1
|
Howe CS, Chulkina M, Syrcle R, McAninch C, McAninch S, Pinchuk IV, Beswick EJ. MK2 Inhibition in CD4+ T Cells Protects Against IFNγ and IL-17A, Chronic Inflammation, and Fibrosis in Inflammatory Bowel Disease Models. Inflamm Bowel Dis 2025:izaf026. [PMID: 39937137 DOI: 10.1093/ibd/izaf026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Indexed: 02/13/2025]
Abstract
BACKGROUND CD4+ T cells contribute to chronic inflammation and fibrosis in inflammatory bowel disease (IBD), but the cellular mechanisms remain elusive. We have found that the mitogen-activated protein kinase 2 (MK2) pathway plays a major role in inflammation and overall pathology in IBD. Thus, here, we examined the role of MK2 in regulating CD4+ T cell responses in IBD models. METHODS Interleukin-10 (IL-10) knockout (KO) mice treated with MK2 inhibitors (MK2i) and CD4-specific MK2 knockdown mice treated with chronic dextran sodium sulfate (DSS) treatments were used to examine inflammation and fibrosis by multiplex array, gene expression, flow cytometry, and histology. Human tissues were treated with MK2i to examine Th1 and Th17 markers. RESULTS IL-10 KO mice treated with MK2i therapeutically showed significantly reduced interferon gamma (IFNγ) and interleukin-17A (IL-17A) and a significantly reduced number of IFNγ+ and IL-17A+ producing CD4+ T cells by flow cytometry. To investigate the direct role of MK2 in CD4+ T cells during IBD, we utilized CD4-specific MK2 knockdown mice in chronic DSS colitis. A decrease in colonic inflammation, IFNγ and IL-17, pro-fibrotic genes, and extracellular matrix deposition was observed in mice with MK2 knockdown in CD4+ T cells compared to control mice. Additionally, IL-17A and IFNγ directly regulated the expression of fibrosis genes in colon tissues. CONCLUSIONS The MK2 pathway regulates inflammatory CD4+ T cells and fibrosis in IBD models and is a potential therapeutic target.
Collapse
Affiliation(s)
- Cody S Howe
- Department of Internal Medicine, University of Kentucky, Lexington, KY, USA
| | - Marina Chulkina
- Department of Medicine, Penn State College of Medicine, Hershey, PA, USA
| | - Ryan Syrcle
- Department of Internal Medicine, University of Kentucky, Lexington, KY, USA
| | - Christina McAninch
- Department of Medicine, Penn State College of Medicine, Hershey, PA, USA
| | - Steven McAninch
- Department of Medicine, Penn State College of Medicine, Hershey, PA, USA
| | - Irina V Pinchuk
- Department of Medicine, Penn State College of Medicine, Hershey, PA, USA
| | - Ellen J Beswick
- Department of Internal Medicine, University of Kentucky, Lexington, KY, USA
| |
Collapse
|
2
|
Ko JH, Kim S, Ryu JS, Song HJ, Oh JY. Interferon-γ elicits the ocular surface pathology mimicking dry eye through direct modulation of resident corneal cells. Cell Death Discov 2023; 9:209. [PMID: 37391421 DOI: 10.1038/s41420-023-01511-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 06/15/2023] [Accepted: 06/20/2023] [Indexed: 07/02/2023] Open
Abstract
Despite accumulating evidence indicating a key role of interferon-γ (IFN-γ)-producing immune cells in ocular infection and immunity, little is known about the direct effects of IFN-γ on resident corneal cells or on the ocular surface. Here, we report that IFN-γ impacts corneal stromal fibroblasts and epithelial cells to promote inflammation, opacification, and barrier disruption on the ocular surface, leading to dry eye. Our results demonstrated that IFN-γ dose-dependently induced cytotoxicity, pro-inflammatory cytokine/chemokine production, and expression of major histocompatibility complex class II and CD40 in cultures of corneal stromal fibroblasts and epithelial cells while increasing myofibroblast differentiation of corneal stromal fibroblasts. In mice, subconjunctival IFN-γ administration caused corneal epithelial defects and stromal opacity in dose- and time-dependent manners while promoting neutrophil infiltration and inflammatory cytokine expression in the cornea. Moreover, IFN-γ reduced aqueous tear secretion and the number of conjunctival goblet cells responsible for mucinous tear production. Together, our findings suggest that IFN-γ induces the ocular surface changes characteristic of dry eye disease at least in part through its direct effects on resident corneal cells.
Collapse
Affiliation(s)
- Jung Hwa Ko
- Laboratory of Ocular Regenerative Medicine and Immunology, Biomedical Research Institute, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Korea
| | - Seonghwan Kim
- Laboratory of Ocular Regenerative Medicine and Immunology, Biomedical Research Institute, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Korea
- Department of Ophthalmology, Seoul Metropolitan Government Seoul National University Boramae Medical Center, 20 Boramae-ro 5 Gil, Dongjak-gu, Seoul, 07061, Korea
| | - Jin Suk Ryu
- Laboratory of Ocular Regenerative Medicine and Immunology, Biomedical Research Institute, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Korea
| | - Hyo Jeong Song
- Laboratory of Ocular Regenerative Medicine and Immunology, Biomedical Research Institute, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Korea
| | - Joo Youn Oh
- Laboratory of Ocular Regenerative Medicine and Immunology, Biomedical Research Institute, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Korea.
- Department of Ophthalmology, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul, 03080, Korea.
| |
Collapse
|
3
|
Esnault S, Jarjour NN. Development of Adaptive Immunity and Its Role in Lung Remodeling. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1426:287-351. [PMID: 37464127 DOI: 10.1007/978-3-031-32259-4_14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/20/2023]
Abstract
Asthma is characterized by airflow limitations resulting from bronchial closure, which can be either reversible or fixed due to changes in airway tissue composition and structure, also known as remodeling. Airway remodeling is defined as increased presence of mucins-producing epithelial cells, increased thickness of airway smooth muscle cells, angiogenesis, increased number and activation state of fibroblasts, and extracellular matrix (ECM) deposition. Airway inflammation is believed to be the main cause of the development of airway remodeling in asthma. In this chapter, we will review the development of the adaptive immune response and the impact of its mediators and cells on the elements defining airway remodeling in asthma.
Collapse
|
4
|
Lin SN, Mao R, Qian C, Bettenworth D, Wang J, Li J, Bruining D, Jairath V, Feagan B, Chen M, Rieder F. Development of Anti-fibrotic Therapy in Stricturing Crohn's Disease: Lessons from Randomized Trials in Other Fibrotic Diseases. Physiol Rev 2021; 102:605-652. [PMID: 34569264 DOI: 10.1152/physrev.00005.2021] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Intestinal fibrosis is considered an inevitable complication of Crohn's disease (CD) that results in symptoms of obstruction and stricture formation. Endoscopic or surgical treatment is required to treat the majority of patients. Progress in the management of stricturing CD is hampered by the lack of effective anti-fibrotic therapy; however, this situation is likely to change because of recent advances in other fibrotic diseases of the lung, liver and skin. In this review, we summarized data from randomized controlled trials (RCT) of anti-fibrotic therapies in these conditions. Multiple compounds have been tested for the anti-fibrotic effects in other organs. According to their mechanisms, they were categorized into growth factor modulators, inflammation modulators, 5-hydroxy-3-methylgultaryl-coenzyme A (HMG-CoA) reductase inhibitors, intracellular enzymes and kinases, renin-angiotensin system (RAS) modulators and others. From our review of the results from the clinical trials and discussion of their implications in the gastrointestinal tract, we have identified several molecular candidates that could serve as potential therapies for intestinal fibrosis in CD.
Collapse
Affiliation(s)
- Si-Nan Lin
- Department of Gastroenterology and Hepatology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.,Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, United States.,Department of Gastroenterology, Hepatology and Nutrition, Digestive Disease Institute, Cleveland Clinic, Cleveland, Ohio, United States
| | - Ren Mao
- Department of Gastroenterology and Hepatology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.,Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, United States.,Department of Gastroenterology, Hepatology and Nutrition, Digestive Disease Institute, Cleveland Clinic, Cleveland, Ohio, United States
| | - Chenchen Qian
- Department of Internal Medicine, UPMC Pinnacle, Harrisburg, Pennsylvania, United States
| | - Dominik Bettenworth
- Department of Medicine B, Gastroenterology and Hepatology, University Hospital Münster, Münster, Germany
| | - Jie Wang
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, United States.,Department of Gastroenterology, Hepatology and Nutrition, Digestive Disease Institute, Cleveland Clinic, Cleveland, Ohio, United States.,Henan Key Laboratory of Immunology and Targeted Drug, Xinxiang Medical University, Xinxiang, Henan Province, China
| | - Jiannan Li
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, United States.,Department of Gastroenterology, Hepatology and Nutrition, Digestive Disease Institute, Cleveland Clinic, Cleveland, Ohio, United States
| | - David Bruining
- Division of Gastroenterology and Hepatology, Mayo Clinic College of Medicine, Rochester, Minnesota, United States
| | - Vipul Jairath
- Alimentiv Inc., London, ON, Canada.,Department of Medicine, Western University, London, ON, Canada.,Department of Biostatistics and Epidemiology, Western University, London, ON, Canada
| | - Brian Feagan
- Alimentiv Inc., London, ON, Canada.,Department of Medicine, Western University, London, ON, Canada.,Department of Biostatistics and Epidemiology, Western University, London, ON, Canada
| | - Minhu Chen
- Department of Gastroenterology and Hepatology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | | | - Florian Rieder
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, United States.,Department of Gastroenterology, Hepatology and Nutrition, Digestive Disease Institute, Cleveland Clinic, Cleveland, Ohio, United States
| |
Collapse
|
5
|
Lázaro-Escudero JA, Chu EY. SnapshotDx Quiz: July 2021. J Invest Dermatol 2021. [DOI: 10.1016/j.jid.2021.05.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
6
|
IFN-γ–Stimulated Apoptotic Keratinocytes Promote Sclerodermatous Changes in Chronic Graft-Versus-Host Disease. J Invest Dermatol 2021; 141:1473-1481.e4. [DOI: 10.1016/j.jid.2020.09.033] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 08/22/2020] [Accepted: 09/14/2020] [Indexed: 01/16/2023]
|
7
|
Liu G, Philp AM, Corte T, Travis MA, Schilter H, Hansbro NG, Burns CJ, Eapen MS, Sohal SS, Burgess JK, Hansbro PM. Therapeutic targets in lung tissue remodelling and fibrosis. Pharmacol Ther 2021; 225:107839. [PMID: 33774068 DOI: 10.1016/j.pharmthera.2021.107839] [Citation(s) in RCA: 148] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Accepted: 03/03/2021] [Indexed: 02/07/2023]
Abstract
Structural changes involving tissue remodelling and fibrosis are major features of many pulmonary diseases, including asthma, chronic obstructive pulmonary disease (COPD) and idiopathic pulmonary fibrosis (IPF). Abnormal deposition of extracellular matrix (ECM) proteins is a key factor in the development of tissue remodelling that results in symptoms and impaired lung function in these diseases. Tissue remodelling in the lungs is complex and differs between compartments. Some pathways are common but tissue remodelling around the airways and in the parenchyma have different morphologies. Hence it is critical to evaluate both common fibrotic pathways and those that are specific to different compartments; thereby expanding the understanding of the pathogenesis of fibrosis and remodelling in the airways and parenchyma in asthma, COPD and IPF with a view to developing therapeutic strategies for each. Here we review the current understanding of remodelling features and underlying mechanisms in these major respiratory diseases. The differences and similarities of remodelling are used to highlight potential common therapeutic targets and strategies. One central pathway in remodelling processes involves transforming growth factor (TGF)-β induced fibroblast activation and myofibroblast differentiation that increases ECM production. The current treatments and clinical trials targeting remodelling are described, as well as potential future directions. These endeavours are indicative of the renewed effort and optimism for drug discovery targeting tissue remodelling and fibrosis.
Collapse
Affiliation(s)
- Gang Liu
- Centre for Inflammation, Centenary Institute and University of Technology Sydney, Sydney, NSW, Australia
| | - Ashleigh M Philp
- Centre for Inflammation, Centenary Institute and University of Technology Sydney, Sydney, NSW, Australia; St Vincent's Medical School, UNSW Medicine, UNSW, Sydney, NSW, Australia
| | - Tamera Corte
- Royal Prince Alfred Hospital, Camperdown, NSW, Australia; Sydney Medical School, University of Sydney, Sydney, NSW, Australia
| | - Mark A Travis
- The Lydia Becker Institute of Immunology and Inflammation, Faculty of Biology, Medicine and Health, Manchester Academic Health Sciences Centre and Wellcome Trust Centre for Cell-Matrix Research, University of Manchester, Manchester, United Kingdom
| | - Heidi Schilter
- Pharmaxis Ltd, 20 Rodborough Road, Frenchs Forest, Sydney, NSW, Australia
| | - Nicole G Hansbro
- Centre for Inflammation, Centenary Institute and University of Technology Sydney, Sydney, NSW, Australia
| | - Chris J Burns
- Walter and Eliza Hall Institute of Medical Research, Department of Medical Biology, The University of Melbourne, Melbourne, VIC 3010, Australia
| | - Mathew S Eapen
- Respiratory Translational Research Group, Department of Laboratory Medicine, School of Health Sciences, University of Tasmania, Launceston, TAS, Australia
| | - Sukhwinder S Sohal
- Respiratory Translational Research Group, Department of Laboratory Medicine, School of Health Sciences, University of Tasmania, Launceston, TAS, Australia
| | - Janette K Burgess
- University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD (GRIAC), Department of Pathology and Medical Biology, Groningen, The Netherlands; Woolcock Institute of Medical Research, Discipline of Pharmacology, The University of Sydney, Sydney, NSW, Australia
| | - Philip M Hansbro
- Centre for Inflammation, Centenary Institute and University of Technology Sydney, Sydney, NSW, Australia.
| |
Collapse
|
8
|
Vu TN, Chen X, Foda HD, Smaldone GC, Hasaneen NA. Interferon-γ enhances the antifibrotic effects of pirfenidone by attenuating IPF lung fibroblast activation and differentiation. Respir Res 2019; 20:206. [PMID: 31511015 PMCID: PMC6737625 DOI: 10.1186/s12931-019-1171-2] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Accepted: 08/23/2019] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Idiopathic pulmonary fibrosis (IPF) pathogenesis involves multiple pathways, and combined antifibrotic therapy is needed for future IPF therapy. Inhaled interferon-γ (IFN-γ) was recently shown to be safe and without systemic effects in patients with IPF. AIM To examine the in vitro effects of individual and combined treatment with IFN-γ and pirfenidone (PFD) on normal and IPF fibroblast activation and extracellular matrix remodeling after TGF-β1 and PDGF-BB stimulation. METHODS IPF and normal human lung fibroblasts (NHLF) were treated with IFN-γ, PFD or a combination of both drugs in the presence of either TGF-β1 or PDGF-BB. The effects of TGF-β1 and PDGF-BB treatment on cell viability, proliferation, differentiation and migration were examined. The expression of collagen 1, matrix metalloproteinases (MMPs) and tissue inhibitors of MMP (TIMPs) was analyzed using qPCR, Western blotting and gelatin zymography. Total collagen content in conditioned media was also measured using a Sircol assay. RESULTS Compared to that of PFD, the effect of IFN-γ in downregulating normal and IPF lung fibroblast differentiation to myofibroblasts in response to TGF-β1 was more potent. Importantly, the combination of IFN-γ and PFD had a possibly synergistic/additive effect in inhibiting the TGF-β1- and PDGF-BB-induced proliferation, migration and differentiation of normal and IPF lung fibroblasts. Furthermore, both drugs reversed TGF-β1-induced effects on MMP-1, - 2, - 3, - 7, and - 9, while only PFD promoted TIMP-1 and-2 expression and release. CONCLUSIONS Our findings demonstrate that the antifibrotic effects of IFN-γ and PFD on normal and IPF lung fibroblasts are different and complementary. Combination therapy with inhaled IFN-γ and PFD in IPF is promising and should be further explored in IPF clinical trials.
Collapse
Affiliation(s)
- Tuong N Vu
- Pulmonary, Critical Care and Sleep Division, Department of Medicine, Stony Brook Medicine, Health Science Center, State University of New York at Stony Brook, HSC T17 Room 040, Stony Brook, NY, 11794-8172, USA
| | - Xuesong Chen
- Pulmonary, Critical Care and Sleep Division, Department of Medicine, Stony Brook Medicine, Health Science Center, State University of New York at Stony Brook, HSC T17 Room 040, Stony Brook, NY, 11794-8172, USA
| | - Hussein D Foda
- Pulmonary, Critical Care and Sleep Division, Department of Medicine, Stony Brook Medicine, Health Science Center, State University of New York at Stony Brook, HSC T17 Room 040, Stony Brook, NY, 11794-8172, USA.,Department of Medicine and Research, VAMC Northport, Stony Brook, NY, USA
| | - Gerald C Smaldone
- Pulmonary, Critical Care and Sleep Division, Department of Medicine, Stony Brook Medicine, Health Science Center, State University of New York at Stony Brook, HSC T17 Room 040, Stony Brook, NY, 11794-8172, USA
| | - Nadia A Hasaneen
- Pulmonary, Critical Care and Sleep Division, Department of Medicine, Stony Brook Medicine, Health Science Center, State University of New York at Stony Brook, HSC T17 Room 040, Stony Brook, NY, 11794-8172, USA. .,Department of Medicine and Research, VAMC Northport, Stony Brook, NY, USA.
| |
Collapse
|
9
|
Dolivo DM, Larson SA, Dominko T. Tryptophan metabolites kynurenine and serotonin regulate fibroblast activation and fibrosis. Cell Mol Life Sci 2018; 75:3663-3681. [PMID: 30027295 PMCID: PMC11105268 DOI: 10.1007/s00018-018-2880-2] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Revised: 06/27/2018] [Accepted: 07/16/2018] [Indexed: 01/06/2023]
Abstract
Fibrosis is a pathological form of aberrant tissue repair, the complications of which account for nearly half of all deaths in the industrialized world. All tissues are susceptible to fibrosis under particular pathological sets of conditions. Though each type of fibrosis has characteristics and hallmarks specific to that particular condition, there appear to be common factors underlying fibrotic diseases. One of these ubiquitous factors is the paradigm of the activated myofibroblast in the promotion of fibrotic phenotypes. Recent research has implicated metabolic byproducts of the amino acid tryptophan, namely serotonin and kynurenines, in the pathology or potential pharmacologic therapy of fibrosis, in part through their effects on development of myofibroblast phenotypes. Here, we review literature underlying what is known mechanistically about the effects of these compounds and their respective pathways on fibrosis. Pharmacologic administration of kynurenine improves scarring outcomes in vivo likely not only through its well-characterized immunosuppressive properties but also via its demonstrated antagonism of fibroblast activation and of collagen deposition. In contrast, serotonin directly promotes activation of fibroblasts via activation of canonical TGF-β signaling, and overstimulation with serotonin leads to fibrotic outcomes in vivo. Recently discovered feedback inhibition between serotonin and kynurenine pathways also reveals more information about the cellular physiology of tryptophan metabolism and may also underlie possible paradigms for anti-fibrotic therapy. Together, understanding of the effects of tryptophan metabolism on modulation of fibrosis may lead to the development of new therapeutic avenues for treatment through exploitation of these effects.
Collapse
Affiliation(s)
- David M Dolivo
- Biology and Biotechnology Department, Worcester Polytechnic Institute, 100 Institute Road, Worcester, MA, 01609, USA
| | - Sara A Larson
- Biology and Biotechnology Department, Worcester Polytechnic Institute, 100 Institute Road, Worcester, MA, 01609, USA
| | - Tanja Dominko
- Biology and Biotechnology Department, Worcester Polytechnic Institute, 100 Institute Road, Worcester, MA, 01609, USA.
| |
Collapse
|
10
|
Stunova A, Vistejnova L. Dermal fibroblasts—A heterogeneous population with regulatory function in wound healing. Cytokine Growth Factor Rev 2018; 39:137-150. [DOI: 10.1016/j.cytogfr.2018.01.003] [Citation(s) in RCA: 86] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Accepted: 01/11/2018] [Indexed: 02/06/2023]
|
11
|
Wang J, Lesko M, Badri MH, Kapoor BC, Wu BG, Li Y, Smaldone GC, Bonneau R, Kurtz ZD, Condos R, Segal LN. Lung microbiome and host immune tone in subjects with idiopathic pulmonary fibrosis treated with inhaled interferon-γ. ERJ Open Res 2017; 3:00008-2017. [PMID: 28717640 PMCID: PMC5507144 DOI: 10.1183/23120541.00008-2017] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Accepted: 04/14/2017] [Indexed: 01/27/2023] Open
Abstract
Therapies targeting inflammation reveal inconsistent results in idiopathic pulmonary fibrosis (IPF). Aerosolised interferon (IFN)-γ has been proposed as a novel therapy. Changes in the host airway microbiome are associated with the inflammatory milieu and may be associated with disease progression. Here, we evaluate whether treatment with aerosolised IFN-γ in IPF impacts either the lower airway microbiome or the host immune phenotype. Patients with IPF who enrolled in an aerosolised IFN-γ trial underwent bronchoscopy at baseline and after 6 months. 16S rRNA sequencing of bronchoalveolar lavage fluid (BALF) was used to evaluate the lung microbiome. Biomarkers were measured by Luminex assay in plasma, BALF and BAL cell supernatant. The compPLS framework was used to evaluate associations between taxa and biomarkers. IFN-γ treatment did not change α or β diversity of the lung microbiome and few taxonomic changes occurred. While none of the biomarkers changed in plasma, there was an increase in IFN-γ and a decrease in Fit-3 ligand, IFN-α2 and interleukin-5 in BAL cell supernatant, and a decrease in tumour necrosis factor-β in BALF. Multiple correlations between microbial taxa common to the oral mucosa and host inflammatory biomarkers were found. These data suggest that the lung microbiome is independently associated with the host immune tone and may have a potential mechanistic role in IPF. Lower airway microbiome and immunological tone are associated in IPF, an effect independent of IFN-γ treatmenthttp://ow.ly/cTDo30bsJiN
Collapse
Affiliation(s)
- Jing Wang
- Division of Pulmonary and Critical Care Medicine, Beijing Chaoyang Hospital, The Capital University of Medicine, Beijing, China.,Division of Pulmonary, Critical Care and Sleep Medicine, New York University School of Medicine, New York, NY, USA
| | - Melissa Lesko
- Division of Pulmonary, Critical Care and Sleep Medicine, New York University School of Medicine, New York, NY, USA
| | - Michelle H Badri
- Dept of Biology, Center for Genomics and Systems Biology, New York University, New York, NY, USA
| | - Bianca C Kapoor
- Division of Pulmonary, Critical Care and Sleep Medicine, New York University School of Medicine, New York, NY, USA
| | - Benjamin G Wu
- Division of Pulmonary, Critical Care and Sleep Medicine, New York University School of Medicine, New York, NY, USA
| | - Yonghua Li
- Division of Pulmonary, Critical Care and Sleep Medicine, New York University School of Medicine, New York, NY, USA
| | - Gerald C Smaldone
- Division of Pulmonary, Critical Care and Sleep Medicine, State University of New York at Stony Brook, Stony Brook, NY, USA
| | - Richard Bonneau
- Dept of Biology, Center for Genomics and Systems Biology, New York University, New York, NY, USA.,Courant Institute of Mathematical Sciences, New York University, New York, NY, USA.,Simons Center for Data Analysis, Simons Foundation, New York, NY, USA
| | - Zachary D Kurtz
- Dept of Microbiology, New York University School of Medicine, New York, NY, USA
| | - Rany Condos
- Division of Pulmonary, Critical Care and Sleep Medicine, New York University School of Medicine, New York, NY, USA
| | - Leopoldo N Segal
- Division of Pulmonary, Critical Care and Sleep Medicine, New York University School of Medicine, New York, NY, USA
| |
Collapse
|
12
|
Richards CD. Innate Immune Cytokines, Fibroblast Phenotypes, and Regulation of Extracellular Matrix in Lung. J Interferon Cytokine Res 2017; 37:52-61. [PMID: 28117653 DOI: 10.1089/jir.2016.0112] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Chronic inflammation can be caused by adaptive immune responses in autoimmune and allergic conditions, driven by a T lymphocyte subset balance (TH1, TH2, Th17, Th22, and/or Treg) and skewed cellular profiles in an antigen-specific manner. However, several chronic inflammatory diseases have no clearly defined adaptive immune mechanisms that drive chronicity. These conditions include those that affect the lung such as nonatopic asthma or idiopathic pulmonary fibrosis comprising significant health problems. The remodeling of extracellular matrix (ECM) causes organ dysfunction, and it is largely generated by fibroblasts as the major cell controlling net ECM. As such, these are potential targets of treatment approaches in the context of ECM pathology. Fibroblast phenotypes contribute to ECM and inflammatory cell accumulation, and they are integrated into chronic disease mechanisms including cancer. Evidence suggests that innate cytokine responses may be critical in nonallergic/nonautoimmune disease, and they enable environmental agent exposure mechanisms that are independent of adaptive immunity. Innate immune cytokines derived from macrophage subsets (M1/M2) and innate lymphoid cell (ILC) subsets can directly regulate fibroblast function. We also suggest that STAT3-activating gp130 cytokines can sensitize fibroblasts to the innate cytokine milieu to drive phenotypes and exacerbate existing adaptive responses. Here, we review evidence exploring innate cytokine regulation of fibroblast behavior.
Collapse
Affiliation(s)
- Carl D Richards
- Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre, McMaster University , Hamilton, Canada
| |
Collapse
|
13
|
Baroke E, Gauldie J, Kolb M. New treatment and markers of prognosis for idiopathic pulmonary fibrosis: lessons learned from translational research. Expert Rev Respir Med 2014; 7:465-78. [PMID: 24138691 DOI: 10.1586/17476348.2013.838015] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a chronic progressive interstitial lung disease with increasing prevalence, high mortality rates and poor treatment options. The diagnostic process is complex and often requires an interdisciplinary approach between different specialists. Information gained over the past 10 years of intense research resulted in improved diagnostic algorithms, a better understanding of the underlying pathogenesis and the development of new therapeutic options. Specifically, the change from the traditional concept that viewed IPF as a chronic inflammatory disorder to the current belief that is primarily resulting from aberrant wound healing enabled the identification of novel treatment targets. This increased the clinical trial activity dramatically and resulted in the approval of the first IPF-specific therapy in many countries. Still, the natural history and intrinsic behavior of IPF are very difficult to predict. There is an urgent need for new therapies and also for development and validation of prognostic markers that predict disease progression, survival and also response to antifibrotic drugs. This review provides an up to date summary of the most relevant clinical trials, novel therapeutic drug targets and outlines a spectrum of potential prognostic biomarkers for IPF.
Collapse
Affiliation(s)
- Eva Baroke
- Department of Medicine, McMaster University, ON, Canada, L8S4L8 and Department of Pathology & Molecular Medicine, McMaster University, Ontario ON, Canada, L8S4L8
| | | | | |
Collapse
|
14
|
Cytokine-like factor 1 gene expression is enriched in idiopathic pulmonary fibrosis and drives the accumulation of CD4+ T cells in murine lungs: evidence for an antifibrotic role in bleomycin injury. THE AMERICAN JOURNAL OF PATHOLOGY 2012; 180:1963-78. [PMID: 22429962 DOI: 10.1016/j.ajpath.2012.01.010] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/16/2011] [Revised: 11/22/2011] [Accepted: 01/20/2012] [Indexed: 11/21/2022]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a progressive and typically fatal lung disease. To gain insight into the pathogenesis of IPF, we reanalyzed our previously published gene expression data profiling IPF lungs. Cytokine receptor-like factor 1 (CRLF1) was among the most highly up-regulated genes in IPF lungs, compared with normal controls. The protein product (CLF-1) and its partner, cardiotrophin-like cytokine (CLC), function as members of the interleukin 6 (IL-6) family of cytokines. Because of earlier work implicating IL-6 family members in IPF pathogenesis, we tested whether CLF-1 expression contributes to inflammation in experimental pulmonary fibrosis. In IPF, we detected CLF-1 expression in both type II alveolar epithelial cells and macrophages. We found that the receptor for CLF-1/CLC signaling, ciliary neurotrophic factor receptor (CNTFR), was expressed only in type II alveolar epithelial cells. Administration of CLF-1/CLC to both uninjured and bleomycin-injured mice led to the pulmonary accumulation of CD4(+) T cells. We also found that CLF-1/CLC administration increased inflammation but decreased pulmonary fibrosis. CLF-1/CLC leads to significantly enriched expression of T-cell-derived chemokines and cytokines, including the antifibrotic cytokine interferon-γ. We propose that, in IPF, CLF-1 is a selective stimulus of type II alveolar epithelial cells and may potentially drive an antifibrotic response by augmenting both T-helper-1-driven and T-regulatory-cell-driven inflammatory responses in the lung.
Collapse
|
15
|
Selman M, Pardo A, Richeldi L, Cerri S. Emerging drugs for idiopathic pulmonary fibrosis. Expert Opin Emerg Drugs 2011; 16:341-62. [DOI: 10.1517/14728214.2011.565049] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
16
|
Coward WR, Saini G, Jenkins G. The pathogenesis of idiopathic pulmonary fibrosis. Ther Adv Respir Dis 2010; 4:367-88. [PMID: 20952439 DOI: 10.1177/1753465810379801] [Citation(s) in RCA: 222] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a progressive fibrotic lung disease with an appalling prognosis. The failure of anti-inflammatory therapies coupled with the observation that deranged epithelium overlies proliferative myofibroblasts to form the fibroblastic focus has lead to the emerging concept that IPF is a disease of deregulated epithelial-mesenchymal crosstalk. IPF is triggered by an as yet unidentified alveolar injury that leads to activation of transforming growth factor-β (TGF-β) and alveolar basement membrane disruption. In the presence of persisting injurious pathways, or disrupted repair pathways, activated TGF-β can lead to enhanced epithelial apoptosis and epithelial-to-mesenchymal transition (EMT) as well as fibroblast, and fibrocyte, transformation into myofibroblasts which are resistant to apoptosis. The resulting deposition of excess disrupted matrix by these myofibroblasts leads to the development of IPF.
Collapse
Affiliation(s)
- William R Coward
- Nottingham Respiratory Biomedical Research Unit, Clinical Sciences Building, Nottingham City Campus, Nottingham, UK
| | | | | |
Collapse
|
17
|
Corte TJ, Wells AU. Treatment of idiopathic interstitial pneumonias. Expert Rev Respir Med 2009; 3:81-91. [PMID: 20477284 DOI: 10.1586/17476348.3.1.81] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The idiopathic interstitial pneumonias can be grouped, for treatment purposes, into primary inflammatory disorders, fibrotic nonspecific interstitial pneumonia (in which inflammation is thought to precede and progress to fibrosis) as well as the most common of the idiopathic interstitial pneumonia subgroups, idiopathic pulmonary fibrosis. Over the past decade, there have been several paradigm shifts in the understanding of idiopathic interstitial pneumonias and their treatment. In particular, we highlight changes in the use of prognostic markers, clinical trial end points and the understanding of pathogenesis of idiopathic pulmonary fibrosis. We outline a practical approach to the treatment of these three patient groups.
Collapse
Affiliation(s)
- Tamera J Corte
- Clinical Research Fellow, Department of Interstitial Lung Disease, Royal Brompton Hospital, Sydney Street, London, SW3 6NP, UK.
| | | |
Collapse
|
18
|
Vannella KM, Moore BB. Viruses as co-factors for the initiation or exacerbation of lung fibrosis. FIBROGENESIS & TISSUE REPAIR 2008; 1:2. [PMID: 19014649 PMCID: PMC2577044 DOI: 10.1186/1755-1536-1-2] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/12/2008] [Accepted: 10/13/2008] [Indexed: 12/27/2022]
Abstract
Idiopathic pulmonary fibrosis (IPF) remains exactly that. The disease originates from an unknown cause, and little is known about the mechanisms of pathogenesis. While the disease is likely multi-factorial, evidence is accumulating to implicate viruses as co-factors (either as initiating or exacerbating agents) of fibrotic lung disease. This review summarizes the available clinical and experimental observations that form the basis for the hypothesis that viral infections may augment fibrotic responses. We review the data suggesting a link between hepatitis C virus, adenovirus, human cytomegalovirus and, in particular, the Epstein-Barr gammaherpesvirus, in IPF. In addition, we highlight the recent associations made between gammaherpesvirus infection and lung fibrosis in horses and discuss the various murine models that have been used to investigate the contribution of gammaherpesviruses to fibrotic progression. We review the work demonstrating that gammaherpesvirus infection of Th2-biased mice leads to multi-organ fibrosis and highlight studies showing that gammaherpesviral infections of mice either pre- or post-fibrotic challenge can augment the development of fibrosis. Finally, we discuss potential mechanisms whereby viral infections may amplify the development of fibrosis. While none of these studies prove causality, we believe the evidence suggests that viral infections should be considered as potential initiators or exacerbating agents in at least some cases of IPF and thereby justify further study.
Collapse
Affiliation(s)
- Kevin M Vannella
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI 48109, USA.
| | | |
Collapse
|
19
|
Abraham LC, Dice JF, Finn PF, Mesires NT, Lee K, Kaplan DL. Extracellular matrix remodeling--methods to quantify cell-matrix interactions. Biomaterials 2006; 28:151-61. [PMID: 16893566 DOI: 10.1016/j.biomaterials.2006.07.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2006] [Accepted: 07/03/2006] [Indexed: 11/29/2022]
Abstract
Tissue turnover during wound healing, regeneration or integration of biomedical materials depends on the rate and extent of materials trafficking into and out of cells involved in extracellular matrix (ECM) remodeling. To exploit these processes, we report the first model for matrix trafficking in which these issues are quantitatively assessed for cells grown on both native collagen (normal tissue) and denatured collagen (wound state) substrates. Human fibroblasts more rapidly remodeled denatured versus normal collagen type I to form new ECM. Fluxes to and from the cells from the collagen substrates and the formation of new ECM were quantified using radioactively labeled substrates. The model can be employed for the systematic and quantitative study of the impact of a broad range of physiological factors and disease states on tissue remodeling, integrating extracellular matrix structures and cell biology.
Collapse
Affiliation(s)
- Leah C Abraham
- Department of Chemical and Biological Engineering, and Bioengineering and Biotechnology Center, Tufts University, Medford, MA 02155, USA
| | | | | | | | | | | |
Collapse
|
20
|
Abstract
Idiopathic pulmonary fibrosis (IPF) is associated with a poor prognosis, although considerable interpatient heterogeneity may be observed. Baseline dyspnea, lung function abnormalities, and gas exchange impairments are predictive of outcome, but recent evidence suggests the change in these parameters over time is as predictive, and in some cases more predictive, of outcome. A change in forced vital capacity > or =10% may be an important surrogate marker for mortality risk in IPF patients. Histologic analysis shows the deposition of interstitial connective tissue and granulation tissue leading to formation of fibroblastic foci. Thus, ongoing epithelial damage and repair processes associated with persistent fibroblastic proliferation seem important in IPF natural history.
Collapse
Affiliation(s)
- Paul W Noble
- Section of Pulmonary and Critical Care Medicine, Yale University School of Medicine, 441-C CAB, 333 Cedar Street, New Haven, CT 06520-8057, USA.
| |
Collapse
|
21
|
Nathan SD. Therapeutic Management of Idiopathic Pulmonary Fibrosis: An Evidence-Based Approach. Clin Chest Med 2006; 27:S27-35, vi. [PMID: 16545630 DOI: 10.1016/j.ccm.2005.08.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The treatment of idiopathic pulmonary fibrosis (IPF) remains controversial. The benefits of conventional treatment with corticosteroids plus either azathioprine or cyclophosphamide have not been established in randomized, controlled trials. Other treatment strategies have been suggested as understanding of the pathogenesis of IPF has increased. Recent clinical and animal studies suggest that modulating the effects of profibrotic growth factors and cytokines holds significant promise. As additional well controlled prospective studies are completed, these trials should provide the clinical evidence necessary for identifying optimal treatment strategies for future patients who have IPF.
Collapse
Affiliation(s)
- Steven D Nathan
- Lung Transplant and Advanced Lung Disease Program, Inova Heart and Vascular Institute, 3300 Gallows Road, Falls Church, VA 22042, USA.
| |
Collapse
|
22
|
Abstract
Idiopathic pulmonary fibrosis (IPF) is a chronic, progressive, and usually fatal pulmonary disease for which there are no proven drug therapies. Anti-inflammatory and immunosuppressive agents have been largely ineffective. The precise relationship of IPF to other idiopathic interstitial pneumonias (IIPs) is not known, despite the observation that different histopathologic patterns of IIP may coexist in the same patient. We propose that these different histopathologic 'reaction' patterns may be determined by complex interactions between host and environmental factors that alter the local alveolar milieu. Recent paradigms in IPF pathogenesis have focused on dysregulated epithelial-mesenchymal interactions, an imbalance in T(H)1/T(H)2 cytokine profile and potential roles for aberrant angiogenesis. In this review, we discuss these evolving concepts in disease pathogenesis and emerging therapies designed to target pro-fibrogenic pathways in IPF.
Collapse
Affiliation(s)
| | - Victor J. Thannickal
- Address correspondence to: Victor J. Thannickal, M.D. Division of Pulmonary and Critical Care Medicine University of Michigan Medical Center 6301 MSRB III 1150 W. Medical Center Dr. Ann Arbor, Michigan 48109 United States of America Phone: 734−936−9371 Fax: 734−764−4556 e-mail:
| |
Collapse
|
23
|
Vayalil PK, Olman M, Murphy-Ullrich JE, Postlethwait EM, Liu RM. Glutathione restores collagen degradation in TGF-beta-treated fibroblasts by blocking plasminogen activator inhibitor-1 expression and activating plasminogen. Am J Physiol Lung Cell Mol Physiol 2005; 289:L937-45. [PMID: 16258002 DOI: 10.1152/ajplung.00150.2005] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Transforming growth factor (TGF)-beta plays an important role in tissue fibrogenesis. We previously demonstrated that reduced glutathione (GSH) supplementation blocked collagen accumulation induced by TGF-beta in NIH-3T3 cells. In the present study, we show that supplementation of GSH restores the collagen degradation rate in TGF-beta-treated NIH-3T3 cells. Restoration of collagen degradation by GSH is associated with a reduction of type I plasminogen activator inhibitor (PAI)-1 expression/activity as well as recovery of the activities of cell/extracellular matrix-associated tissue-type plasminogen activator and plasmin. Furthermore, we find that NIH-3T3 cells constitutively express plasminogen mRNA and possess plasmin activity. Blockade of cell surface binding of plasminogen/plasminogen activation with tranexamic acid (TXA) or inhibition of plasmin activity with aprotinin significantly reduces the basal level of collagen degradation both in the presence or absence of exogenous plasminogen. Most importantly, addition of TXA or active PAI-1 almost completely eliminates the restorative effects of GSH on collagen degradation in TGF-beta treated cells. Together, our results suggest that the major mechanism by which GSH restores collagen degradation in TGF-beta-treated cells is through blocking PAI-1 expression, leading to increased PA/plasmin activity and consequent proteolytic degradation of collagens. This study provides mechanistic evidence for GSH's putative therapeutic effect in the treatment of fibrotic disorders.
Collapse
Affiliation(s)
- Praveen K Vayalil
- Dept. of Environmental Health Sciences, School of Public Health, University of Alabama at Birmingham, Birmingham, AL, USA
| | | | | | | | | |
Collapse
|
24
|
Natividad A, Wilson J, Koch O, Holland MJ, Rockett K, Faal N, Jallow O, Joof HM, Burton MJ, Alexander NDE, Kwiatkowski DP, Mabey DCW, Bailey RL. Risk of trachomatous scarring and trichiasis in Gambians varies with SNP haplotypes at the interferon-gamma and interleukin-10 loci. Genes Immun 2005; 6:332-40. [PMID: 15789056 DOI: 10.1038/sj.gene.6364182] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Experimental evidence implicates interferon gamma (IFNgamma) in protection from and resolution of chlamydial infection. Conversely, interleukin 10 (IL10) is associated with susceptibility and persistence of infection and pathology. We studied genetic variation within the IL10 and IFNgamma loci in relation to the risk of developing severe complications of human ocular Chlamydia trachomatis infection. A total of 651 Gambian subjects with scarring trachoma, of whom 307 also had potentially blinding trichiasis and pair-matched controls with normal eyelids, were screened for associations between single-nucleotide polymorphisms (SNPs), SNP haplotypes and the risk of disease. MassEXTEND (Sequenom) and MALDI-TOF mass spectrometry were used for detection and analysis of SNPs and the programs PHASE and SNPHAP used to infer haplotypes from population genetic data. Multivariate conditional logistic regression analysis identified IL10 and IFNgamma SNP haplotypes associated with increased risk of both trachomatous scarring and trichiasis. SNPs in putative IFNgamma and IL10 regulatory regions lay within the disease-associated haplotypes. The IFNgamma +874A allele, previously linked to lower IFNgamma production, lies in the IFNgamma risk haplotype and was more common among cases than controls, but not significantly so. The promoter IL10-1082G allele, previously associated with high IL10 expression, is in both susceptibility and resistance haplotypes.
Collapse
Affiliation(s)
- A Natividad
- London School of Hygiene and Tropical Medicine, London University, London, UK.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Pacanowski MA, Amsden GW. Interferon Gamma-1b in the Treatment of Idiopathic Pulmonary Fibrosis. Ann Pharmacother 2005; 39:1678-86. [PMID: 16160001 DOI: 10.1345/aph.1e672] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Objective: To examine the clinical aspects of idiopathic pulmonary fibrosis (IPF) and the efficacy and safety of interferon gamma-1b (IFNγ−1b) in its treatment. Data Sources: Epidemiologic, preclinical, and clinical studies published in the English language were identified by a MEDLINE search (1966–January 2005) using the search terms idiopathic pulmonary fibrosis, cryptogenic fibrosing alveolitis, and interferon. Additional citations were identified from the reference lists of related publications. Study Selection and Data Extraction: Selected preclinical studies describing the pathophysiologic basis for IFNγ−1b therapy and all clinical studies were included. Additional trials describing other treatment modalities and the determinants of response to therapy in patients with IPF were also reviewed. Data Synthesis: IFNγ−1b targets the fibrotic rather than inflammatory processes of IPF. The efficacy of IFNγ−1b in patients with IPF is inconsistent with regard to changes in pulmonary function and mortality, although a modest survival benefit was observed in the largest clinical trial. Adverse events related to IFNγ−1b are frequent although transient. Several cases of respiratory failure occurring subsequent to the administration of IFNγ−1b are documented. CONCLUSIONS: To date, although trials suggest that earlier-stage IPF may be responsive to IFNγ−1b, study results overall are inconsistent; further investigation is needed.
Collapse
Affiliation(s)
- Michael A Pacanowski
- Section of Clinical Pharmacology, Department of Pharmaceutical Care Services, Bassett Healthcare, Cooperstown, NY 13326-1394, USA
| | | |
Collapse
|
26
|
Bajwa EK, Ayas NT, Schulzer M, Mak E, Ryu JH, Malhotra A. Interferon-γ1b Therapy in Idiopathic Pulmonary Fibrosis. Chest 2005; 128:203-6. [PMID: 16002936 DOI: 10.1378/chest.128.1.203] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
CONTEXT Despite the investigation of multiple therapeutic options, idiopathic pulmonary fibrosis (IPF) remains a devastating, progressively fatal disease. Much interest has focused on the use of interferon (IFN)-gamma1b therapy, but the efficacy of this treatment has not been proven. OBJECTIVE To determine whether IFN treatment reduces mortality in patients with IPF. DESIGN A meta-analysis of randomized controlled trials evaluating the use of IFN-gamma1b as treatment for IPF. MAIN OUTCOME MEASURE Mortality in patients treated with IFN-gamma1b was compared to mortality in patients treated with control therapies. RESULTS A total of three studies involving 390 patients was included in the analysis. IFN-gamma1b therapy was associated with reduced mortality (hazard ratio [HR], 0.418; 95% confidence interval [CI], 0.253 to 0.690; p = 0.0003). A comparison of mortality at different time points revealed that IFN-gamma1b therapy was associated with significantly reduced mortality at 1 year (0.0861; 95% CI, 0.0244 to 0.1478; p = 0.0063), 18 months (0.1682; 95% CI, 0.1065 to 0.2299; p < 0.0001), 650 days (0.1939; 95% CI, 0.1386 to 0.2492; p < 0.0001), and 2 years (0.2652; 95% CI, 0.1652 to 0.3652; p < 0.0001). CONCLUSION When the results of multiple studies are combined in a meta-analysis, IFN-gamma1b therapy is associated with reduced mortality.
Collapse
Affiliation(s)
- Ednan K Bajwa
- Brigham and Women's Hospital, 75 Francis Street, Boston, MA 02115, USA
| | | | | | | | | | | |
Collapse
|
27
|
Affiliation(s)
- Thomas A Wynn
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 50 South Drive, Room 6154, MSC 8003, Bethesda, Maryland 20892, USA.
| |
Collapse
|
28
|
Raghu G, Brown KK, Bradford WZ, Starko K, Noble PW, Schwartz DA, King TE. A placebo-controlled trial of interferon gamma-1b in patients with idiopathic pulmonary fibrosis. N Engl J Med 2004; 350:125-33. [PMID: 14711911 DOI: 10.1056/nejmoa030511] [Citation(s) in RCA: 462] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
BACKGROUND Idiopathic pulmonary fibrosis is a progressive, fatal disease with no known efficacious therapy. METHODS In a double-blind, multinational trial, we randomly assigned 330 patients with idiopathic pulmonary fibrosis that was unresponsive to corticosteroid therapy to receive subcutaneous interferon gamma-1b or placebo. RESULTS Over a median of 58 weeks, interferon gamma-1b therapy did not significantly affect the primary end point of progression-free survival, defined as the time to disease progression or death, and no significant treatment effect was observed on measures of lung function, gas exchange, or the quality of life. Ten percent of patients in the interferon gamma-1b group died, as compared with 17 percent of patients in the placebo group (P=0.08). Treatment with interferon gamma-1b was associated with more frequent constitutional symptoms. However, the rates of treatment adherence and premature discontinuation of treatment were similar in the two groups. More pneumonias were reported among patients in the interferon gamma-1b group, but the incidence of severe or life-threatening respiratory tract infections was similar in the two groups. CONCLUSIONS In a well-defined population of patients with idiopathic pulmonary fibrosis, interferon gamma-1b did not affect progression-free survival, pulmonary function, or the quality of life. Owing to the size and duration of the trial, a clinically significant survival benefit could not be ruled out.
Collapse
Affiliation(s)
- Ganesh Raghu
- Division of Pulmonary Medicine, University of Washington, Seattle 98195, USA.
| | | | | | | | | | | | | |
Collapse
|
29
|
Whitehead GS, Grasman KA, Kimmel EC. Lung function and airway inflammation in rats following exposure to combustion products of carbon-graphite/epoxy composite material: comparison to a rodent model of acute lung injury. Toxicology 2003; 183:175-97. [PMID: 12504350 DOI: 10.1016/s0300-483x(02)00542-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Pulmonary function and inflammation in the lungs of rodents exposed by inhalation to carbon/graphite/epoxy advanced composite material (ACM) combustion products were compared to that of a rodent model of acute lung injury (ALI) produced by pneumotoxic paraquat dichloride. This investigation was undertaken to determine if short-term exposure to ACM smoke induces ALI; and to determine if smoke-related responses were similar to the pathogenic mechanisms of a model of lung vascular injury. We examined the time-course for mechanical lung function, infiltration of inflammatory cells into the lung, and the expression of three inflammatory cytokines, tumor necrosis factor-alpha (TNF-alpha), macrophage inflammatory protein-2 (MIP-2) and interferon-gamma (IFN-gamma). Male Fischer-344 rats were either exposed to 26.8-29.8 g/m(3) nominal concentrations of smoke or were given i.p. injections of paraquat dichloride. Measurements were determined at 1, 2, 3, and 7 days post exposure. In the smoke-challenged rats, there were no changes in lung function indicative of ALI throughout the 7-day observation period, despite the acute lethality of the smoke atmosphere. However, the animals showed signs of pulmonary inflammation. The expression of TNF-alpha was significantly increased in the lavage fluid 1 day following exposure, which preceded the maximum leukocyte infiltration. MIP-2 levels were significantly increased in lavage fluid at days 2, 3, and 7. This followed the leukocyte infiltration. IFN-gamma was significantly increased in the lung tissue at day 7, which occurred during the resolution of the inflammatory response. The paraquat, which was also lethal to a small percentage of the animals, caused several physiologic changes characteristic of ALI, including significant decreases in lung compliance, lung volumes/capacities, distribution of ventilation, and gas exchange capacity. The expression of TNF-alpha and MIP-2 increased significantly in the lung tissue as well as in the lavage fluid. Increased MIP-2 levels also preceded the maximum neutrophil infiltration. The differences in the time-course and primary site of TNF-alpha, MIP-2, and IFN-gamma expression; and the differences in the temporal relationship between their expression and infiltration of inflammatory cells may have accounted for the differences in lung function between paraquat treated and ACM smoke exposed animals.
Collapse
Affiliation(s)
- Gregory S Whitehead
- Department of Pulmonary and Critical Care Medicine, Duke University Medical Center, Durham, NC 27710-0001, USA.
| | | | | |
Collapse
|
30
|
Abstract
Fibroproliferative lung disease is prevalent and associated with high mortality. The pathogenesis of fibrotic lung disease involves inflammation, mesenchymal cell proliferation, and deposition of interstitial matrix components, such as collagen and fibronectin. Corticosteroids and other immunosuppressive medications have been routinely employed, but have demonstrated only marginal efficacy. Even though this information has been known for some time, the optimal medical regimen for treating pulmonary fibrosis remains largely undefined. This article addresses the rationale for, and efficacy of, antifibrotic regimens used to treat humans with fibrotic lung diseases.
Collapse
Affiliation(s)
- J A Lasky
- Department of Medicine, Tulane University Medical Center, New Orleans, Louisiana 70112, USA.
| | | |
Collapse
|
31
|
Keane MP, Belperio JA, Burdick MD, Strieter RM. IL-12 attenuates bleomycin-induced pulmonary fibrosis. Am J Physiol Lung Cell Mol Physiol 2001; 281:L92-7. [PMID: 11404251 DOI: 10.1152/ajplung.2001.281.1.l92] [Citation(s) in RCA: 101] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Interleukin (IL)-12 is a potent inducer of interferon (IFN)-gamma. We postulated that IL-12 would attenuate bleomycin-induced pulmonary fibrosis. To test this hypothesis, we administered IL-12 or murine serum albumin to bleomycin-treated mice by daily intraperitoneal injection until day 12. Mice treated with IL-12 demonstrated decreased hydroxyproline levels compared with control treated mice. Furthermore, administration of IL-12 led to a time-dependent increase in both lung and bronchoalveolar lavage fluid IFN-gamma. The antifibrotic effect of IL-12 could be attenuated with simultaneous administration of neutralizing anti-IFN-gamma antibodies. These findings support the notion that IL-12 attenuates bleomycin-induced pulmonary fibrosis via modulation of IFN-gamma production.
Collapse
Affiliation(s)
- M P Keane
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of California, Los Angeles School of Medicine, Los Angeles, California 90095-1922, USA.
| | | | | | | |
Collapse
|
32
|
Ebrahimi B, Dutia BM, Brownstein DG, Nash AA. Murine gammaherpesvirus-68 infection causes multi-organ fibrosis and alters leukocyte trafficking in interferon-gamma receptor knockout mice. THE AMERICAN JOURNAL OF PATHOLOGY 2001; 158:2117-25. [PMID: 11395389 PMCID: PMC1892003 DOI: 10.1016/s0002-9440(10)64683-4] [Citation(s) in RCA: 60] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 03/13/2001] [Indexed: 11/19/2022]
Abstract
Murine gammaherpesvirus-68 (MHV-68) infection in interferon-gamma receptor knockout mice (IFN-gammaR(-)/(-)) results in splenic fibrosis and excessive loss of splenocytes. In our present study we found that MHV-68 infection in IFN-gammaR(-)/(-) mice also resulted in fibrosis and atrophy of the mediastinal lymph nodes, interstitial pulmonary fibrosis and fibrotic changes in the liver. Atrophy and cellular depletion of the spleen in IFN-gammaR(-)/(-) was not the result of increased cell death. The loss of splenocytes in IFN-gammaR(-)/(-) mice, which was most evident on day 23 after infection, correlated with an increase in the number of leukocytes in peripheral blood. At the peak of leukocytosis, on day 23 after infection, peripheral blood cells from infected IFN-gammaR(-)/(-) mice were unable to traffic through the fibrosed spleens of IFN-gammaR(-)/(-) mice but were able to enter the spleens of wild-type mice. This indicates that leukocytosis was in part the result of emigration of cells from the spleen and their subsequent exclusion of re-entry at the height of fibrosis. Significant cytokine and chemokine changes were observed in spleens of IFN-gammaR(-)/(-) mice. IFN-gamma, tumor necrosis factor-alpha (TNF-alpha ), TNF-beta, interleukin-1beta (IL-1beta), transforming growth factor-beta1 (TGF-beta1), lymphotactin, and MIP-1beta were elevated on day 14 after infection whereas chemokines IP-10 and MIG were significantly reduced. These changes suggest a role for dysregulated cytokines and chemokines in severe organ-specific fibrosis with implications for immune-mediated fibrotic disorders.
Collapse
Affiliation(s)
- B Ebrahimi
- Laboratory for Clinical and Molecular Virology, Department of Veterinary Pathology, University of Edinburgh, United Kingdom
| | | | | | | |
Collapse
|
33
|
Aggarwal AN, Behera D. Interferon-gamma 1b: impact of new indications (idiopathic pulmonary fibrosis). Expert Opin Pharmacother 2000; 1:1423-7. [PMID: 11249475 DOI: 10.1517/14656566.1.7.1423] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a relentlessly progressive disease with inadequate response to conventional treatment with corticosteroids and/or immunosuppressive agents in most patients. Interferon-gamma (IFN-gamma), an antifibrotic agent, has been proposed as a novel therapeutic approach. Several investigators have shown a relative decrease in systemic and pulmonary IFN-gamma activity in patients with IPF. Experimental evidence from animal and human studies also suggests that IFN-gamma administration may ameliorate lung fibrosis. Clinical experience is, however, limited to a single clinical trial that showed objective functional improvement in a small number of patients treated with IFN-gamma and low-dose corticosteroids. Further research is needed to characterise the efficacy, safety and optimum route of administration of this agent before it can be recommended for use in routine clinical practice.
Collapse
Affiliation(s)
- A N Aggarwal
- Department of Pulmonary Medicine, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | | |
Collapse
|
34
|
Yuan W, Yufit T, Li L, Mori Y, Chen SJ, Varga J. Negative modulation of alpha1(I) procollagen gene expression in human skin fibroblasts: transcriptional inhibition by interferon-gamma. J Cell Physiol 1999; 179:97-108. [PMID: 10082137 DOI: 10.1002/(sici)1097-4652(199904)179:1<97::aid-jcp12>3.0.co;2-e] [Citation(s) in RCA: 47] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Interferon-gamma (IFN-gamma), a multifunctional cytokine produced by activated Th1 lymphocytes, exerts potent effects on the extracellular matrix by regulating fibroblast function. In this study, we examined the modulation of alpha1(I) procollagen gene (COL1A1) expression by recombinant IFN-gamma. The results showed that IFN-gamma stimulated the rapid accumulation of interferon regulated factor (IRF)-1 mRNA, followed by a delayed and dose-dependent inhibition of alpha1(I) procollagen mRNA expression in skin fibroblasts from several different donors. The inhibitory response was abrogated in fibroblasts stably expressing IRF-1 in the antisense orientation. A marked decrease in the amount of heterogeneous nuclear pre-mRNA preceded the inhibition of COL1A1 mRNA expression. In fibroblasts transiently transfected with COL1A1 promoter-chloramphenicol acetyltransferase reporter gene plasmids, IFN-gamma selectively inhibited promoter activity and abrogated its stimulation induced by TGF-beta. The inhibition by IFN-gamma was not due to downregulation of TGF-beta receptor mRNA expression in the fibroblasts or decreased ligand binding to the receptor. IFN-alpha and IFN-beta by themselves had little effect on promoter activity, but IFN-alpha augmented the inhibitory effect of IFN-gamma. Using a series of 5' deletion constructs, a proximal region of the COL1A1 promoter was shown to function as an IFN-gamma response element. This region of the gene harbors overlapping binding sites for transcription factors Sp1, Sp3, and NF-1 but no homologs of previously characterized IFN-gamma response elements. The putative IFN-gamma response region was sufficient to confer inhibition of reporter gene expression by treatment with IFN-gamma. Gel mobility shift analysis showed that two distinct and specific DNA-protein complexes were formed when fibroblast nuclear extracts were incubated with oligonucleotides spanning the IFN-gamma response region. IFN-gamma did not modify the ability of nuclear proteins to bind to this region. The results indicate that IFN-gamma inhibits COL1A1 expression in fibroblasts principally at the level of gene transcription. Inhibition involves IRF-1 and is mediated through a short proximal promoter segment but without an apparent change in promoter occupancy. The findings provide novel insight into the mechanism of IFN-gamma regulation of fibroblast function.
Collapse
MESH Headings
- Adult
- Binding Sites
- Cell Nucleus/chemistry
- Cells, Cultured
- Chloramphenicol O-Acetyltransferase/biosynthesis
- Chloramphenicol O-Acetyltransferase/genetics
- DNA/metabolism
- DNA-Binding Proteins/physiology
- Depression, Chemical
- Fibroblasts/drug effects
- Fibroblasts/metabolism
- Gene Expression Regulation/drug effects
- Genes, Reporter
- Humans
- Infant, Newborn
- Interferon Regulatory Factor-1
- Interferon-alpha/pharmacology
- Interferon-gamma/pharmacology
- Male
- Phosphoproteins/physiology
- Procollagen/genetics
- Promoter Regions, Genetic/drug effects
- Promoter Regions, Genetic/genetics
- Protein Isoforms/biosynthesis
- Protein Isoforms/genetics
- RNA, Heterogeneous Nuclear/biosynthesis
- RNA, Heterogeneous Nuclear/genetics
- RNA, Messenger/biosynthesis
- RNA, Messenger/genetics
- Receptors, Transforming Growth Factor beta/drug effects
- Recombinant Proteins
- Sequence Deletion
- Skin/cytology
- Transcription Factors/metabolism
- Transcription, Genetic/drug effects
- Transfection
- Transforming Growth Factor beta/pharmacology
Collapse
Affiliation(s)
- W Yuan
- Section of Rheumatology, University of Illinois College of Medicine, Chicago 60607-7171, USA
| | | | | | | | | | | |
Collapse
|
35
|
Awad M, Pravica V, Perrey C, El Gamel A, Yonan N, Sinnott PJ, Hutchinson IV. CA repeat allele polymorphism in the first intron of the human interferon-gamma gene is associated with lung allograft fibrosis. Hum Immunol 1999; 60:343-6. [PMID: 10363726 DOI: 10.1016/s0198-8859(98)00133-5] [Citation(s) in RCA: 94] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Interferon-gamma (IFN-gamma) is an inflammatory cytokine that has been implicated in the development of fibrosis in inflamed tissues. In this study we have analysed the association between genetically-determined high IFN-gamma production and development of fibrosis in lung transplants. The human IFN-gamma gene has a variable length CA repeat in the first intron. Our previous study showed that polymorphism of this microsatellite is associated with individual variation in the levels of IFN-gamma production. In vitro production of IFN-gamma showed significant correlation with presence of allele #2 (p < 0.01). In this study allele #2 was found to be associated with allograft fibrosis defined by transbronchial biopsy. An analysis of two groups of lung transplant recipients showed a significant increase in the frequency of allele #2 in the group which developed fibrosis after transplantation compared to the group that did not (p < 0.005). We postulate that the production of IFN-gamma, which is under genetic control, can influence the development of fibrosis in lung allografts.
Collapse
Affiliation(s)
- M Awad
- School of Biological Sciences, University of Manchester, UK
| | | | | | | | | | | | | |
Collapse
|
36
|
Yokozeki M, Baba Y, Shimokawa H, Moriyama K, Kuroda T. Interferon-gamma inhibits the myofibroblastic phenotype of rat palatal fibroblasts induced by transforming growth factor-beta1 in vitro. FEBS Lett 1999; 442:61-4. [PMID: 9923605 DOI: 10.1016/s0014-5793(98)01626-3] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Interferon-gamma (IFN-gamma), a multifunctional cytokine, has been noted as a potential therapeutic agent for various fibrotic disorders, including excessive scar tissue formation. We previously reported that transforming growth factor-beta1 (TGF-beta1) induced the myofibroblastic phenotype in palatal fibroblasts derived from palatal mucosa, and that such effects might have a close link to palatal scar formation. In the present study, we examined the effects of IFN-gamma on TGF-beta1-pretreated palatal fibroblasts for the purpose of clarifying the suppressive potency against myofibroblastic phenotype expression in vitro. IFN-gamma significantly altered the spindle morphology of TGF-beta1-pretreated palatal fibroblasts into the polygonal one that was similar to the non-treated palatal fibroblasts. This change was parallel with a decrease in the expression of alpha-smooth muscle actin protein, a marker for myofibroblast, as determined by immunoblot analysis. Northern blot analysis showed that IFN-gamma inhibited proalpha2(I) collagen mRNA expression that was stimulated by TGF-beta1 pretreatment for 24 h. Furthermore, IFN-gamma decreased the cell contractility enhanced by TGF-beta1 pretreatment for 24 h in a three-dimensional collagen gel culture system. These results suggest that IFN-gamma may have negative effects with regard to controlling the myofibroblastic phenotype induced by TGF-beta1 in palatal fibroblasts.
Collapse
Affiliation(s)
- M Yokozeki
- Second Department of Orthodontics, Faculty of Dentistry, Tokyo Medical and Dental University, Japan
| | | | | | | | | |
Collapse
|
37
|
|
38
|
Badorff C, Noutsias M, Kühl U, Schultheiss HP. Cell-mediated cytotoxicity in hearts with dilated cardiomyopathy: correlation with interstitial fibrosis and foci of activated T lymphocytes. J Am Coll Cardiol 1997; 29:429-34. [PMID: 9015000 DOI: 10.1016/s0735-1097(96)00475-5] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
OBJECTIVES The purpose of this study was to investigate the expression of perforin and T-cell intracellular antigen-1, two crucial components of lymphocyte-mediated cytotoxicity, in endomyocardial biopsies from patients with idiopathic dilated cardiomyopathy. BACKGROUND Previous reports have demonstrated the presence of myocardial interstitial fibrosis and increased infiltrating lymphocytes in patients with dilated cardiomyopathy. However, the pathogenic significance of these lymphocytic infiltrates remains unclear. METHODS Endomyocardial biopsies from 134 patients with idiopathic dilated cardiomyopathy were histologically and immunohistologically analyzed. Monoclonal antibodies against diverse T-lymphocyte antigens, perforin and T-cell intracellular antigen-1 were used with the highly sensitive avidin-biotin complex technique. Positive cells were counted in at least 10 high power field. RESULTS Perforin and T-cell intracellular antigen-1 were immunohistologically detected in all biopsies. Immunoreactivity was restricted to the cytoplasm and was granular in nature, indicating specific staining of cytoplasmic granules. Correlations were established between the expression of perforin and T-cell intracellular antigen-1 and the abundance of foci of various T-lymphocyte subpopulations and, most importantly, the degree of interstitial fibrosis on routine histologic examination (p = 0.015). CONCLUSIONS Cytotoxic activity is clearly present in endomyocardial biopsies from patients with idiopathic dilated cardiomyopathy. Local activation-that is, focal accumulation of T lymphocytes-seems to be important for the generation of lymphocyte-mediated cytotoxicity. The interstitial fibrosis commonly seen in dilated cardiomyopathy may be caused by cytotoxic T-lymphocyte damage to the myocardium.
Collapse
Affiliation(s)
- C Badorff
- Department of Cardiology, Benjamin Franklin-Hospital, Free University of Berlin, Germany.
| | | | | | | |
Collapse
|
39
|
Diaz A, Jiménez SA. Interferon-gamma regulates collagen and fibronectin gene expression by transcriptional and post-transcriptional mechanisms. Int J Biochem Cell Biol 1997; 29:251-60. [PMID: 9076960 DOI: 10.1016/s1357-2725(96)00112-4] [Citation(s) in RCA: 43] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Interferon-gamma (IFN-gamma) regulates the expression of collagen and fibronectin genes by molecular mechanisms not completely understood. We investigated the effects of IFN-gamma on the expression of the genes encoding alpha 1 (I) procollagen (COL1A1), alpha 1 (III) procollagen (COL3A1), and fibronectin (FN) in cultured normal human lung fibroblasts. Labeled newly synthesized proteins were analysed by electrophoresis, mRNA levels and stability by Northern hybridizations, and transcription rates by in vitro assays. IFN-gamma caused a reduction in the production of alpha 1 (I) and alpha 1 (III) procollagens and of fibronectin. The reduction in the production of procollagen chains was shown to result from a combination of IFN-gamma-induced inhibition of the transcription rates of the COL1A1 and COL3A1 genes and destabilization of the corresponding transcripts. IFN-gamma increased the transcription rate of FN, but also decreased the stability of the corresponding transcripts. The net results indicate that the regulation of the expression of extracellular matrix genes by IFN-gamma is a complex process that involves changes in gene transcription rates, alterations in mRNAs stability, and possibly, modulation of the rates of translation.
Collapse
Affiliation(s)
- A Diaz
- Department of Medicine, Jefferson Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | | |
Collapse
|
40
|
Gailit J, Xu J, Bueller H, Clark RA. Platelet-derived growth factor and inflammatory cytokines have differential effects on the expression of integrins alpha 1 beta 1 and alpha 5 beta 1 by human dermal fibroblasts in vitro. J Cell Physiol 1996; 169:281-9. [PMID: 8908195 DOI: 10.1002/(sici)1097-4652(199611)169:2<281::aid-jcp7>3.0.co;2-k] [Citation(s) in RCA: 55] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Dermal fibroblasts are essential for the repair of cutaneous wounds. Fibroblasts presumably use cell surface receptors of the integrin family during migration into a wound from the adjacent uninjured tissue and for the subsequent matrix repairs. We have investigated the possible roles of platelet-derived growth factor and inflammatory cytokines in the regulation of integrin expression on wound fibroblasts using a porcine cutaneous wound model and cultured human cells. Tissue specimens collected from 4-day pig wounds were stained with antibodies specific for the alpha 1 and alpha 5 integrin subunits. Staining for alpha 1 was markedly decreased on fibroblasts adjacent to the wound and in the granulation tissue, while staining for alpha 5 was clearly enhanced in both locations. Normal adult human dermal fibroblasts in culture express the integrins alpha 1 beta 1, a collagen receptor, and alpha 5 beta 1 a fibronectin receptor. Quantitative flow cytometry was used to measure cell surface integrin expression after treatment with platelet-derived growth factor (PDGF)-AA, PDGF-AB, or PDGF BB. Each isoform of PDGF produced a significant decrease in the level of alpha 1 present on the cell surface and an increase in the level of alpha 5. Furthermore, PDGF-BB produced a corresponding decrease in alpha 1 mRNA and an increase in alpha 5 mRNA. In contrast, treatment with three inflammatory cytokines, IL-1 beta, TNF-alpha, and IFN-gamma, produced clear increases in the levels of alpha 1 and alpha 5 present on the cell surface. Our observations suggest that the differential effects of PDGF and inflammatory cytokines may be part of the mechanism regulating the expression of alpha 1 and alpha 5 integrins by dermal fibroblasts during wound repair.
Collapse
Affiliation(s)
- J Gailit
- Department of Dermatology, State University of New York at Stony Brook 11794, USA
| | | | | | | |
Collapse
|
41
|
Abstract
It is now evident that persistent overproduction of collagen and other connective tissue macromolecules results in excessive tissue deposition, and is responsible for the progressive nature of fibrosis in SSc. Up-regulation of collagen gene expression in SSc fibroblasts appears to be a critical event in the development of tissue fibrosis. The coordinate transcriptional activation of a number of extracellular matrix genes suggests a fundamental alteration in the regulatory control of gene expression in SSc fibroblasts. Trans-acting nuclear factors that bind to cis-acting elements in enhancer and promoter regions of the genes modulate the basal and inducible transcriptional activity of the collagen genes. The identity of the nuclear transcriptional factors that regulate normal collagen gene expression remains to be firmly established, and to date, no alterations in the level or in the activity of such DNA binding factors has been demonstrated in SSc fibroblasts. In addition to important interactions between fibroblasts and the extracellular matrix, cytokines and other cellular mediators can positively and negatively influence fibroblast collagen synthesis. Some of these signaling molecules may have physiologic roles, and their aberrant expression, or altered responsiveness of SSc fibroblasts to them, may result in the acquisition of the activated phenotype. The rapid expansion of knowledge regarding the effects of cytokines on extracellular matrix synthesis has led to an appreciation of the enormous complexity of regulatory networks that operate in the physiologic maintenance of connective tissue and which may be responsible for the occurrence of pathologic fibrosis. The ubiquitous growth factor TGF beta is the most potent inducer of collagen gene expression and connective tissue accumulation yet discovered. The expression of TGF beta in activated infiltrating mononuclear cells suggests a role for this cytokine as a mediator of fibroblast activation in SSc. Furthermore, the recognition that TGF beta is capable of inducing its own expression in a variety of cell types, coupled with the demonstration that a subpopulation of SSc dermal fibroblasts produces TGF beta, indicates the existence of a possible autocrine loop whereby lymphocyte-derived TGF beta in early SSc not only signals biosynthetic activation of fibroblasts in a paracrine manner, but autoinduces endogenous TGF beta production by the target fibroblasts themselves. Such an autocrine loop involving TGF beta may explain the persistent activation of collagen gene expression in SSc fibroblasts, and could be responsible for the progressive nature of fibrosis in SSc. Numerous other cytokines, as well as cell-matrix interactions, also modify collagen gene expression and can significantly influence the effects of TGF beta. Although their physiologic function in tissue remodeling or their involvement in abnormal fibrogenesis has not yet been conclusively demonstrated, the study of the biologic effects of these cytokines may provide important clues to understanding the pathogenesis of SSc, and to the development of rational drug therapy aimed at interrupting the abnormal fibrogenic process in this disease.
Collapse
Affiliation(s)
- S A Jimenez
- Jefferson Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | | | | |
Collapse
|
42
|
Varga J, Yufit T, Brown RR. Inhibition of collagenase and stromelysin gene expression by interferon-gamma in human dermal fibroblasts is mediated in part via induction of tryptophan degradation. J Clin Invest 1995; 96:475-81. [PMID: 7615820 PMCID: PMC185221 DOI: 10.1172/jci118058] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
The expression of the matrix-degrading enzymes collagenase and stromelysin is modulated by a variety of biologic and pharmacologic agents. IFN-gamma has potent effects on metalloproteinase production and therefore may play an important role in preventing excessive connective tissue degradation during inflammation and repair. We investigated the mechanisms of collagenase and stromelysin regulation by IFN-gamma in human dermal fibroblasts. IFN-gamma (300 U/ml) prevented the stimulation of metalloproteinase gene expression by IL-1 beta. In addition, incubation of fibroblasts with IFN-gamma resulted in a marked increase in cellular indoleamine 2,3-dioxygenase (IDO) mRNA, a > 90% depletion of tryptophan, and a corresponding > 30-fold increase in the tryptophan metabolite kynurenine in the culture media. Reducing the concentration of tryptophan from 25 microM to 0 markedly diminished the ability of fibroblasts to increase collagenase and stromelysin mRNA and collagenase production in response to IL-1 beta. Addition of exogenous tryptophan (25-50 micrograms/ml) to cultures that had been tryptophan depleted by pretreatment with IFN-gamma for 48 h restored the fibroblast response to IL-1 beta or PMA, but had no effect on IFN-gamma-induced HLA-DR alpha chain mRNA expression. These results indicate that inhibition of collagenase and stromelysin gene expression by IFN-gamma in fibroblasts is associated with activation of IDO and enhanced cellular tryptophan metabolism. Tryptophan degradation and ensuing tryptophan depletion may account, at least in part, for the inhibitory effect of IFN-gamma on metalloproteinase production in dermal fibroblasts.
Collapse
Affiliation(s)
- J Varga
- Department of Medicine, Jefferson Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania 19107, USA
| | | | | |
Collapse
|
43
|
Affiliation(s)
- S H Phan
- Department of Pathology, University of Michigan Medical School, Ann Arbor 48109-0602, USA
| |
Collapse
|
44
|
Affiliation(s)
- S Sollberg
- Department of Dermatology, University of Cologne, Germany
| | | | | | | |
Collapse
|
45
|
Görlach A, Herter P, Hentschel H, Frosch PJ, Acker H. Effects of nIFN beta and rIFN gamma on growth and morphology of two human melanoma cell lines: comparison between two- and three-dimensional culture. Int J Cancer 1994; 56:249-54. [PMID: 8314308 DOI: 10.1002/ijc.2910560218] [Citation(s) in RCA: 28] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
We compared the anti-proliferative effects of natural interferon beta (nIFN beta) and recombinant interferon gamma (rIFN gamma) on 2 human melanoma cell lines, IGRI and SK-Mel28, grown in 2-dimensional monolayer and in 3-dimensional spheroid culture. In monolayer culture, growth of both lines was inhibited in a dose-dependent manner by 5-day treatments with IFN in concentrations ranging between 1 and 5,000 IU/ml. Incubations with 120 IU/ml nIFN beta or 25 IU/ml rIFN gamma led to a 50% growth inhibition of IGRI cells. A 50% growth inhibition of SK-Me128 cells was obtained with 60 IU/ml nIFN beta, whereas even 5,000 IU/ml rIFN gamma inhibited the growth of this line by only 30%. Growing these melanoma cell lines in 3-dimensional spheroid culture for 5 days reduced their sensitivity to interferon. Growth inhibition values of 50% were achieved with 3,000 IU/ml rIFN gamma or 9,000 IU/ml nIFN beta for IGRI spheroids and 10,000 IU/ml nIFN beta for SK-Me128 spheroids, while 10,000 IU/ml rIFN gamma reduced the growth of SK-Me128 spheroids by only 25%. Outgrowth tests showed that the proliferative capacity after 5-day incubations with IFN was only reduced in IGRI spheroids treated with high doses of nIFN beta. The macroscopically observed increased density of interferon-treated spheroids could be confirmed by light microscopy as corresponding to reduced intercellular space in these spheroids. Scanning electron microscopy furthermore showed variations on the surface of IFN-treated spheroids as well as in cellular organization and structures between cells, hinting at a possible involvement of extracellular matrix substances in the reaction to interferons.(ABSTRACT TRUNCATED AT 250 WORDS)
Collapse
Affiliation(s)
- A Görlach
- Max-Planck Institute for Molecular Physiology, Dortmund, Germany
| | | | | | | | | |
Collapse
|
46
|
Berthod F, Hayek D, Damour O, Collombel C. Collagen synthesis by fibroblasts cultured within a collagen sponge. Biomaterials 1993; 14:749-54. [PMID: 8218724 DOI: 10.1016/0142-9612(93)90039-5] [Citation(s) in RCA: 84] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
We prepared a collagen sponge made of type I and III bovine collagen, glycosaminoglycans (GAG) and chitosan. Fibroblasts grown within the collagen sponge express a sixfold increase of their collagen synthesis, compared with fibroblasts embedded in a collagen gel. Moreover, collagen synthesis is twice as high in the collagen sponge than in a monolayer culture. The collagen sponge culture system promotes a dynamic model for us to perform studies on the regulations of collagen synthesis. Increased collagen production within the collagen sponge leads fibroblasts to reconstitute their own extracellular matrix, which should be more physiological than a bovine collagen gel.
Collapse
Affiliation(s)
- F Berthod
- Laboratoire des Substituts Cutanés, Hôpital Edouard Herriot, Lyon, France
| | | | | | | |
Collapse
|
47
|
Clark JG, Madtes DK, Raghu G. Effects of platelet-derived growth factor isoforms on human lung fibroblast proliferation and procollagen gene expression. Exp Lung Res 1993; 19:327-44. [PMID: 8319603 DOI: 10.3109/01902149309064350] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Platelet-derived growth factor (PDGF), a potent mitogen for fibroblasts, is a potentially important cytokine in the pathogenesis of fibroproliferative disorders of lung. Different isoforms of PDGF include a heterodimer composed of A and B chains and homodimers composed of A or B chains. The biological significance of the different isoforms is unknown, but they have been shown to differ in their mitogenic potency in some systems. Their effects on other fibroblast functions have not been fully examined. We undertook this study to determine the effect of PDGF isoforms on human lung fibroblast proliferation and procollagen synthesis. Cultured lung fibroblasts (IMR-90, WI-38, GeNA) were incubated in the presence of varying concentrations of highly purified PDGF-AB obtained from platelets or recombinant PDGF-AA or -BB homodimers. Incorporation of [3H]thymidine was determined as a measure of mitogenic activity. Fetal lung fibroblasts (IMR-90, WI-38) and an adult fibroblast strain (GeNA) responded similarly to the different isoforms, with maximum mitogenic activity observed at 5-10 ng/mL. Cell cycle analysis using three additional normal adult lung fibroblast strains indicated that all PDGF isoforms stimulated similar proportions of cells to cycle over a 7-day period. Fibroblast procollagen synthesis, measured after pulse labeling with [3H]proline, was not increased even at concentrations of the PDGF isoforms that were maximally mitogenic. Moreover, steady-state levels of alpha 1(I) and alpha 1(III) procollagen mRNA levels, determined by northern analysis and dot blot hybridization, were not changed after exposure to any of the PDGF isoforms. While all PDGF isoforms are potent mitogens for fetal and adult lung fibroblasts, it was concluded that they do not directly stimulate procollagen gene expression or procollagen synthesis in vitro. The results suggest that PDGF isoforms are potentially important mitogens for lung fibroblasts, but other factors are likely to be involved in the stimulation of fibroblast procollagen synthesis that is observed in fibroproliferative disorders of lung.
Collapse
Affiliation(s)
- J G Clark
- Pulmonary Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98104
| | | | | |
Collapse
|
48
|
Kakizaki Y, Kraft N, Atkins RC. Interferon-gamma stimulates the secretion of IL-1, but not of IL-6, by glomerular mesangial cells. Clin Exp Immunol 1993; 91:521-5. [PMID: 8383024 PMCID: PMC1554711 DOI: 10.1111/j.1365-2249.1993.tb05935.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
IL-1 activity in culture supernatant and cell lysate from rat mesangial cells stimulated with interferon-gamma (IFN-gamma) was measured by a thymocyte proliferation assay. While IFN-gamma alone had no effect on the secretion or the intracellular pool of IL-1, the enhancement by IFN-gamma of IL-1 secretion in response to lipopolysaccharide (LPS) was observed. The stimulatory effect of culture supernatant on thymocyte proliferation was abrogated by preincubation with the anti-IL-1 antibody. At least 4-h incubation with IFN-gamma and LPS was required to detect enhancing effect of IFN-gamma. The addition of as little as 1 U/ml IFN-gamma significantly increased IL-1 secretion in the presence of 10 micrograms/ml LPS. The IL-6 activity in culture supernatants was determined by measurement of thymidine uptake in mouse IL-6-dependent cell line (MH60.BSF2). Mesangial cells secreted IL-6 in culture supernatant without additional stimuli and LPS distinctly increased it as described previously. However, in contrast to IL-1 production, no effect of IFN-gamma on IL-6 secretion was observed in the presence or absence of LPS. Moreover, we determined whether enhanced IL-1 release is associated with Ia expression on mesangial cells. IFN-gamma alone and the combination with LPS induced marked expression of Ia antigen, whereas LPS alone did not. We conclude that IFN-gamma stimulates the production of IL-1, but not IL-6, by mesangial cells and suggest an important role of IFN-gamma in the pathogenesis of glomerulonephritis by regulating the mesangial production of IL-1 and the accessory cell function of mesangial cells.
Collapse
Affiliation(s)
- Y Kakizaki
- Monash Medical Centre, Monash University, Clayton, Victoria, Australia
| | | | | |
Collapse
|
49
|
Slack JL, Liska DJ, Bornstein P. Regulation of expression of the type I collagen genes. AMERICAN JOURNAL OF MEDICAL GENETICS 1993; 45:140-51. [PMID: 8456796 DOI: 10.1002/ajmg.1320450203] [Citation(s) in RCA: 89] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
The identification and functional analysis of DNA-protein interactions in the intronic and 5' flanking regions of the type I collagen genes has begun to define a series of cis-elements and trans-acting factors which regulate transcription of these genes. Studies such as these will eventually be expected to elucidate the mechanisms responsible for coordinate transcription of the alpha 1 and alpha 2 genes, a question which remains central to the field of collagen research. Although it is relatively straightforward to define sites of DNA-protein binding, interpretation of the functional importance of such interactions can be extremely complex. Furthermore, while mutation or deletion of a particular binding site may alter the functional activity of a construct transfected into cultured cells, there is no guarantee that a similar change will have the same effect in vivo, where the entire gene locus is present in its native chromosomal context. Nevertheless, these kinds of in vitro studies offer the best current approach to defining and isolating transcription factors that control expression of the alpha 1 and alpha 2 genes. Ultimately, it will be necessary to test the activity of such factors (and their respective cis-elements) in defined systems in vivo.
Collapse
Affiliation(s)
- J L Slack
- Department of Biochemistry and Medicine, University of Washington, Seattle 98195
| | | | | |
Collapse
|
50
|
Unemori EN, Bauer EA, Amento EP. Relaxin alone and in conjunction with interferon-gamma decreases collagen synthesis by cultured human scleroderma fibroblasts. J Invest Dermatol 1992; 99:337-42. [PMID: 1512471 DOI: 10.1111/1523-1747.ep12616665] [Citation(s) in RCA: 49] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Fibroblasts derived from the involved skin of scleroderma patients frequently display a phenotype of supernormal collagen expression when cultured. Fibroblasts displaying this phenotype derived from seven patients were treated with relaxin (1-100 ng/ml) and interferon-gamma (1-100 U/ml), individually and in combination, to assess the relative abilities of these cytokines to down-modulate collagen synthesis and secretion. Scleroderma fibroblasts displayed varying sensitivities to both relaxin and interferon-gamma. Relaxin (100 ng/ml) decreased expression of collagen by six of seven lines tested from 8 to 59% compared to untreated cultures. Interferon-gamma (100 U/ml) depressed collagen secretion by all seven lines in a range from 7 to 89%. When relaxin and interferon-gamma were used in combination, relaxin augmented IFN-gamma-induced decreases in collagen secretion in four of seven lines. In three of these lines, the use of relaxin in conjunction with suboptimal doses of interferon-gamma resulted in decreases equivalent to or greater than that seen with a tenfold higher concentration of interferon-gamma. This study demonstrates the ability of relaxin to directly alter the excessive collagen-producing phenotype of scleroderma fibroblasts. In addition, in some cases, combining relaxin and interferon-gamma resulted in a cooperative effect in decreasing collagen expression by scleroderma cells in vitro.
Collapse
Affiliation(s)
- E N Unemori
- Inflammation, Bone and Connective Tissue Research, Genentech, Inc., South San Francisco, CA 94080
| | | | | |
Collapse
|