhang
- Department of Rheumatology, Xiangya Hospital; Central South University; Changsha People's Republic of China
| | - Hui Luo
- Department of Rheumatology, Xiangya Hospital; Central South University; Changsha People's Republic of China
| | - Yisha Li
- Department of Rheumatology, Xiangya Hospital; Central South University; Changsha People's Republic of China
| | - Honglin Zhu
- Department of Rheumatology, Xiangya Hospital; Central South University; Changsha People's Republic of China
| |
Collapse
|
24
|
Interstitial lung disease in systemic sclerosis: current and future treatment. Rheumatol Int 2017; 37:853-863. [PMID: 28063071 DOI: 10.1007/s00296-016-3636-7] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2016] [Accepted: 12/19/2016] [Indexed: 12/28/2022]
Abstract
Systemic sclerosis (SSc) has the highest fatality rate among connective tissue diseases and is characterized by vascular damage, inflammation and fibrosis of the skin and various internal organs. Interstitial lung disease (ILD) frequently complicates SSc and can be a debilitating disorder with a poor prognosis. ILD is the most frequent cause of death in SSc, and the management of SSc-ILD patients is a great challenge. Early detection of pulmonary involvement based on a recent decline of lung function tests and on the extent of lung involvement at high-resolution computed tomography is critical for the best management of these patients. This article summarizes classification, pathogenesis, diagnosis, prognosis, survival and finally current and future treatment options in SSc-ILD.
Collapse
|
25
|
Fuschiotti P. Current perspectives on the immunopathogenesis of systemic sclerosis. Immunotargets Ther 2016; 5:21-35. [PMID: 27529059 PMCID: PMC4970639 DOI: 10.2147/itt.s82037] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Systemic sclerosis (SSc or scleroderma) is a progressive and highly debilitating autoimmune disorder characterized by inflammation, vasculopathy, and extensive fibrosis. SSc is highly heterogeneous in its clinical presentation, extent and severity of skin and internal organ involvement, and clinical course and has the highest fatality rate among connective tissue diseases. While clinical outcomes have improved in recent years, no current therapy is able to reverse or slow the natural progression of SSc, a reflection of its complex pathogenesis. Although activation of the immune system has long been recognized, the mechanisms responsible for the initiation of autoimmunity and the role of immune effector pathways in the pathogenesis of SSc remain incompletely understood. This review summarizes recent progress in disease pathogenesis with particular focus on the immunopathogenetic mechanisms of SSc.
Collapse
Affiliation(s)
- Patrizia Fuschiotti
- Division of Rheumatology and Clinical Immunology, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| |
Collapse
|
26
|
Luzina IG, Lockatell V, Hyun SW, Kopach P, Kang PH, Noor Z, Liu A, Lillehoj EP, Lee C, Miranda-Ribera A, Todd NW, Goldblum SE, Atamas SP. Elevated expression of NEU1 sialidase in idiopathic pulmonary fibrosis provokes pulmonary collagen deposition, lymphocytosis, and fibrosis. Am J Physiol Lung Cell Mol Physiol 2016; 310:L940-54. [PMID: 26993524 DOI: 10.1152/ajplung.00346.2015] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Accepted: 03/17/2016] [Indexed: 01/08/2023] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) poses challenges to understanding its underlying cellular and molecular mechanisms and the development of better therapies. Previous studies suggest a pathophysiological role for neuraminidase 1 (NEU1), an enzyme that removes terminal sialic acid from glycoproteins. We observed increased NEU1 expression in epithelial and endothelial cells, as well as fibroblasts, in the lungs of patients with IPF compared with healthy control lungs. Recombinant adenovirus-mediated gene delivery of NEU1 to cultured primary human cells elicited profound changes in cellular phenotypes. Small airway epithelial cell migration was impaired in wounding assays, whereas, in pulmonary microvascular endothelial cells, NEU1 overexpression strongly impacted global gene expression, increased T cell adhesion to endothelial monolayers, and disrupted endothelial capillary-like tube formation. NEU1 overexpression in fibroblasts provoked increased levels of collagen types I and III, substantial changes in global gene expression, and accelerated degradation of matrix metalloproteinase-14. Intratracheal instillation of NEU1 encoding, but not control adenovirus, induced lymphocyte accumulation in bronchoalveolar lavage samples and lung tissues and elevations of pulmonary transforming growth factor-β and collagen. The lymphocytes were predominantly T cells, with CD8(+) cells exceeding CD4(+) cells by nearly twofold. These combined data indicate that elevated NEU1 expression alters functional activities of distinct lung cell types in vitro and recapitulates lymphocytic infiltration and collagen accumulation in vivo, consistent with mechanisms implicated in lung fibrosis.
Collapse
Affiliation(s)
- Irina G Luzina
- Baltimore Veterans Affairs Medical Center, Baltimore, Maryland; University of Maryland School of Medicine, Baltimore, Maryland; and
| | - Virginia Lockatell
- Baltimore Veterans Affairs Medical Center, Baltimore, Maryland; University of Maryland School of Medicine, Baltimore, Maryland; and
| | - Sang W Hyun
- University of Maryland School of Medicine, Baltimore, Maryland; and
| | - Pavel Kopach
- University of Maryland School of Medicine, Baltimore, Maryland; and
| | - Phillip H Kang
- University of Maryland School of Medicine, Baltimore, Maryland; and
| | - Zahid Noor
- University of Maryland School of Medicine, Baltimore, Maryland; and
| | - Anguo Liu
- University of Maryland School of Medicine, Baltimore, Maryland; and
| | - Erik P Lillehoj
- University of Maryland School of Medicine, Baltimore, Maryland; and
| | - Chunsik Lee
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, People's Republic of China
| | | | - Nevins W Todd
- Baltimore Veterans Affairs Medical Center, Baltimore, Maryland; University of Maryland School of Medicine, Baltimore, Maryland; and
| | - Simeon E Goldblum
- Baltimore Veterans Affairs Medical Center, Baltimore, Maryland; University of Maryland School of Medicine, Baltimore, Maryland; and
| | - Sergei P Atamas
- Baltimore Veterans Affairs Medical Center, Baltimore, Maryland; University of Maryland School of Medicine, Baltimore, Maryland; and
| |
Collapse
|
27
|
Desbois AC, Cacoub P. Systemic sclerosis: An update in 2016. Autoimmun Rev 2016; 15:417-26. [PMID: 26802722 DOI: 10.1016/j.autrev.2016.01.007] [Citation(s) in RCA: 104] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Accepted: 01/11/2016] [Indexed: 01/27/2023]
Abstract
Systemic sclerosis (SSc) is a chronic immune disorder of unknown origin, dominated by excessive fibrosis responsible for cutaneous and pulmonary fibrosis, and by vascular endothelial dysfunction at the origin of skin ischemia, renal and pulmonary artery lesions. Renal and pulmonary complications are mainly responsible for the severity of the disease. Recent advances led to a better understanding of pathological mechanisms and a more accurate classification of patients according to clinical and biological (auto-antibodies) phenotype. Recent trials provided interesting data on different therapeutic strategies, depending on organ involvement. These data are of particular importance in such disease, still characterized by increased mortality and morbidity rates. In this review, we aim to synthetize recent advances in diagnosis and prognosis leading to better classification of SSc patients, and in therapeutic management.
Collapse
Affiliation(s)
- Anne Claire Desbois
- Sorbonne Universités, UPMC Université Paris 06, UMR 7211, Paris, France; Inflammation-Immunopathology-Biotherapy Department (DHU i2B), F-75005 Paris, France; INSERM, UMR_S 959, F-75013 Paris, France; CNRS, FRE3632, F-75005 Paris, France
| | - Patrice Cacoub
- Sorbonne Universités, UPMC Université Paris 06, UMR 7211, Paris, France; Inflammation-Immunopathology-Biotherapy Department (DHU i2B), F-75005 Paris, France; AP-HP, Groupe Hospitalier Pitié-Salpêtrière, Department of Internal Medicine and Clinical Immunology, 83 boulevard de l'hôpital, F-75013 Paris, France.
| |
Collapse
|
28
|
Luzina IG, Lockatell V, Todd NW, Kopach P, Pentikis HS, Atamas SP. Pharmacological In Vivo Inhibition of S-Nitrosoglutathione Reductase Attenuates Bleomycin-Induced Inflammation and Fibrosis. J Pharmacol Exp Ther 2015; 355:13-22. [PMID: 26209236 DOI: 10.1124/jpet.115.224675] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2015] [Accepted: 07/23/2015] [Indexed: 01/17/2023] Open
Abstract
Interstitial lung disease (ILD) characterized by pulmonary fibrosis and inflammation poses a substantial biomedical challenge due to often negative disease outcomes combined with the need to develop better, more effective therapies. We assessed the in vivo effect of administration of a pharmacological inhibitor of S-nitrosoglutathione reductase, SPL-334 (4-{[2-[(2-cyanobenzyl)thio]-4-oxothieno[3,2-d]pyrimidin-3(4H)-yl]methyl}benzoic acid), in a mouse model of ILD induced by intratracheal instillation of bleomycin (BLM). Daily i.p. administration of SPL-334 alone at 0.3, 1.0, or 3.0 mg/kg had no effect on animal body weight, appearance, behavior, total and differential bronchoalveolar lavage (BAL) cell counts, or collagen accumulation in the lungs, showing no toxicity of our investigational compound. Similar administration of SPL-334 for 7 days before and for an additional 14 days after BLM instillation resulted in a preventive protective effect on the BLM challenge-induced decline in total body weight and changes in total and differential BAL cellularity. In the therapeutic treatment regimen, SPL-334 was administered at days 7-21 after BLM challenge. Such treatment attenuated the BLM challenge-induced decline in total body weight, changes in total and differential BAL cellularity, and magnitudes of histologic changes and collagen accumulation in the lungs. These changes were accompanied by an attenuation of BLM-induced elevations in pulmonary levels of profibrotic cytokines interleukin-6, monocyte chemoattractant protein-1, and transforming growth factor-β (TGF-β). Experiments in cell cultures of primary normal human lung fibroblast have demonstrated attenuation of TGF-β-induced upregulation in collagen by SPL-334. It was concluded that SPL-334 is a potential therapeutic agent for ILD.
Collapse
Affiliation(s)
- Irina G Luzina
- Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland (I.G.L., V.L., N.W.T., P.K., S.P.A); and SAJE Pharma, Baltimore, Maryland (H.S.P.)
| | - Virginia Lockatell
- Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland (I.G.L., V.L., N.W.T., P.K., S.P.A); and SAJE Pharma, Baltimore, Maryland (H.S.P.)
| | - Nevins W Todd
- Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland (I.G.L., V.L., N.W.T., P.K., S.P.A); and SAJE Pharma, Baltimore, Maryland (H.S.P.)
| | - Pavel Kopach
- Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland (I.G.L., V.L., N.W.T., P.K., S.P.A); and SAJE Pharma, Baltimore, Maryland (H.S.P.)
| | - Helen S Pentikis
- Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland (I.G.L., V.L., N.W.T., P.K., S.P.A); and SAJE Pharma, Baltimore, Maryland (H.S.P.)
| | - Sergei P Atamas
- Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland (I.G.L., V.L., N.W.T., P.K., S.P.A); and SAJE Pharma, Baltimore, Maryland (H.S.P.)
| |
Collapse
|
29
|
Ayano M, Tsukamoto H, Kohno K, Ueda N, Tanaka A, Mitoma H, Akahoshi M, Arinobu Y, Niiro H, Horiuchi T, Akashi K. Increased CD226 Expression on CD8+ T Cells Is Associated with Upregulated Cytokine Production and Endothelial Cell Injury in Patients with Systemic Sclerosis. THE JOURNAL OF IMMUNOLOGY 2015; 195:892-900. [PMID: 26109642 DOI: 10.4049/jimmunol.1403046] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/05/2014] [Accepted: 05/26/2015] [Indexed: 02/06/2023]
Abstract
Systemic sclerosis (SSc) is an autoimmune disease characterized by vascular damage and fibrosis of the skin and internal organs. Because activated and oligoclonally expanded CD8(+) T cells can be detected in peripheral blood and lungs of SSc patients, effector memory CD8(+) T cells may play a critical role for organ involvement in SSc; however, the pathogenic functions of effector memory CD8(+) T cells remain incompletely understood. In this study, we performed DNA microarray analysis of the sort-purified effector memory CD8(+) T cells from SSc patients and healthy controls, and showed that the expression of genes related to immune response and cell adhesion, including CD226 (also known as DNAX accessory molecule-1 [DNAM-1]), was significantly altered. Moreover, detailed analysis of CD226 revealed that CD226(high)CD8(+) T cells were increased in SSc patients (mean, 50.7%) compared with healthy controls (32.9%) and were appreciably associated with the severity of skin sclerosis and interstitial lung disease. Furthermore, CD226(+)CD8(+) T cells produced higher amount of various cytokines than CD226(-) ones, and CD226(high)CD8(+) T cells from SSc patients showed upregulated IL-13 production and positive correlation with the cytotoxic capacity of CD8(+) T cells against HUVECs. Finally, the neutralization of CD226 in CD8(+) T cells impaired costimulation, cytokine productions, and cytolysis against HUVECs. These findings indicate that upregulated CD226 expression on CD8(+) T cells reflects disease severity and is involved in SSc pathogenesis via the production of various cytokines, including profibrotic IL-13 and endothelial cell injury, and that CD226 may be a useful target in the treatment of SSc.
Collapse
Affiliation(s)
- Masahiro Ayano
- Department of Medicine and Biosystemic Science, Kyushu University Graduate School of Medical Sciences, Fukuoka 812-8582, Japan
| | - Hiroshi Tsukamoto
- Department of Medicine and Biosystemic Science, Kyushu University Graduate School of Medical Sciences, Fukuoka 812-8582, Japan;
| | - Kentaro Kohno
- Department of Hematology, National Kyushu Medical Center, Fukuoka 810-8563, Japan
| | - Naoyasu Ueda
- Department of Internal Medicine, Miyazaki Prefectural Miyazaki Hospital, Miyazaki 880-8510, Japan
| | - Atsushi Tanaka
- Department of Medicine and Biosystemic Science, Kyushu University Graduate School of Medical Sciences, Fukuoka 812-8582, Japan
| | - Hiroki Mitoma
- Department of Medicine and Biosystemic Science, Kyushu University Graduate School of Medical Sciences, Fukuoka 812-8582, Japan
| | - Mitsuteru Akahoshi
- Department of Medicine and Biosystemic Science, Kyushu University Graduate School of Medical Sciences, Fukuoka 812-8582, Japan
| | - Yojiro Arinobu
- Department of Medicine and Biosystemic Science, Kyushu University Graduate School of Medical Sciences, Fukuoka 812-8582, Japan
| | - Hiroaki Niiro
- Clinical Education Center, Kyushu University Hospital, Fukuoka 812-8582, Japan; and
| | - Takahiko Horiuchi
- Department of Internal Medicine, Kyushu University Beppu Hospital, Beppu 874-0836, Japan
| | - Koichi Akashi
- Department of Medicine and Biosystemic Science, Kyushu University Graduate School of Medical Sciences, Fukuoka 812-8582, Japan
| |
Collapse
|
30
|
Oncostatin M regulates osteogenic differentiation of murine adipose-derived mesenchymal progenitor cells through a PKCdelta-dependent mechanism. Cell Tissue Res 2015; 360:309-19. [DOI: 10.1007/s00441-014-2099-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2014] [Accepted: 12/18/2014] [Indexed: 10/24/2022]
|
31
|
|
32
|
Dumoitier N, Lofek S, Mouthon L. Pathophysiology of systemic sclerosis: state of the art in 2014. Presse Med 2014; 43:e267-78. [PMID: 25179277 DOI: 10.1016/j.lpm.2014.08.001] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2014] [Accepted: 08/05/2014] [Indexed: 02/06/2023] Open
Abstract
Major work has been done in order to improve the understanding of systemic sclerosis (SSc) pathogenesis. A number of new experimental models have been set up, that should help to understand the disease pathogenesis and test new therapeutic targets. Reactive oxygen species represent a hallmark of the pathogenesis of SSc, both at the fibroblast and at the endothelial cell levels. Although a large number of genetic studies have been conducted, it is still difficult to identify a genetic background specific to SSc, and the major progress in this setting is probably the identification of an interferon signature. Besides endothelial cells and fibroblasts, major development has been made in the understanding of the role of B cells and autoantibodies in the pathogenesis of SSc. Plasmacytoid dendritic cells seem to play a major role in the pathogenesis of SSc through the secretion of CXCL4, although these data will need to be confirmed in the near future.
Collapse
Affiliation(s)
- Nicolas Dumoitier
- Institut Cochin, Inserm U1016, CNRS UMR 8104, 8, rue Méchain, 75014 Paris, France; Université Paris Diderot, 75013 Paris, France
| | - Sébastien Lofek
- Institut Cochin, Inserm U1016, CNRS UMR 8104, 8, rue Méchain, 75014 Paris, France
| | - Luc Mouthon
- Institut Cochin, Inserm U1016, CNRS UMR 8104, 8, rue Méchain, 75014 Paris, France; Assistance publique-Hôpitaux de Paris (AP-HP), hôpital Cochin, centre de référence pour les vascularites nécrosantes et la sclérodermie systémique, service de médecine interne, Université Paris Descartes, 75014 Paris, France.
| |
Collapse
|
33
|
Shiomi A, Usui T, Ishikawa Y, Shimizu M, Murakami K, Mimori T. GM-CSF but not IL-17 is critical for the development of severe interstitial lung disease in SKG mice. THE JOURNAL OF IMMUNOLOGY 2014; 193:849-59. [PMID: 24951817 DOI: 10.4049/jimmunol.1303255] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Interstitial lung disease (ILD) is a common complication and sometimes a prognostic factor of connective tissue diseases (CTDs) in humans. However, suitable animal model of severe CTD-associated ILD (CTD-ILD) has been limited. In this study, we showed that zymosan-treated SKG mice developed not only arthritis but also chronic-progressive ILD with high mortality over several months. The pathological and clinical features of ILD in zymosan-treated SKG mice were similar to that of human severe CTD-ILD. ILD in this mouse was characterized by massive infiltration of Th17 cells, GM-CSF-producing CD4(+) T cells, and CD11b(+) Gr1(+) neutrophils with fibrosis. Naive SKG T cells were skewed to differentiate into GM-CSF-producing cells, and GM-CSF secreted by T cells enhanced IL-6 and IL-1β production by macrophages, which in turn enhanced differentiation of IL-17A- and/or GM-CSF-producing T cells and infiltration of neutrophils into lung. Neutralization of GM-CSF completely blocked the development of this ILD, and the blocking of IL-6 signaling resulted in partial prevention of it, whereas neutralization of IL-17A did not. In contrast, the progression of arthritis was inhibited by the neutralization of GM-CSF and slightly by the neutralization of IL-17A, but not by the blocking of IL-6 signaling. These data suggested zymosan-treated SKG mice could be a useful mouse model of severe CTD-ILD, and GM-CSF, rather than IL-17A or IL-6, contributed to the development of ILD in zymosan-treated SKG mice, indicating that neutralization of GM-CSF would be a useful therapeutic strategy for severe CTD-ILD.
Collapse
Affiliation(s)
- Aoi Shiomi
- Department of Rheumatology and Clinical Immunology, Graduate School of Medicine, Kyoto University, Kyoto 606-8507, Japan
| | - Takashi Usui
- Department of Rheumatology and Clinical Immunology, Graduate School of Medicine, Kyoto University, Kyoto 606-8507, Japan
| | - Yuki Ishikawa
- Department of Rheumatology and Clinical Immunology, Graduate School of Medicine, Kyoto University, Kyoto 606-8507, Japan
| | - Masakazu Shimizu
- Department of Rheumatology and Clinical Immunology, Graduate School of Medicine, Kyoto University, Kyoto 606-8507, Japan
| | - Kosaku Murakami
- Department of Rheumatology and Clinical Immunology, Graduate School of Medicine, Kyoto University, Kyoto 606-8507, Japan
| | - Tsuneyo Mimori
- Department of Rheumatology and Clinical Immunology, Graduate School of Medicine, Kyoto University, Kyoto 606-8507, Japan
| |
Collapse
|
34
|
Luzina IG, Kopach P, Lockatell V, Kang PH, Nagarsekar A, Burke AP, Hasday JD, Todd NW, Atamas SP. Interleukin-33 potentiates bleomycin-induced lung injury. Am J Respir Cell Mol Biol 2014; 49:999-1008. [PMID: 23837438 DOI: 10.1165/rcmb.2013-0093oc] [Citation(s) in RCA: 101] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
The mechanisms of interstitial lung disease (ILD) remain incompletely understood, although recent observations have suggested an important contribution by IL-33. Substantial elevations in IL-33 expression were found in the lungs of patients with idiopathic pulmonary fibrosis and scleroderma lung disease, as well as in the bleomycin injury mouse model. Most of the observed IL-33 expression was intracellular and intranuclear, suggesting involvement of the full-length (fl) protein, but not of the proteolytically processed mature IL-33 cytokine. The effects of flIL-33 on mouse lungs were assessed independently and in combination with bleomycin injury, using recombinant adenovirus-mediated gene delivery. Bleomycin-induced changes were not affected by gene deficiency of the IL-33 receptor T1/ST2. Combined flIL-33 expression and bleomycin injury exerted a synergistic effect on pulmonary lymphocyte and collagen accumulation, which could be explained by synergistic regulation of the cytokines transforming growth factor-β, IL-6, monocyte chemotactic protein-1, macrophage inflammatory protein\x{2013}1α, and tumor necrosis factor-α. By contrast, no increase in the levels of the Th2 cytokines IL-4, IL-5, or IL-13 was evident. Moreover, flIL-33 was found to increase the expression of several heat shock proteins (HSPs) significantly, and in particular HSP70, which is known to be associated with ILD. Thus, flIL-33 is a synergistic proinflammatory and profibrotic regulator that acts by stimulating the expression of several non-Th2 cytokines, and activates the expression of HSP70.
Collapse
Affiliation(s)
- Irina G Luzina
- 1 Division of Rheumatology and Clinical Immunology, Department of Medicine, University of Maryland School of Medicine, Baltimore; and
| | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Richards CD. The enigmatic cytokine oncostatin m and roles in disease. ISRN INFLAMMATION 2013; 2013:512103. [PMID: 24381786 PMCID: PMC3870656 DOI: 10.1155/2013/512103] [Citation(s) in RCA: 154] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/03/2013] [Accepted: 09/29/2013] [Indexed: 12/11/2022]
Abstract
Oncostatin M is a secreted cytokine involved in homeostasis and in diseases involving chronic inflammation. It is a member of the gp130 family of cytokines that have pleiotropic functions in differentiation, cell proliferation, and hematopoetic, immunologic, and inflammatory networks. However, Oncostatin M also has activities novel to mediators of this cytokine family and others and may have fundamental roles in mechanisms of inflammation in pathology. Studies have explored Oncostatin M functions in cancer, bone metabolism, liver regeneration, and conditions with chronic inflammation including rheumatoid arthritis, lung and skin inflammatory disease, atherosclerosis, and cardiovascular disease. This paper will review Oncostatin M biology in a historical fashion and focus on its unique activities, in vitro and in vivo, that differentiate it from other cytokines and inspire further study or consideration in therapeutic approaches.
Collapse
Affiliation(s)
- Carl D. Richards
- McMaster Immunology Research Centre, Department of Pathology and Molecular Medicine, McMaster University, 1280 Main Street, West, Hamilton, ON, Canada L8S 4K1
| |
Collapse
|
36
|
Funk SD, Orr AW. Ephs and ephrins resurface in inflammation, immunity, and atherosclerosis. Pharmacol Res 2013; 67:42-52. [DOI: 10.1016/j.phrs.2012.10.008] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2012] [Revised: 10/04/2012] [Accepted: 10/10/2012] [Indexed: 01/13/2023]
|
37
|
Mikami Y, Yamauchi Y, Horie M, Kase M, Jo T, Takizawa H, Kohyama T, Nagase T. Tumor necrosis factor superfamily member LIGHT induces epithelial-mesenchymal transition in A549 human alveolar epithelial cells. Biochem Biophys Res Commun 2012; 428:451-7. [PMID: 23131560 DOI: 10.1016/j.bbrc.2012.10.097] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2012] [Accepted: 10/12/2012] [Indexed: 02/06/2023]
Abstract
Fibrosis is an abnormal response to organ injury, characterized by accumulation of activated fibroblasts at the sites of injury. Fibroblasts arise from several sources, including resident fibroblasts and circulating fibrocytes that infiltrate organ tissue. Recently, epithelial-mesenchymal transition (EMT) has been recognized as a source of mesenchymal cells. EMT is induced by various growth factors, such as transforming growth factor (TGF)-β1, and enhanced by inflammatory cytokines. Recently the tumor necrosis factor superfamily member LIGHT has been implicated in the pathogenesis of inflammatory disease and airway remodeling in severe asthma. We hypothesized that LIGHT might contribute to the pathogenesis of airway fibrosis via enhancement of EMT. Therefore, we investigated LIGHT's ability to induce EMT. A549 cells were stimulated with LIGHT, TGF-β1 or both for 48h. To estimate EMT, we evaluated the expression of epithelial and mesenchymal markers using immunocytochemistry, Western blotting and quantitative RT-PCR. Signaling pathways for EMT were characterized by Western analysis to detect phosphorylation of Erk1/2 and smad2. LIGHT enhanced TGF-β1-induced EMT both morphologically, by suppressing E-cadherin and enhancing vimentin, and functionally, by enhancing cell contractility. Additionally, LIGHT induced EMT without TGF-β1. Evaluation of the mechanism showed that LIGHT did not induce TGF-β1 production or affect the smad-snai1 pathway. Inhibition of Erk1/2 phosphorylation reduced LIGHT-induced EMT, indicating the Erk1/2 pathway to be a key pathway in LIGHT-induced EMT. In summary, LIGHT enhanced TGF-β1-induced EMT but also induced EMT via the Erk1/2 pathway by itself, without TGF-β1 signaling. LIGHT may contribute to the pathogenesis of airway fibrosis through enhancement of EMT.
Collapse
Affiliation(s)
- Yu Mikami
- Department of Respiratory Medicine, Graduate School of Medicine, The University of Tokyo, Japan
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Todd NW, Luzina IG, Atamas SP. Molecular and cellular mechanisms of pulmonary fibrosis. FIBROGENESIS & TISSUE REPAIR 2012; 5:11. [PMID: 22824096 PMCID: PMC3443459 DOI: 10.1186/1755-1536-5-11] [Citation(s) in RCA: 309] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/02/2012] [Accepted: 06/28/2012] [Indexed: 12/22/2022]
Abstract
Pulmonary fibrosis is a chronic lung disease characterized by excessive accumulation of extracellular matrix (ECM) and remodeling of the lung architecture. Idiopathic pulmonary fibrosis is considered the most common and severe form of the disease, with a median survival of approximately three years and no proven effective therapy. Despite the fact that effective treatments are absent and the precise mechanisms that drive fibrosis in most patients remain incompletely understood, an extensive body of scientific literature regarding pulmonary fibrosis has accumulated over the past 35 years. In this review, we discuss three broad areas which have been explored that may be responsible for the combination of altered lung fibroblasts, loss of alveolar epithelial cells, and excessive accumulation of ECM: inflammation and immune mechanisms, oxidative stress and oxidative signaling, and procoagulant mechanisms. We discuss each of these processes separately to facilitate clarity, but certainly significant interplay will occur amongst these pathways in patients with this disease.
Collapse
Affiliation(s)
- Nevins W Todd
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA.
| | | | | |
Collapse
|
39
|
Luzina IG, Keegan AD, Heller NM, Rook GAW, Shea-Donohue T, Atamas SP. Regulation of inflammation by interleukin-4: a review of "alternatives". J Leukoc Biol 2012; 92:753-64. [PMID: 22782966 DOI: 10.1189/jlb.0412214] [Citation(s) in RCA: 258] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Studies of IL-4 have revealed a wealth of information on the diverse roles of this cytokine in homeostatic regulation and disease pathogenesis. Recent data suggest that instead of simple linear regulatory pathways, IL-4 drives regulation that is full of alternatives. In addition to the well-known dichotomous regulation of Th cell differentiation by IL-4, this cytokine is engaged in several other alternative pathways. Its own production involves alternative mRNA splicing, yielding at least two functional isoforms: full-length IL-4, encoded by the IL-4 gene exons 1-4, and IL-4δ2, encoded by exons 1, 3, and 4. The functional effects of these two isoforms are in some ways similar but in other ways quite distinct. When binding to the surface of target cells, IL-4 may differentially engage two different types of receptors. By acting on macrophages, a cell type critically involved in inflammation, IL-4 induces the so-called alternative macrophage activation. In this review, recent advances in understanding these three IL-4-related branch points--alternative splicing of IL-4, differential receptor engagement by IL-4, and differential regulation of macrophage activation by IL-4--are summarized in light of their contributions to inflammation.
Collapse
Affiliation(s)
- Irina G Luzina
- University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | | | | | | | | | | |
Collapse
|
40
|
O'Reilly S, Hügle T, van Laar JM. T cells in systemic sclerosis: a reappraisal. Rheumatology (Oxford) 2012; 51:1540-9. [PMID: 22577083 DOI: 10.1093/rheumatology/kes090] [Citation(s) in RCA: 170] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
SSc is an autoimmune disease characterized by inflammation and extracellular matrix deposition that ultimately leads to loss of organ function. T cells appear to play a prominent role in its pathogenesis. The evidence for this comes from their being at the site of fibrosis, their activated phenotype and alteration in their number and frequency in peripheral blood. This review examines the role of T cells in the pathogenesis of SSc and specifically examines the key soluble profibrotic mediators (IL-4, IL-6, IL-13) secreted by Th2 cells and their interactions with fibroblasts that deposit excess extracellular matrix leading to fibrosis. We finally examine possible therapeutic options in targeting T-cell mediators to disrupt the cellular interactions between T cells and fibroblasts that serve to drive the fibrotic response. One of the factors driving fibrosis is IL-6 and this can be neutralized in vivo not only to limit IL-6-driven tissue fibrosis but concomitantly to suppress switching of Tregs to Th17 T cells that will provide more IL-6, thus perpetuating the fibrosis. Taken together, these data implicate the role of T cells in SSc and suggest that Th2-polarized T cells and the fibrotic mediators subsequently released directly induce fibrosis. Targeting such cytokines may be therapeutic not only in SSc but more generally in diseases where fibrosis is directed by inflammatory signals.
Collapse
Affiliation(s)
- Steven O'Reilly
- Musculoskeletal Research Group, Institute of Cellular Medicine, Newcastle University, Framlington Place, Newcastle upon Tyne, UK. steven.o'
| | | | | |
Collapse
|
41
|
Cho JY. Recent advances in mechanisms and treatments of airway remodeling in asthma: a message from the bench side to the clinic. Korean J Intern Med 2011; 26:367-83. [PMID: 22205837 PMCID: PMC3245385 DOI: 10.3904/kjim.2011.26.4.367] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2011] [Accepted: 09/15/2011] [Indexed: 12/31/2022] Open
Abstract
Airway remodeling in asthma is a result of persistent inflammation and epithelial damage in response to repetitive injury. Recent studies have identified several important mediators associated with airway remodeling in asthma, including transforming growth factor-β, interleukin (IL)-5, basic fibroblast growth factor, vascular endothelial growth factor, LIGHT, tumor necrosis factor (TNF)-α, thymic stromal lymphopoietin, IL-33, and IL-25. In addition, the epithelium mesenchymal transformation (EMT) induced by environmental factors may play an important role in initiating this process. Diagnostic methods using sputum and blood biomarkers as well as radiological interventions have been developed to distinguish between asthma sub-phenotypes. Human clinical trials have been conducted to evaluate biological therapies that target individual inflammatory cells or mediators including anti IgE, anti IL-5, and anti TNF-α. Furthermore, new drugs such as c-kit/platelet-derived growth factor receptor kinase inhibitors, endothelin-1 receptor antagonists, calcium channel inhibitors, and HMG-CoA reductase inhibitors have been developed to treat asthma-related symptoms. In addition to targeting specific inflammatory cells or mediators, preventing the initiation of EMT may be important for targeted treatment. Interestingly, bronchial thermoplasty reduces smooth muscle mass in patients with severe asthma and improves asthma-specific quality of life, particularly by reducing severe exacerbation and healthcare use. A wide range of different therapeutic approaches has been developed to address the immunological processes of asthma and to treat this complex chronic illness. An important future direction may be to investigate the role of mediators involved in the development of airway remodeling to enhance asthma therapy.
Collapse
Affiliation(s)
- Jae Youn Cho
- Division of Allergy/Immunology, University of California San Diego School of Medicine, La Jolla, CA 92093-0635, USA.
| |
Collapse
|
42
|
Atamas SP, Fontenot AP. Regulatory T Cells and Lung Fibrosis: A Good Cell Gone Bad. Am J Respir Crit Care Med 2011; 184:1224-6. [DOI: 10.1164/rccm.201108-1572ed] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
|
43
|
Sargent JL, Whitfield ML. Capturing the heterogeneity in systemic sclerosis with genome-wide expression profiling. Expert Rev Clin Immunol 2011; 7:463-73. [PMID: 21790289 DOI: 10.1586/eci.11.41] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Heterogeneity in the clinical presentation and basic science findings of systemic sclerosis (SSc) has hindered the understanding of pathogenesis and development of effective treatments. Genome-wide profiling of SSc has measured this heterogeneity. Gene expression studies of diffuse SSc skin have shown reproducible, disease-specific gene expression signatures when compared with healthy controls and, surprisingly, disease-specific gene expression was found in both lesional and non-lesional skin. SSc-specific gene expression in peripheral blood cells and the lungs has also been demonstrated. Hypothesis-driven approaches that assess the contribution of individual pathways provide insight into the etiology of gene expression subsets.
Collapse
Affiliation(s)
- Jennifer L Sargent
- Department of Genetics, Dartmouth Medical School, Hanover, NH 03755, USA
| | | |
Collapse
|
44
|
Michels-van Amelsfort JMR, Walter GJ, Taams LS. CD4+CD25+ regulatory T cells in systemic sclerosis and other rheumatic diseases. Expert Rev Clin Immunol 2011; 7:499-514. [PMID: 21790293 DOI: 10.1586/eci.11.28] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Systemic sclerosis (SSc) is a generalized connective tissue disorder, characterized by a wide spectrum of microvascular and immunological abnormalities, leading to a progressive thickening and fibrosis of the skin and other organs, such as the lungs, GI tract, heart and kidneys. SSc is thought to be an autoimmune disease owing to the presence of high affinity antibodies and possible clinical overlap with other autoimmune diseases such as systemic lupus erythematosus and rheumatoid arthritis. Autoimmune diseases arise because of a breakdown in immunological self tolerance. Self tolerance is maintained via multiple regulatory mechanisms within the immune system, including the thymic deletion of self-reactive T cells and mechanisms of peripheral tolerance. In recent years, the presence of CD4(+)CD25(+)FOXP3(+) Tregs has been identified as a major mechanism of peripheral tolerance, and accumulating evidence indicates that alterations in Treg frequencies and/or function may contribute to autoimmune diseases. Here, we will review recent data on the percentage, function and phenotype of CD4(+)CD25(+) Tregs in rheumatic disease, and discuss how recent developments may guide research in this area in SSc.
Collapse
|
45
|
Hoermann G, Cerny-Reiterer S, Herrmann H, Blatt K, Bilban M, Gisslinger H, Gisslinger B, Müllauer L, Kralovics R, Mannhalter C, Valent P, Mayerhofer M. Identification of oncostatin M as a JAK2 V617F-dependent amplifier of cytokine production and bone marrow remodeling in myeloproliferative neoplasms. FASEB J 2011; 26:894-906. [PMID: 22051730 DOI: 10.1096/fj.11-193078] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
The JAK2 mutation V617F is detectable in a majority of patients with Philadelphia chromosome-negative myeloproliferative neoplasms (MPNs). Enforced expression of JAK2 V617F in mice induces myeloproliferation and bone marrow (BM) fibrosis, suggesting a causal role for the JAK2 mutant in the pathogenesis of MPNs. However, little is known about mechanisms and effector molecules contributing to JAK2 V617F-induced myeloproliferation and fibrosis. We show that JAK2 V617F promotes expression of oncostatin M (OSM) in neoplastic myeloid cells. Correspondingly, OSM mRNA levels were increased in the BM of patients with MPNs (median 287% of ABL, range 22-1450%) compared to control patients (median 59% of ABL, range 12-264%; P < 0.0001). OSM secreted by JAK2 V617F+ cells stimulated growth of fibroblasts and microvascular endothelial cells and induced the production of angiogenic and profibrogenic cytokines (HGF, VEGF, and SDF-1) in BM fibroblasts. All effects of MPN cell-derived OSM were blocked by a neutralizing anti-OSM antibody, whereas the production of OSM in MPN cells was suppressed by a pharmacologic JAK2 inhibitor or RNAi-mediated knockdown of JAK2. In summary, JAK2 V617F-mediated up-regulation of OSM may contribute to fibrosis, neoangiogenesis, and the cytokine storm observed in MPNs, suggesting that OSM might serve as a novel therapeutic target molecule in these neoplasms.
Collapse
Affiliation(s)
- Gregor Hoermann
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Abstract
Systemic sclerosis (SSc) is a progressive and highly debilitating autoimmune disorder characterized by inflammation, fibrosis, and vascular damage of the connective tissue. T cell-derived cytokines have been implicated in the induction of fibrosis. We found that high levels of the profibrotic type-2 cytokine IL-13 are produced by peripheral blood effector CD8(+) T cells from SSc patients compared to normal controls. This abnormality correlates with increased expression of the transcription factor GATA-3 and the extent of skin fibrosis. Together, the data provide new insights into SSc pathogenesis and identify a specific T cell phenotype that can be used as a biomarker of immune dysfunction in patients with SSc and as a novel therapeutic target for this currently incurable disease.
Collapse
Affiliation(s)
- Patrizia Fuschiotti
- Department of Immunology, University of Pittsburgh School of Medicine, 200 Lothrop Street, BST W1052, Pittsburgh, PA 15261, USA.
| |
Collapse
|
47
|
Maddigan A, Truitt L, Arsenault R, Freywald T, Allonby O, Dean J, Narendran A, Xiang J, Weng A, Napper S, Freywald A. EphB receptors trigger Akt activation and suppress Fas receptor-induced apoptosis in malignant T lymphocytes. THE JOURNAL OF IMMUNOLOGY 2011; 187:5983-94. [PMID: 22039307 DOI: 10.4049/jimmunol.1003482] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Treatment of hematopoietic malignancies often requires allogeneic bone marrow transplantation, and the subsequent graft-versus-leukemia response is crucial for the elimination of malignant cells. Cytotoxic T lymphocytes and NK cells responsible for the immunoelimination express Fas ligand and strongly rely on the induction of Fas receptor-mediated apoptosis for their action. Although cancer cells are removed successfully by graft-versus-leukemia reactions in myeloid malignancies, their efficiency is low in T cell leukemias. This may be partially because of the ability of malignant T cells to escape apoptosis. Our work shows that Eph family receptor EphB3 is consistently expressed by malignant T lymphocytes, most frequently in combination with EphB6, and that stimulation with their common ligands, ephrin-B1 and ephrin-B2, strongly suppresses Fas-induced apoptosis in these cells. This effect is associated with Akt activation and with the inhibition of the Fas receptor-initiated caspase proteolytic cascade. Akt proved to be crucial for the prosurvival response, because inhibition of Akt, but not of other molecules central to T cell biology, including Src kinases, MEK1 and MEK2, blocked the antiapoptotic effect. Overall, this demonstrates a new role for EphB receptors in the protection of malignant T cells from Fas-induced apoptosis through Akt engagement and prevention of caspase activation. Because Fas-triggered apoptosis is actively involved in the graft-versus-leukemia response and cytotoxic T cells express ephrin-Bs, our observations suggest that EphB receptors are likely to support immunoevasivenes of T cell malignancies and may represent promising targets for therapies, aiming to enhance immunoelimination of cancerous T cells.
Collapse
Affiliation(s)
- Alison Maddigan
- Department of Chemistry and Biochemistry, University of Regina, Regina, Saskatchewan S4S 0A2, Canada
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Farkas L, Gauldie J, Voelkel NF, Kolb M. Pulmonary Hypertension and Idiopathic Pulmonary Fibrosis. Am J Respir Cell Mol Biol 2011; 45:1-15. [DOI: 10.1165/rcmb.2010-0365tr] [Citation(s) in RCA: 149] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
|
49
|
The tumor necrosis factor family member LIGHT is a target for asthmatic airway remodeling. Nat Med 2011; 17:596-603. [PMID: 21499267 DOI: 10.1038/nm.2356] [Citation(s) in RCA: 143] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2010] [Accepted: 03/16/2011] [Indexed: 01/10/2023]
Abstract
Individuals with chronic asthma show a progressive decline in lung function that is thought to be due to structural remodeling of the airways characterized by subepithelial fibrosis and smooth muscle hyperplasia. Here we show that the tumor necrosis factor (TNF) family member LIGHT is expressed on lung inflammatory cells after allergen exposure. Pharmacological inhibition of LIGHT using a fusion protein between the IgG Fc domain and lymphotoxin β receptor (LTβR) reduces lung fibrosis, smooth muscle hyperplasia and airway hyperresponsiveness in mouse models of chronic asthma, despite having little effect on airway eosinophilia. LIGHT-deficient mice also show a similar impairment in fibrosis and smooth muscle accumulation. Blockade of LIGHT suppresses expression of lung transforming growth factor-β (TGF-β) and interleukin-13 (IL-13), cytokines implicated in remodeling in humans, whereas exogenous administration of LIGHT to the airways induces fibrosis and smooth muscle hyperplasia, Thus, LIGHT may be targeted to prevent asthma-related airway remodeling.
Collapse
|
50
|
Avouac J, Clemessy M, Distler JH, Gasc JM, Ruiz B, Vacher-Lavenu MC, Wipff J, Kahan A, Boileau C, Corvol P, Allanore Y. Enhanced expression of ephrins and thrombospondins in the dermis of patients with early diffuse systemic sclerosis: potential contribution to perturbed angiogenesis and fibrosis. Rheumatology (Oxford) 2011; 50:1494-504. [DOI: 10.1093/rheumatology/keq448] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
|