51
|
Eurlings IMJ, Reynaert NL, van de Wetering C, Aesif SW, Mercken EM, de Cabo R, van der Velden JL, Janssen-Heininger YM, Wouters EFM, Dentener MA. Involvement of c-Jun N-Terminal Kinase in TNF-α-Driven Remodeling. Am J Respir Cell Mol Biol 2017; 56:393-401. [PMID: 27875656 DOI: 10.1165/rcmb.2015-0195oc] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Lung tissue remodeling in chronic obstructive pulmonary disease (COPD) is characterized by airway wall thickening and/or emphysema. Although the bronchial and alveolar compartments are functionally independent entities, we recently showed comparable alterations in matrix composition comprised of decreased elastin content and increased collagen and hyaluronan contents of alveolar and small airway walls. Out of several animal models tested, surfactant protein C (SPC)-TNF-α mice showed remodeling in alveolar and airway walls similar to what we observed in patients with COPD. Epithelial cells are able to undergo a phenotypic shift, gaining mesenchymal properties, a process in which c-Jun N-terminal kinase (JNK) signaling is involved. Therefore, we hypothesized that TNF-α induces JNK-dependent epithelial plasticity, which contributes to lung matrix remodeling. To this end, the ability of TNF-α to induce a phenotypic shift was assessed in A549, BEAS2B, and primary bronchial epithelial cells, and phenotypic markers were studied in SPC-TNF-α mice. Phenotypic markers of mesenchymal cells were elevated both in vitro and in vivo, as shown by the expression of vimentin, plasminogen activator inhibitor-1, collagen, and matrix metalloproteinases. Concurrently, the expression of the epithelial markers, E-cadherin and keratin 7 and 18, was attenuated. A pharmacological inhibitor of JNK attenuated this phenotypic shift in vitro, demonstrating involvement of JNK signaling in this process. Interestingly, activation of JNK signaling was also clearly present in lungs of SPC-TNF-α mice and patients with COPD. Together, these data show a role for TNF-α in the induction of a phenotypic shift in vitro, resulting in increased collagen production and the expression of elastin-degrading matrix metalloproteinases, and provide evidence for involvement of the TNF-α-JNK axis in extracellular matrix remodeling.
Collapse
Affiliation(s)
- Irene M J Eurlings
- 1 Department of Respiratory Medicine, School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Niki L Reynaert
- 1 Department of Respiratory Medicine, School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Cheryl van de Wetering
- 1 Department of Respiratory Medicine, School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Scott W Aesif
- 2 Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota
| | - Evi M Mercken
- 3 Experimental Gerontology Section, Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, Maryland; and
| | - Rafael de Cabo
- 3 Experimental Gerontology Section, Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, Maryland; and
| | - Jos L van der Velden
- 4 Department of Pathology, University of Vermont College of Medicine, Burlington, Vermont
| | | | - Emiel F M Wouters
- 1 Department of Respiratory Medicine, School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Mieke A Dentener
- 1 Department of Respiratory Medicine, School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, Maastricht, the Netherlands
| |
Collapse
|
52
|
Abstract
Increased levels of tumor necrosis factor (TNF) α have been linked to a number of pulmonary inflammatory diseases including asthma, chronic obstructive pulmonary disease (COPD), acute lung injury (ALI)/acute respiratory distress syndrome (ARDS), sarcoidosis, and interstitial pulmonary fibrosis (IPF). TNFα plays multiple roles in disease pathology by inducing an accumulation of inflammatory cells, stimulating the generation of inflammatory mediators, and causing oxidative and nitrosative stress, airway hyperresponsiveness and tissue remodeling. TNFα-targeting biologics, therefore, present a potentially highly efficacious treatment option. This review summarizes current knowledge on the role of TNFα in pulmonary disease pathologies, with a focus on the therapeutic potential of TNFα-targeting agents in treating inflammatory lung diseases.
Collapse
Affiliation(s)
- Rama Malaviya
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ, USA
| | - Jeffrey D Laskin
- Department of Environmental and Occupational Health, School of Public Health, Rutgers University, Piscataway, NJ, USA
| | - Debra L Laskin
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ, USA.
| |
Collapse
|
53
|
Li H, Hao Y, Zhang H, Ying W, Li D, Ge Y, Ying B, Cheng B, Lian Q, Jin S. Posttreatment with Protectin DX ameliorates bleomycin-induced pulmonary fibrosis and lung dysfunction in mice. Sci Rep 2017; 7:46754. [PMID: 28466866 PMCID: PMC5413938 DOI: 10.1038/srep46754] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Accepted: 03/21/2017] [Indexed: 02/06/2023] Open
Abstract
Protectin DX (10S,17S-dihydroxydocosa-4Z,7Z,11E,13Z,15E,19Z-hexaenoic acid) (PDX), generated from Ω-3 fatty docosahexaenoic acids, is believed to exert anti-inflammatory and proresolution bioactions. To date, few studies have been performed regarding its effect on pulmonary fibrosis. Herein we show that PDX exerts a potential therapeutic effect which is distinct from its anti-inflammation and pro-resolution activity on mice with pulmonary fibrosis. In the present study, we showed that bleomycin (BLM) increased inflammatory infiltration, collagen deposition, and lung dysfunction on day7 after challenged in mice. Posttreatment with PDX ameliorated BLM-induced inflammatory responses, extracellular matrix (ECM) deposition and the level of cytokines related to fibrosis as evaluated by histology analysis, transformation electron microscope (TEM), lung hydroxyproline content and cytokines test. Moreover, PDX improved lung respiratory function, remedied BLM-induced hypoxemia and prolonged life span. In addition, we found that PDX reversed epithelial–mesenchymal transition (EMT) phenotypic transformation in vivo and in vitro, reinforcing a potential mechanism of promoting fibrosis resolution. In summary, our findings showed that posttreatment with PDX could ameliorate BLM-induced pulmonary fibrosis and lung dysfunction in mice and PDX may be considered as a promising therapeutic approached to fibrotic lung diseases.
Collapse
Affiliation(s)
- Hui Li
- Department of Anesthesia and Critical Care, Second Affiliated Hospital of Wenzhou Medical University, Zhejiang 325027, China
| | - Yu Hao
- Department of Anesthesia and Critical Care, Second Affiliated Hospital of Wenzhou Medical University, Zhejiang 325027, China
| | - Huawei Zhang
- Department of Anesthesia and Critical Care, Second Affiliated Hospital of Wenzhou Medical University, Zhejiang 325027, China
| | - Weiyang Ying
- Department of Anesthesia and Critical Care, Second Affiliated Hospital of Wenzhou Medical University, Zhejiang 325027, China
| | - Dan Li
- Department of Anesthesia and Critical Care, Second Affiliated Hospital of Wenzhou Medical University, Zhejiang 325027, China
| | - Yahe Ge
- Department of Anesthesia and Critical Care, Second Affiliated Hospital of Wenzhou Medical University, Zhejiang 325027, China
| | - Binyu Ying
- Department of Anesthesia and Critical Care, Second Affiliated Hospital of Wenzhou Medical University, Zhejiang 325027, China
| | - Bihuan Cheng
- Department of Anesthesia and Critical Care, Second Affiliated Hospital of Wenzhou Medical University, Zhejiang 325027, China
| | - Qingquan Lian
- Department of Anesthesia and Critical Care, Second Affiliated Hospital of Wenzhou Medical University, Zhejiang 325027, China
| | - Shengwei Jin
- Department of Anesthesia and Critical Care, Second Affiliated Hospital of Wenzhou Medical University, Zhejiang 325027, China
| |
Collapse
|
54
|
Chen J, Chi S, Li F, Yang J, Cho WC, Liu X. Biologics-induced interstitial lung diseases in rheumatic patients: facts and controversies. Expert Opin Biol Ther 2017; 17:265-283. [PMID: 28117616 DOI: 10.1080/14712598.2017.1287169] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
INTRODUCTION Interstitial lung disease (ILD) is a common, devastating pulmonary complication. An increased number of reports suggesting that biological disease modifying antirheumatic drugs (DMARDs) induced or exacerbated ILDs in rheumatoid arthritis (RA) patients has garnered increased attention. Areas covered: This article discusses ILDs induced by or exacerbated during biological therapy in RA patients. The article summarizes the efficacy and safety of a variety of licensed and off-label biologics clinically used for rheumatic diseases, focusing on the onset or exacerbation of RA-associated ILDs (RA-ILDs) in RA patients treated with biologics targeting tumor necrosis factor, CD20, interleukin 1 (IL-1) and IL-6 receptors. Additionally, the pathogenesis of RA-ILDs is discussed. Expert opinion: To some extent, the possibility of biologic-induced RA-ILDs increases the difficulty in choosing an optimal regimen for RA treatment with biological agents, as the relationship between biological therapy safety and the induction or exacerbation of RA-ILDs has not been established. A framework to assess baseline disease severity, particularly standardizing the evaluation of the pulmonary condition stage in RA patients and monitoring the outcome during the biological therapy treatment, is highly needed and may substantially help guide treatment decisions and predict the treatment benefits.
Collapse
Affiliation(s)
- Juan Chen
- a Department of Pulmonary and Critical Care Medicine , General Hospital of Ningxia Medical University , Yinchuan , China
| | - Shuhong Chi
- b Department of Rheumatology , General Hospital of Ningxia Medical University , Yinchuan , China
| | - Feng Li
- c Center of Laboratory Medicine , General Hospital of Ningxia Medical University , Yinchuan , Ningxia , China
| | - Jiali Yang
- c Center of Laboratory Medicine , General Hospital of Ningxia Medical University , Yinchuan , Ningxia , China.,d Human Stem Cell Institute , General Hospital of Ningxia Medical University , Yinchuan , Ningxia , China
| | - William C Cho
- e Department of Clinical Oncology , Queen Elizabeth Hospital , Kowloon , Hong Kong
| | - Xiaoming Liu
- c Center of Laboratory Medicine , General Hospital of Ningxia Medical University , Yinchuan , Ningxia , China.,d Human Stem Cell Institute , General Hospital of Ningxia Medical University , Yinchuan , Ningxia , China
| |
Collapse
|
55
|
Cooke KR, Luznik L, Sarantopoulos S, Hakim FT, Jagasia M, Fowler DH, van den Brink MRM, Hansen JA, Parkman R, Miklos DB, Martin PJ, Paczesny S, Vogelsang G, Pavletic S, Ritz J, Schultz KR, Blazar BR. The Biology of Chronic Graft-versus-Host Disease: A Task Force Report from the National Institutes of Health Consensus Development Project on Criteria for Clinical Trials in Chronic Graft-versus-Host Disease. Biol Blood Marrow Transplant 2017; 23:211-234. [PMID: 27713092 PMCID: PMC6020045 DOI: 10.1016/j.bbmt.2016.09.023] [Citation(s) in RCA: 280] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2016] [Accepted: 09/30/2016] [Indexed: 12/12/2022]
Abstract
Chronic graft-versus-host disease (GVHD) is the leading cause of late, nonrelapse mortality and disability in allogeneic hematopoietic cell transplantation recipients and a major obstacle to improving outcomes. The biology of chronic GVHD remains enigmatic, but understanding the underpinnings of the immunologic mechanisms responsible for the initiation and progression of disease is fundamental to developing effective prevention and treatment strategies. The goals of this task force review are as follows: This document is intended as a review of our understanding of chronic GVHD biology and therapies resulting from preclinical studies, and as a platform for developing innovative clinical strategies to prevent and treat chronic GVHD.
Collapse
Affiliation(s)
- Kenneth R Cooke
- Department of Oncology, Sidney Kimmel Cancer Center at Johns Hopkins Hospital, Baltimore, Maryland.
| | - Leo Luznik
- Department of Oncology, Sidney Kimmel Cancer Center at Johns Hopkins Hospital, Baltimore, Maryland
| | - Stefanie Sarantopoulos
- Division of Hematological Malignancies and Cellular Therapy, Department of Immunology and Duke Cancer Institute, Duke University, Durham, North Carolina
| | - Frances T Hakim
- Experimental Transplantation and Immunology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Madan Jagasia
- Division of Hematology-Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Daniel H Fowler
- Experimental Transplantation and Immunology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Marcel R M van den Brink
- Departments of Immunology and Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - John A Hansen
- Division of Clinical Research, Fred Hutchinson Cancer Research Center, Department of Medicine, University of Washington, Seattle, Washington
| | - Robertson Parkman
- Division of Pediatric Stem Cell Transplantation and Regenerative Medicine, Stanford University, Palo Alto, California
| | - David B Miklos
- Division of Blood and Marrow Transplantation, Stanford University, Palo Alto, California
| | - Paul J Martin
- Division of Clinical Research, Fred Hutchinson Cancer Research Center, Department of Medicine, University of Washington, Seattle, Washington
| | - Sophie Paczesny
- Departments of Pediatrics and Immunology, Wells Center for Pediatric Research, Melvin and Bren Simon Cancer Center, Indiana University School of Medicine, Indianapolis, Indiana
| | - Georgia Vogelsang
- Department of Oncology, Sidney Kimmel Cancer Center at Johns Hopkins Hospital, Baltimore, Maryland
| | - Steven Pavletic
- Experimental Transplantation and Immunology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Jerome Ritz
- Division of Hematologic Malignancies, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Kirk R Schultz
- Michael Cuccione Childhood Cancer Research Program, Department of Pediatrics, BC Children's Hospital, University of British Columbia, Vancouver, British Columbia, Canada.
| | - Bruce R Blazar
- Masonic Cancer Center and Department of Pediatrics, Division of Blood and Marrow Transplantation, University of Minnesota, Minneapolis, Minnesota.
| |
Collapse
|
56
|
Qin Y, Li S, Zhao G, Fu X, Xie X, Huang Y, Cheng X, Wei J, Liu H, Lai Z. Long-term intravenous administration of carboxylated single-walled carbon nanotubes induces persistent accumulation in the lungs and pulmonary fibrosis via the nuclear factor-kappa B pathway. Int J Nanomedicine 2016; 12:263-277. [PMID: 28115845 PMCID: PMC5221802 DOI: 10.2147/ijn.s123839] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Numerous studies have demonstrated promising application of single-walled carbon nanotubes (SWNTs) in drug delivery, diagnosis, and targeted therapy. However, the adverse health effects resulting from intravenous injection of SWNTs are not completely understood. Studies have shown that levels of “pristine” or carboxylated carbon nanotubes are very high in mouse lungs after intravenous injection. We hypothesized that long-term and repeated intravenous administration of carboxylated SWNTs (c-SWNTs) can result in persistent accumulation and induce histopathologic changes in rat lungs. Here, c-SWNTs were administered repeatedly to rats via tail-vein injection for 90 days. Long-term intravenous injection of c-SWNTs caused sustained embolization in lung capillaries and granuloma formation. It also induced a persistent inflammatory response that was regulated by the nuclear factor-kappa B signaling pathway, and which resulted in pulmonary fibrogenesis. c-SWNTs trapped within lung capillaries traversed capillary walls and injured alveolar epithelial cells, thereby stimulating production of pro-inflammatory cytokines (tumor necrosis factor-alpha and interleukin-1 beta) and pro-fibrotic growth factors (transforming growth factor-beta 1). Protein levels of type-I and type-III collagens, matrix metalloproteinase-2, and the tissue inhibitor of metalloproteinase-2 were upregulated after intravenous exposure to c-SWNTs as determined by immunohistochemical assays and Western blotting, which suggested collagen deposition and remodeling of the extracellular matrix. These data suggest that chronic and cumulative toxicity of nanomaterials to organs with abundant capillaries should be assessed if such nanomaterials are applied via intravenous administration.
Collapse
Affiliation(s)
- Yue Qin
- Pharmaceutical College, Guangxi Medical University
| | - Suning Li
- Department of Pharmacy, The Maternal and Child Health Hospital of Guangxi Zhuang Autonomous Region
| | - Gan Zhao
- Department of Pharmacy, The Maternal and Child Health Hospital of Guangxi Zhuang Autonomous Region
| | - Xuanhao Fu
- Pharmaceutical College, Guangxi Medical University
| | - Xueping Xie
- Pharmaceutical College, Guangxi Medical University
| | - Yiyi Huang
- Pharmaceutical College, Guangxi Medical University
| | - Xiaojing Cheng
- Life Sciences Institute, Guangxi Medical University, Nanning, Guangxi, People's Republic of China
| | - Jinbin Wei
- Pharmaceutical College, Guangxi Medical University
| | - Huagang Liu
- Pharmaceutical College, Guangxi Medical University
| | - Zefeng Lai
- Pharmaceutical College, Guangxi Medical University
| |
Collapse
|
57
|
Dong J, Ma Q. Myofibroblasts and lung fibrosis induced by carbon nanotube exposure. Part Fibre Toxicol 2016; 13:60. [PMID: 27814727 PMCID: PMC5097370 DOI: 10.1186/s12989-016-0172-2] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Accepted: 10/25/2016] [Indexed: 01/07/2023] Open
Abstract
Carbon nanotubes (CNTs) are newly developed materials with unique properties and a range of industrial and commercial applications. A rapid expansion in the production of CNT materials may increase the risk of human exposure to CNTs. Studies in rodents have shown that certain forms of CNTs are potent fibrogenic inducers in the lungs to cause interstitial, bronchial, and pleural fibrosis characterized by the excessive deposition of collagen fibers and the scarring of involved tissues. The cellular and molecular basis underlying the fibrotic response to CNT exposure remains poorly understood. Myofibroblasts are a major type of effector cells in organ fibrosis that secrete copious amounts of extracellular matrix proteins and signaling molecules to drive fibrosis. Myofibroblasts also mediate the mechano-regulation of fibrotic matrix remodeling via contraction of their stress fibers. Recent studies reveal that exposure to CNTs induces the differentiation of myofibroblasts from fibroblasts in vitro and stimulates pulmonary accumulation and activation of myofibroblasts in vivo. Moreover, mechanistic analyses provide insights into the molecular underpinnings of myofibroblast differentiation and function induced by CNTs in the lungs. In view of the apparent fibrogenic activity of CNTs and the emerging role of myofibroblasts in the development of organ fibrosis, we discuss recent findings on CNT-induced lung fibrosis with emphasis on the role of myofibroblasts in the pathologic development of lung fibrosis. Particular attention is given to the formation and activation of myofibroblasts upon CNT exposure and the possible mechanisms by which CNTs regulate the function and dynamics of myofibroblasts in the lungs. It is evident that a fundamental understanding of the myofibroblast and its function and regulation in lung fibrosis will have a major influence on the future research on the pulmonary response to nano exposure, particle and fiber-induced pneumoconiosis, and other human lung fibrosing diseases.
Collapse
Affiliation(s)
- Jie Dong
- Receptor Biology Laboratory, Toxicology and Molecular Biology Branch, Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Centers for Disease Control and Prevention, 1095 Willowdale Road, Morgantown, WV, USA
| | - Qiang Ma
- Receptor Biology Laboratory, Toxicology and Molecular Biology Branch, Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Centers for Disease Control and Prevention, 1095 Willowdale Road, Morgantown, WV, USA.
| |
Collapse
|
58
|
Picchianti Diamanti A, Markovic M, Argento G, Giovagnoli S, Ricci A, Laganà B, D'Amelio R. Therapeutic management of patients with rheumatoid arthritis and associated interstitial lung disease: case report and literature review. Ther Adv Respir Dis 2016; 11:64-72. [PMID: 27733490 PMCID: PMC5941974 DOI: 10.1177/1753465816668780] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Rheumatoid arthritis (RA) is an inflammatory autoimmune disease that can present
different extrarticular manifestations involving heart, lungs and kidneys. In
recent years there has been a growing awareness of the central role played by
the lungs in the onset and progression of RA. In particular interstitial lung
disease (ILD) is a common pulmonary manifestation that may be related to the
inflammatory process itself, infectious complications and to the treatments
used. Management of patients with ILD/RA is still a challenge for clinicians,
both synthetic [mainly methotrexate (MTX), leflunomide] and biologic
immunosuppressors [mainly anti-tumor necrosis factor (TNF)α] have in fact been
related to the onset or worsening of lung diseases with conflicting data. Here
we report the case of a 61-year-old male patient with severely active early RA,
previously treated with MTX, who developed subacute ILD, along with a review of
ILD/RA topic. Tocilizumab (humanized monoclonal antibody against the
interleukin-6 receptor) was introduced on the basis of its effectiveness in RA
without concomitant MTX and the ability to overcome the profibrotic effects of
interleukin (IL)-6. After 3 months of treatment the clinical condition of the
patient strongly improved until it reached low disease activity. He no longer
complained of cough and dyspnea and bilateral basal crackles were no more
present. Considering its distinctive features, tocilizumab, in such a
challenging clinical condition, appears to be a safe and effective therapy, thus
it enables RA remission without deteriorating ILD, at 1-year follow up, as
confirmed by ultrasonography of the affected joints and chest high-resolution
computed tomography (HRCT).
Collapse
Affiliation(s)
- Andrea Picchianti Diamanti
- Department of Clinical and Molecular Medicine, School of Medicine and Psychology, 'Sapienza' University of Rome, Sant'Andrea University Hospital, Via di Grottarossa 1039, Rome 00189, Italy
| | - Milica Markovic
- Department of Clinical and Molecular Medicine, School of Medicine and Psychology, 'Sapienza' University of Rome, Sant'Andrea University Hospital, Rome, Italy
| | - Giuseppe Argento
- Radiology Unit, School of Medicine and Psychology, "Sapienza" University of Rome, Sant'Andrea University Hospital, Rome, Italy
| | - Simonetta Giovagnoli
- Department of Clinical and Molecular Medicine, School of Medicine and Psychology, 'Sapienza' University of Rome, Sant'Andrea University Hospital, Rome, Italy
| | - Alberto Ricci
- Department of Clinical and Molecular Medicine, School of Medicine and Psychology, 'Sapienza' University of Rome, Sant'Andrea University Hospital, Rome, Italy
| | - Bruno Laganà
- Department of Clinical and Molecular Medicine, School of Medicine and Psychology, 'Sapienza' University of Rome, Sant'Andrea University Hospital, Rome, Italy
| | - Raffaele D'Amelio
- Department of Clinical and Molecular Medicine, School of Medicine and Psychology, 'Sapienza' University of Rome, Sant'Andrea University Hospital, Rome, Italy
| |
Collapse
|
59
|
Lipsa D, Leva P, Barrero-Moreno J, Coelhan M. Inflammatory effects induced by selected limonene oxidation products: 4-OPA, IPOH, 4-AMCH in human bronchial (16HBE14o-) and alveolar (A549) epithelial cell lines. Toxicol Lett 2016; 262:70-79. [PMID: 27575568 DOI: 10.1016/j.toxlet.2016.08.023] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Revised: 08/24/2016] [Accepted: 08/25/2016] [Indexed: 12/24/2022]
Abstract
Limonene, a monoterpene abundantly present in most of the consumer products (due to its pleasant citrus smell), easily undergoes ozonolysis leading to several limonene oxidation products (LOPs) such as 4-acetyl-1-methylcyclohexene (4-AMCH), 4-oxopentanal (4-OPA) and 3-isopropenyl-6-oxoheptanal (IPOH). Toxicological studies have indicated that human exposure to limonene and ozone can cause adverse airway effects. However, little attention has been paid to the potential health impact of specific LOPs, in particular of IPOH, 4-OPA and 4-AMCH. This study evaluates the cytotoxic effects of the selected LOPs on human bronchial epithelial (16HBE14o-) and alveolar epithelial (A549) cell lines by generating concentration-response curves using the neutral red uptake assay and analyzing the inflammatory response with a series of cytokines/chemokines. The cellular viability was mostly reduced by 4-OPA [IC50=1.6mM (A549) and 1.45mM (16HBE14o-)] when compared to IPOH [IC50=3.5mM (A549) and 3.4mM (16HBE14o-)] and 4-AMCH [IC50 could not be calculated]. As a result from the inflammatory response, IPOH [50μM] induced an increase of both IL-6 and IL-8 secretion in A549 (1.5-fold change) and in 16HBE14o- (2.8- and 7-fold change respectively). 4-OPA [50μM] treatment of A549 increased IL-6 (1.4-times) and IL-8 (1.3-times) levels, while in 16HBE14o- had an opposite effect. A549 treated with 4-AMCH [50μM] elevate both IL-6 and IL-8 levels by 1.2-times, while in 16HBE14o- had an opposite effect. Based on our results, lung cellular injury characterized by inflammatory cytokine release was observed for both cell lines treated with the selected chemicals at concentrations that did not affect their cellular viability.
Collapse
Affiliation(s)
- Dorelia Lipsa
- Technische Universität München, Research Center Weihenstephan for Brewing and Food Quality, Alte Akademie 3, Freising-Weihenstephan, Germany; European Commission, Joint Research Centre, Institute for Health and Consumer Protection, Chemical Assessment and Testing Unit, Ispra (VA), Italy.
| | - Paolo Leva
- European Commission, Joint Research Centre, Institute for Health and Consumer Protection, Chemical Assessment and Testing Unit, Ispra (VA), Italy
| | - Josefa Barrero-Moreno
- European Commission, Joint Research Centre, Institute for Health and Consumer Protection, Chemical Assessment and Testing Unit, Ispra (VA), Italy
| | - Mehmet Coelhan
- Technische Universität München, Research Center Weihenstephan for Brewing and Food Quality, Alte Akademie 3, Freising-Weihenstephan, Germany
| |
Collapse
|
60
|
Fujita M, Ouchi H, Ikegame S, Harada E, Matsumoto T, Uchino J, Nakanishi Y, Watanabe K. Critical role of tumor necrosis factor receptor 1 in the pathogenesis of pulmonary emphysema in mice. Int J Chron Obstruct Pulmon Dis 2016; 11:1705-12. [PMID: 27555760 PMCID: PMC4968668 DOI: 10.2147/copd.s108919] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
COPD is a major cause of chronic morbidity and mortality throughout the world. Although tumor necrosis factor-α (TNF-α) has a critical role in the development of COPD, the role of different TNF receptors (TNFRs) in pulmonary emphysema has not been resolved. We aimed to clarify the role of TNFRs in the development of pulmonary emphysema. TNF-α transgenic mice, a murine model of COPD in which the mice spontaneously develop emphysema with a large increase in lung volume and pulmonary hypertension, were crossed with either TNFR1-deficient mice or TNFR2-deficient mice. After 6 months, the gross appearance of the lung, lung histology, and pulmonary and cardiac physiology were determined. In addition, the relationship between apoptosis and emphysema was investigated. Pulmonary emphysema-like changes disappeared with deletion of TNFR1. However, slight improvements were attained with deletion of TNFR2. Apoptotic cells in the interstitium of the lung were observed in TNF-α transgenic mice. The apoptotic signals through TNFR1 appear critical for the pathogenesis of pulmonary emphysema. In contrast, the inflammatory process has a less important role for the development of emphysema.
Collapse
Affiliation(s)
- Masaki Fujita
- Department of Respiratory Medicine, Faculty of Medicine, Fukuoka University
| | - Hiroshi Ouchi
- Research Institute for Diseases of the Chest, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Satoshi Ikegame
- Research Institute for Diseases of the Chest, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Eiji Harada
- Research Institute for Diseases of the Chest, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Takemasa Matsumoto
- Department of Respiratory Medicine, Faculty of Medicine, Fukuoka University
| | - Junji Uchino
- Department of Respiratory Medicine, Faculty of Medicine, Fukuoka University
| | - Yoichi Nakanishi
- Research Institute for Diseases of the Chest, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Kentaro Watanabe
- Department of Respiratory Medicine, Faculty of Medicine, Fukuoka University
| |
Collapse
|
61
|
Bawadekar M, Gendron-Fitzpatrick A, Rebernick R, Shim D, Warner TF, Nicholas AP, Lundblad LKA, Thompson PR, Shelef MA. Tumor necrosis factor alpha, citrullination, and peptidylarginine deiminase 4 in lung and joint inflammation. Arthritis Res Ther 2016; 18:173. [PMID: 27450561 PMCID: PMC4957385 DOI: 10.1186/s13075-016-1068-0] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2016] [Accepted: 06/29/2016] [Indexed: 01/08/2023] Open
Abstract
Background The relationship between lung and joint inflammation in rheumatoid arthritis is poorly understood. Lung inflammation with resultant protein citrullination may trigger anti-citrullinated protein antibodies, inflammation, and arthritis. Alternatively, lung and joint inflammation may be two manifestations of a single underlying pathology. The lung has increased citrullination and TNF-α levels are high in rheumatoid arthritis; however, it is unknown if TNF-α can induce lung protein citrullination. The citrullinating enzyme peptidylarginine deiminase 4 (PAD4) exacerbates TNF-α-induced arthritis, but a role for PAD4 in lung citrullination and TNF-α-induced lung inflammation has not been explored. Our aim was to use TNF-α-overexpressing mice to clarify the intersection of TNF-α, citrullination, PAD4, arthritis, and lung inflammation. Methods Lung protein citrullination in wild-type mice, mice that overexpress TNF-α systemically (TNF+), TNF+PAD4+/+, and TNF+PAD4-/- mice was quantified by both gel electrophoresis using a citrulline probe and western blot. Hematoxylin and eosin (H&E)-stained lung sections from TNF+PAD4+/+ and TNF+PAD4-/- mice were scored for lung inflammation. H&E-stained ankle joint sections from mice that overexpress TNF-α only in the lungs were assessed for arthritis. Results TNF+ mice have increased lung protein citrullination. TNF+PAD4-/- mice do not have significantly reduced lung protein citrullination, but do have decreased lung inflammation compared to TNF+PAD4+/+ mice. Mice that overexpress TNF-α only in the lungs do not develop arthritis. Conclusions PAD4 exacerbates lung inflammation downstream of TNF-α without having a major role in generalized protein citrullination in inflamed lungs. Also, TNF-α-induced lung inflammation is not sufficient to drive murine arthritis. Electronic supplementary material The online version of this article (doi:10.1186/s13075-016-1068-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Mandar Bawadekar
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Annette Gendron-Fitzpatrick
- Research Animal Resource Center Comparative Pathology Lab and Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Ryan Rebernick
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Daeun Shim
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Thomas F Warner
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Anthony P Nicholas
- Department of Neurology and Center for Neurodegeneration and Experimental Therapeutics, University of Alabama at Birmingham and Birmingham VA Medical Center, Birmingham, AL, USA
| | | | - Paul R Thompson
- Departments of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, MA, USA
| | - Miriam A Shelef
- Department of Medicine, University of Wisconsin-Madison and William S. Middleton Memorial Veterans Hospital, Madison, WI, USA.
| |
Collapse
|
62
|
Liu L, Geng X, McDermott J, Shen J, Corbin C, Xuan S, Kim J, Zuo L, Liu Z. Copper Deficiency in the Lungs of TNF-α Transgenic Mice. Front Physiol 2016; 7:234. [PMID: 27378943 PMCID: PMC4906028 DOI: 10.3389/fphys.2016.00234] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Accepted: 05/30/2016] [Indexed: 12/27/2022] Open
Abstract
Tumor necrosis factor (TNF)-α is a well-known pro-inflammatory cytokine. Increased expression of Tnf-α is a feature of inflammatory lung diseases, such as asthma, emphysema, fibrosis, and smoking-induced chronic obstructive pulmonary disease (COPD). Using a mouse line with lung-specific Tnf-α overexpression (SPC-TNF-α) to mimic TNF-α-associated lung diseases, we investigated the role of chronic inflammation in the homeostasis of lung trace elements. We performed a quantitative survey of micronutrients and biometals, including copper (Cu), zinc (Zn), and selenium (Se), in the transgenic mice tissues. We also examined the expression of Cu-dependent proteins in the inflammatory lung tissue to determine whether they were affected by the severe Cu deficiency, including cuproenzymes, Cu transporters, and Cu chaperones. We found consistent lung-specific reduction of the metal Cu, with a mean decrease of 70%; however, Zn and Se were unaffected in all other tissues. RT-PCR showed that two Cu enzymes associated with lung pathology were downregulated: amine oxidase, Cu containing 3 (Aoc3) and lysyl oxidase (Lox). Two factors, vascular endothelial growth factor (Vegf) and focal adhesion kinase (Fak), related with Cu deficiency treatment, showed decreased expression in the transgenic inflammatory lung. We concluded that Cu deficiency occurs following chronic TNF-α-induced lung inflammation and this likely plays an essential role in the inflammation-induced lung damage. These results suggest the restoration of lung Cu status as a potential strategy in both treatment and prevention of chronic lung inflammation and related disorders.
Collapse
Affiliation(s)
- Liu Liu
- Department of Biological Sciences, Oakland University Rochester, MI, USA
| | - Xiangrong Geng
- Department of Biological Sciences, Oakland University Rochester, MI, USA
| | - Joseph McDermott
- Department of Biological Sciences, Oakland University Rochester, MI, USA
| | - Jian Shen
- Department of Pathology, Creighton University School of Medicine Omaha, NE, USA
| | - Cody Corbin
- Department of Biological Sciences, Oakland University Rochester, MI, USA
| | - Stephanie Xuan
- Department of Biological Sciences, Oakland University Rochester, MI, USA
| | - Jae Kim
- Department of Biological Sciences, Oakland University Rochester, MI, USA
| | - Li Zuo
- Radiologic Sciences and Respiratory Therapy Division, School of Health and Rehabilitation Sciences, The Ohio State University College of Medicine, The Ohio State University Wexner Medical Center Columbus, OH, USA
| | - Zijuan Liu
- Department of Biological Sciences, Oakland University Rochester, MI, USA
| |
Collapse
|
63
|
Lee SJ, Park JH, Lee JY, Jeong YJ, Song JA, Lee K, Kim DJ. Establishment of a mouse model for pulmonary inflammation and fibrosis by intratracheal instillation of polyhexamethyleneguanidine phosphate. J Toxicol Pathol 2016; 29:95-102. [PMID: 27182113 PMCID: PMC4866002 DOI: 10.1293/tox.2015-0067] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Accepted: 12/17/2015] [Indexed: 12/18/2022] Open
Abstract
Although several animal models have been developed to study human pulmonary fibrosis, lack of a perfect model has raised the need for various animal models of pulmonary fibrosis. In this study, we evaluated the pulmonary effect of polyhexamethyleneguanidine phosphate instillation into the lungs of mice to determine the potential of these mice as a murine model of pulmonary fibrosis. Intratracheal instillation of polyhexamethyleneguanidine phosphate induced severe lung inflammation manifested by the infiltration of mononuclear cells and neutrophils and increased production of IL-6, TNF-α, CCL2 and CXCL1. The lung inflammation gradually increased until 28 days after polyhexamethyleneguanidine phosphate exposure, and increases of collagen deposition and TGF-β production, which are indicators of pulmonary fibrosis, were seen. Our study showed that intratracheal instillation of polyhexamethyleneguanidine phosphate induces pulmonary inflammation and fibrosis in mice.
Collapse
Affiliation(s)
- Sang Jin Lee
- Department of Biochemistry, College of Medicine, Konyang University, 158 Gwanjeodong-ro, Seo-gu, Daejeon 302-718, Republic of Korea; Inhalation Toxicology Center, Korea Institute of Toxicology, 30 Baekhak 1-Gil, Jeongeup-si, Jeollabuk-do 580-185, Republic of Korea
| | - Jong-Hwan Park
- Laboratory Animal Medicine, College of Veterinary Medicine, Chonnam National University, 77 Yongbong-ro, Buk-gu , Gwangju 61186, Republic of Korea
| | - Jun-Young Lee
- Department of Biochemistry, College of Medicine, Konyang University, 158 Gwanjeodong-ro, Seo-gu, Daejeon 302-718, Republic of Korea
| | - Yu-Jin Jeong
- Department of Biochemistry, College of Medicine, Konyang University, 158 Gwanjeodong-ro, Seo-gu, Daejeon 302-718, Republic of Korea
| | - Jeong Ah Song
- Inhalation Toxicology Center, Korea Institute of Toxicology, 30 Baekhak 1-Gil, Jeongeup-si, Jeollabuk-do 580-185, Republic of Korea
| | - Kyuhong Lee
- Inhalation Toxicology Center, Korea Institute of Toxicology, 30 Baekhak 1-Gil, Jeongeup-si, Jeollabuk-do 580-185, Republic of Korea; Toxicology and Pharmacology, University of Science and Technology, 217 Gajeong-ro, Yuseong-gu, Daejeon 305-350, Republic of Korea
| | - Dong-Jae Kim
- Laboratory Animal Resource Center, Daegu Gyeongbuk Institute of Science & Technology (DGIST), 333 Techno Jungang Daero, Hyeonpung-myeon, Dalseong-gun, Daegu 42988, Republic of Korea
| |
Collapse
|
64
|
Rajasekaran S, Rajaguru P, Sudhakar Gandhi PS. MicroRNAs as potential targets for progressive pulmonary fibrosis. Front Pharmacol 2015; 6:254. [PMID: 26594173 PMCID: PMC4633493 DOI: 10.3389/fphar.2015.00254] [Citation(s) in RCA: 87] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2015] [Accepted: 10/19/2015] [Indexed: 12/19/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a chronic, progressive and devastating disorder. It is characterized by alveolar epithelial cell injury and activation, infiltration of inflammatory cells, initiation of epithelial mesenchymal transition (EMT), aberrant proliferation and activation of fibroblasts, exaggerated deposition of extracellular matrix (ECM) proteins, and finally leading to the destruction of lung parenchyma. MicroRNAs (miRNAs) are endogenous small non-coding RNA molecules that post-transcriptionally regulate gene expression in diverse biological and pathological processes, including cell proliferation, differentiation, apoptosis and metastasis. As a result, miRNAs have emerged as a major area of biomedical research with relevance to pulmonary fibrosis. In this context, the present review discusses specific patterns of dysregulated miRNAs in patients with IPF. Further, we discuss the current understanding of miRNAs involvement in regulating lung inflammation, TGF-β1-mediated EMT and fibroblast differentiation processes, ECM genes expression, and in the progression of lung fibrosis. The possible future directions that might lead to novel therapeutic strategies for the treatment of pulmonary fibrosis are also reviewed.
Collapse
Affiliation(s)
- Subbiah Rajasekaran
- Department of Biotechnology, Bharathidasan Institute of Technology Campus, Anna University Tiruchirappalli, India
| | - P Rajaguru
- Department of Biotechnology, Bharathidasan Institute of Technology Campus, Anna University Tiruchirappalli, India
| | - P S Sudhakar Gandhi
- Department of Biotechnology, Bharathidasan Institute of Technology Campus, Anna University Tiruchirappalli, India
| |
Collapse
|
65
|
TNF-α-stimulated fibroblasts secrete lumican to promote fibrocyte differentiation. Proc Natl Acad Sci U S A 2015; 112:11929-34. [PMID: 26351669 DOI: 10.1073/pnas.1507387112] [Citation(s) in RCA: 92] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
In healing wounds and fibrotic lesions, fibroblasts and monocyte-derived fibroblast-like cells called fibrocytes help to form scar tissue. Although fibrocytes promote collagen production by fibroblasts, little is known about signaling from fibroblasts to fibrocytes. In this report, we show that fibroblasts stimulated with the fibrocyte-secreted inflammatory signal tumor necrosis factor-α secrete the small leucine-rich proteoglycan lumican, and that lumican, but not the related proteoglycan decorin, promotes human fibrocyte differentiation. Lumican competes with the serum fibrocyte differentiation inhibitor serum amyloid P, but dominates over the fibroblast-secreted fibrocyte inhibitor Slit2. Lumican acts directly on monocytes, and unlike other factors that affect fibrocyte differentiation, lumican has no detectable effect on macrophage differentiation or polarization. α2β1, αMβ2, and αXβ2 integrins are needed for lumican-induced fibrocyte differentiation. In lung tissue from pulmonary fibrosis patients with relatively normal lung function, lumican is present at low levels throughout the tissue, whereas patients with advanced disease have pronounced lumican expression in the fibrotic lesions. These data may explain why fibrocytes are increased in fibrotic tissues, suggest that the levels of lumican in tissues may have a significant effect on the decision of monocytes to differentiate into fibrocytes, and indicate that modulating lumican signaling may be useful as a therapeutic for fibrosis.
Collapse
|
66
|
Abstract
Systemic sclerosis is an autoimmune disease that contains an interplay between inflammation and fibrosis. The ultimate effector cell is the myofibroblast that secretes excessive matrix molecules leading to fibrosis. There is no treatment that modifies the disease and this is an unmet clinical need. In this issue of Autoimmunity, Jafarinejad-Farsaangi et al., demonstrate that restitution of microRNA-29a targets the apoptosis rheostat and leads to apoptosis in dermal fibroblasts, suggesting that in vivo targeting of the microRNA through mimics would lead to depletion of the fibroblasts.
Collapse
Affiliation(s)
- Steven O'Reilly
- a School of Biological and Biomedical Sciences, Durham University , Durham , UK
| |
Collapse
|
67
|
Malaviya R, Sunil VR, Venosa A, Verissimo VL, Cervelli JA, Vayas KN, Hall L, Laskin JD, Laskin DL. Attenuation of Nitrogen Mustard-Induced Pulmonary Injury and Fibrosis by Anti-Tumor Necrosis Factor-α Antibody. Toxicol Sci 2015; 148:71-88. [PMID: 26243812 DOI: 10.1093/toxsci/kfv161] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Nitrogen mustard (NM) is a bifunctional alkylating agent that causes acute injury to the lung that progresses to fibrosis. This is accompanied by a prominent infiltration of macrophages into the lung and upregulation of proinflammatory/profibrotic cytokines including tumor necrosis factor (TNF)α. In these studies, we analyzed the ability of anti-TNFα antibody to mitigate NM-induced lung injury, inflammation, and fibrosis. Treatment of rats with anti-TNFα antibody (15 mg/kg, iv, every 9 days) beginning 30 min after intratracheal administration of NM (0.125 mg/kg) reduced progressive histopathologic alterations in the lung including perivascular and peribronchial edema, macrophage/monocyte infiltration, interstitial thickening, bronchiolization of alveolar walls, fibrin deposition, emphysema, and fibrosis. NM-induced damage to the alveolar-epithelial barrier, measured by bronchoalveolar lavage (BAL) protein and cell content, was also reduced by anti-TNFα antibody, along with expression of the oxidative stress marker, heme oxygenase-1. Whereas the accumulation of proinflammatory/cytotoxic M1 macrophages in the lung in response to NM was suppressed by anti-TNFα antibody, anti-inflammatory/profibrotic M2 macrophages were increased or unchanged. Treatment of rats with anti-TNFα antibody also reduced NM-induced increases in expression of the profibrotic mediator, transforming growth factor-β. This was associated with a reduction in NM-induced collagen deposition in the lung. These data suggest that inhibiting TNFα may represent an efficacious approach to mitigating lung injury induced by mustards.
Collapse
Affiliation(s)
- Rama Malaviya
- *Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy
| | - Vasanthi R Sunil
- *Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy
| | - Alessandro Venosa
- *Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy
| | | | - Jessica A Cervelli
- *Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy
| | - Kinal N Vayas
- *Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy
| | - LeRoy Hall
- Department of Drug Safety Sciences, Janssen Research & Development, Raritan, New Jersey 08869
| | - Jeffrey D Laskin
- Department of Environmental and Occupational Medicine, Robert Wood Johnson Medical School, Rutgers University, Piscataway, New Jersey 08854; and
| | - Debra L Laskin
- *Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy,
| |
Collapse
|
68
|
Adegunsoye A, Balachandran J. Inflammatory response mechanisms exacerbating hypoxemia in coexistent pulmonary fibrosis and sleep apnea. Mediators Inflamm 2015; 2015:510105. [PMID: 25944985 PMCID: PMC4402194 DOI: 10.1155/2015/510105] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Accepted: 03/17/2015] [Indexed: 01/02/2023] Open
Abstract
Mediators of inflammation, oxidative stress, and chemoattractants drive the hypoxemic mechanisms that accompany pulmonary fibrosis. Patients with idiopathic pulmonary fibrosis commonly have obstructive sleep apnea, which potentiates the hypoxic stimuli for oxidative stress, culminating in systemic inflammation and generalized vascular endothelial damage. Comorbidities like pulmonary hypertension, obesity, gastroesophageal reflux disease, and hypoxic pulmonary vasoconstriction contribute to chronic hypoxemia leading to the release of proinflammatory cytokines that may propagate clinical deterioration and alter the pulmonary fibrotic pathway. Tissue inhibitor of metalloproteinase (TIMP-1), interleukin- (IL-) 1α, cytokine-induced neutrophil chemoattractant (CINC-1, CINC-2α/β), lipopolysaccharide induced CXC chemokine (LIX), monokine induced by gamma interferon (MIG-1), macrophage inflammatory protein- (MIP-) 1α, MIP-3α, and nuclear factor- (NF-) κB appear to mediate disease progression. Adipocytes may induce hypoxia inducible factor (HIF) 1α production; GERD is associated with increased levels of lactate dehydrogenase (LDH), alkaline phosphatase (ALP), and tumor necrosis factor alpha (TNF-α); pulmonary artery myocytes often exhibit increased cytosolic free Ca2+. Protein kinase C (PKC) mediated upregulation of TNF-α and IL-1β also occurs in the pulmonary arteries. Increased understanding of the inflammatory mechanisms driving hypoxemia in pulmonary fibrosis and obstructive sleep apnea may potentiate the identification of appropriate therapeutic targets for developing effective therapies.
Collapse
Affiliation(s)
- Ayodeji Adegunsoye
- Section of Pulmonary & Critical Care, Department of Medicine, University of Chicago, Chicago, IL 60637, USA
| | - Jay Balachandran
- Section of Pulmonary & Critical Care, Department of Medicine, University of Chicago, Chicago, IL 60637, USA
| |
Collapse
|
69
|
Yang D, Yuan W, Lv C, Li N, Liu T, Wang L, Sun Y, Qiu X, Fu Q. Dihydroartemisinin supresses inflammation and fibrosis in bleomycine-induced pulmonary fibrosis in rats. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2015; 8:1270-1281. [PMID: 25973011 PMCID: PMC4396330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 12/08/2014] [Accepted: 01/23/2015] [Indexed: 06/04/2023]
Abstract
Pulmonary fibrosis is a respiratory disease with a high mortality rate and its pathogenesis involves multiple mechanisms including epithelial cell injury, fibroblast proliferation, inflammation, and collagen coagulation. The treatment regimens still fail to recover this disease. We have previously found that dihydroartemisinin inhibits the development of pulmonary fibrosis in rats. This study aimed to determine the mechanisms of dihydroartemisinin in bleomycin-induced pulmonary fibrosis. The experimental rats were divided into six groups as normal saline control group (NS group), bleomycin group (BLM group), dihydroartemisinin-1, -2, or -3 group (DHA-1, DHA-2 and DHA-3 group) and dexamethasone group (DXM group). In BLM group, rats were treated with intratracheal instillation of bleomycin. NS group received the same volume of saline instead of bleomycin. In DHA-1, DHA-2 and DHA-3 group, in addition to intratracheal instillation of bleomycin, respectively, dihydroartemisinin (25 mg/kg, 50 mg/kg, 100 mg/kg daily) was administrated by intraperitoneal instillation. In DXM group, rats were treated with intraperitoneal instillation of dexamethasone as control. Immunocytochemical assay, reverse transcription PCR and western blot were used for detecting the expression of TGF-β1, TNF-α, α-SMA and NF-κB in lung tissues. What's more, morphological change and collagen deposition were analyzed by hematoxylin-eosin staining and Masson staining. Collagen synthesis was detected by hydroxyproline chromatometry. Results showed that dihydroartemisinin significantly decreased the amount of inflammatory cytokines and collagen synthesis, and inhibited fibroblast proliferation in bleomycin-induced pulmonary fibrosis (P < 0.001). This study provides experimental evidence that dihydroartemisinin could decrease cytokines, alveolar inflammation and attenuates lung injury and fibrosis.
Collapse
Affiliation(s)
- Dongxia Yang
- College of Basic Medicine, China Medical UniversityChina
- College of Basic Medicine, Binzhou Medical UniversityChina
| | - Wendan Yuan
- College of Basic Medicine, Binzhou Medical UniversityChina
| | - Changjun Lv
- College of Basic Medicine, Binzhou Medical UniversityChina
| | - Naie Li
- College of Basic Medicine, Binzhou Medical UniversityChina
| | - Tongshen Liu
- College of Basic Medicine, Binzhou Medical UniversityChina
| | - Liang Wang
- College of Basic Medicine, Binzhou Medical UniversityChina
| | - Yufei Sun
- College of Basic Medicine, Binzhou Medical UniversityChina
| | - Xueshan Qiu
- College of Basic Medicine, China Medical UniversityChina
| | - Qiang Fu
- College of Basic Medicine, Binzhou Medical UniversityChina
| |
Collapse
|
70
|
Honda I, Taki A, Morioka C, Komaki M, Miyasaka N, Oshima N, Iseki S, Morio T, Kubota T, Morita I. Mesenchymal stem cells ameliorate intra-amniotic inflammation-related neonatal complications in rats. Inflamm Regen 2015. [DOI: 10.2492/inflammregen.35.261] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Affiliation(s)
- Izumi Honda
- Department of Comprehensive Reproductive Medicine, Tokyo Medical and Dental University Graduate School
| | - Atsuko Taki
- Department of Pediatrics and Developmental Biology, Tokyo Medical and Dental University Graduate School
| | - Chikako Morioka
- Department of Pediatrics and Developmental Biology, Tokyo Medical and Dental University Graduate School
| | - Motohiro Komaki
- Department of Nanomedicine (DNP), Tokyo Medical and Dental University Graduate School
| | - Naoyuki Miyasaka
- Department of Comprehensive Reproductive Medicine, Tokyo Medical and Dental University Graduate School
| | - Noriko Oshima
- Department of Comprehensive Reproductive Medicine, Tokyo Medical and Dental University Graduate School
| | - Sachiko Iseki
- Department of Molecular Craniofacial Embryology, Tokyo Medical and Dental University Graduate School
| | - Tomohiro Morio
- Department of Pediatrics and Developmental Biology, Tokyo Medical and Dental University Graduate School
| | - Toshiro Kubota
- Department of Comprehensive Reproductive Medicine, Tokyo Medical and Dental University Graduate School
| | - Ikuo Morita
- Department of Cellular Physiological Chemistry, Tokyo Medical and Dental University Graduate School
| |
Collapse
|
71
|
Dong J, Porter DW, Batteli LA, Wolfarth MG, Richardson DL, Ma Q. Pathologic and molecular profiling of rapid-onset fibrosis and inflammation induced by multi-walled carbon nanotubes. Arch Toxicol 2014; 89:621-33. [PMID: 25510677 DOI: 10.1007/s00204-014-1428-y] [Citation(s) in RCA: 70] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2014] [Accepted: 11/26/2014] [Indexed: 12/27/2022]
Abstract
Multi-walled carbon nanotubes (MWCNT) are new materials with a wide range of industrial and commercial applications. However, their nano-scaled size and fiber-like shape render them respirable and potentially fibrogenic if inhaled into the lungs. To understand MWCNT fibrogenesis, we analyzed the pathologic and molecular aspects of the early phase response to MWCNT in mouse lungs. MWCNT induced rapid and pronounced lesions in the lungs characterized by increased cellularity and formation of fibrotic foci, most notably near where MWCNT deposited, within 14 days post-exposure. Deposition of collagen fibers was markedly increased in the alveolar septa and fibrotic foci, accompanied by elevated expression of fibrotic genes Col1a1, Col1a2, and Fn1 at both mRNA and protein levels. Fibrosis was induced rapidly at 40 μg, wherein fibrotic changes were detected on day 1 and reached a maximal intensity on day 7 through day 14. Induction of fibrosis was dose-dependent at the dose range of 5-40 μg, 7 days post-exposure. MWCNT elicited rapid and prominent infiltrations of neutrophils and macrophages alongside fibrosis implicating acute inflammation in the fibrotic response. At the molecular level, MWCNT induced elevated expression of proinflammatory cytokines TNFα, IL1α, IL1β, IL6, and CCL2 in lung tissues as well as the bronchoalveolar lavage fluid, in a dose- and time-dependent manner. MWCNT also increased the expression of fibrogenic growth factors TGF-β1 and PDGF-A in the lungs significantly. These findings underscore the interplay between acute inflammation and the early fibrotic response in the initiation and propagation of pulmonary fibrosis induced by MWCNT.
Collapse
Affiliation(s)
- Jie Dong
- Receptor Biology Laboratory, Toxicology and Molecular Biology Branch, Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Centers for Disease Control and Prevention, 1095 Willowdale Road, Mailstop 3014, Morgantown, WV, 26505, USA
| | | | | | | | | | | |
Collapse
|
72
|
Eurlings IMJ, Dentener MA, Mercken EM, de Cabo R, Bracke KR, Vernooy JHJ, Wouters EFM, Reynaert NL. A comparative study of matrix remodeling in chronic models for COPD; mechanistic insights into the role of TNF-α. Am J Physiol Lung Cell Mol Physiol 2014; 307:L557-65. [PMID: 25106431 DOI: 10.1152/ajplung.00116.2014] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Remodeling in chronic obstructive pulmonary disease (COPD) has at least two dimensions: small airway wall thickening and destruction of alveolar walls. Recently we showed comparable alterations of the extracellular matrix (ECM) compounds collagen, hyaluoran, and elastin in alveolar and small airway walls of COPD patients. The aim of this study was to characterize and assess similarities in alveolar and small airway wall matrix remodeling in chronic COPD models. From this comparative characterization of matrix remodeling we derived and elaborated underlying mechanisms to the matrix changes reported in COPD. Lung tissue sections of chronic models for COPD, either induced by exposure to cigarette smoke, chronic intratracheal lipopolysaccharide instillation, or local tumor necrosis factor (TNF) expression [surfactant protein C (SPC)-TNFα mice], were stained for elastin, collagen, and hyaluronan. Furthermore TNF-α matrix metalloproteinase (MMP)-2, -9, and -12 mRNA expression was analyzed using qPCR and localized using immunohistochemistry. Both collagen and hyaluronan were increased in alveolar and small airway walls of all three models. Interestingly, elastin contents were differentially affected, with a decrease in both alveolar and airway walls in SPC-TNFα mice. Furthermore TNF-α and MMP-2 and -9 mRNA and protein levels were found to be increased in alveolar walls and around airway walls only in SPC-TNFα mice. We show that only SPC-TNFα mice show changes in elastin remodeling that are comparable to what has been observed in COPD patients. This reveals that the SPC-TNFα model is a suitable model to study processes underlying matrix remodeling and in particular elastin breakdown as seen in COPD. Furthermore we indicate a possible role for MMP-2 and MMP-9 in the breakdown of elastin in airways and alveoli of SPC-TNFα mice.
Collapse
Affiliation(s)
- Irene M J Eurlings
- Department of Respiratory Medicine, NUTRIM School for Nutrition, Toxicology and Metabolism, Maastricht University Medical Center, Maastricht, The Netherlands;
| | - Mieke A Dentener
- Department of Respiratory Medicine, NUTRIM School for Nutrition, Toxicology and Metabolism, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Evi M Mercken
- Experimental Gerontology Section, Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, Maryland; and
| | - Rafael de Cabo
- Experimental Gerontology Section, Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, Maryland; and
| | - Ken R Bracke
- Laboratory for Translational Research in Obstructive Pulmonary Diseases, Department of Respiratory Medicine, Ghent University Hospital, Ghent, Belgium
| | - Juanita H J Vernooy
- Department of Respiratory Medicine, NUTRIM School for Nutrition, Toxicology and Metabolism, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Emiel F M Wouters
- Department of Respiratory Medicine, NUTRIM School for Nutrition, Toxicology and Metabolism, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Niki L Reynaert
- Department of Respiratory Medicine, NUTRIM School for Nutrition, Toxicology and Metabolism, Maastricht University Medical Center, Maastricht, The Netherlands
| |
Collapse
|
73
|
Luo Y, Wang M, Pang Z, Jiang F, Chen J, Zhang J. Locally instilled tumor necrosis factor α antisense oligonucleotide contributes to inhibition of TH 2-driven pulmonary fibrosis via induced CD4+ CD25+ Foxp3+ regulatory T cells. J Gene Med 2014; 15:441-52. [PMID: 24339053 DOI: 10.1002/jgm.2750] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2013] [Revised: 10/29/2013] [Accepted: 12/09/2013] [Indexed: 11/11/2022] Open
Abstract
BACKGROUND Anti-tumor necrosis factor α therapeutics has the potential to alleviate pulmonary fibrosis. However, the systemic administration of anti-tumor necrosis factor α agents has brought about contradictory results and frequent adverse effects, such as infections, immunogenicity and malignancies, amongst others. In the present study, we attempted the local administration of tumor necrosis factor α antisense oligonucleotide and evaluated the treatment effects on pulmonary fibrosis in a bleomycin-induced pulmonary fibrosis mouse model. METHODS Flow cytometry for regulatory T cells, reverse transcriptase-polymerase chain reaction for crucial gene expression, western blotting for crucial protein products, immunofluorescent analysis for T(H)2 cells and myofibroblasts, as well as histology analysis for pathological examination, were used. RESULTS By local administration of tumor necrosis factor α antisense oligonucleotide, we investigated whether tumor necrosis factor α expression in epithelial cells was significantly inhibited and extracellular matrix overexpression was dramatically reduced. These treatment effects were associated with induced regulatory T cells, reduced T(H)2 cells and generally decreased T(H)2-type cytokine expression. Systemic immunosuppression was not triggered by local antisense oligonucleotide administration because the proportion of regulatory T cells in the blood, thymus or spleen was not affected. CONCLUSIONS These findings demonstrate that local administration of tumor necrosis factor α antisense oligonucleotide contributes to anti-fibrotic action via a sustained up-regulated level of regulatory T cells, which inhibits T(H)2-biased responses, pro-fibrotic mediator production and extracellular matrix deposition, with no systemic immunosupression associated with systemically induced regulatory T cells.
Collapse
Affiliation(s)
- Yi Luo
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China
| | | | | | | | | | | |
Collapse
|
74
|
Gono T, Kaneko H, Kawaguchi Y, Hanaoka M, Kataoka S, Kuwana M, Takagi K, Ichida H, Katsumata Y, Ota Y, Kawasumi H, Yamanaka H. Cytokine profiles in polymyositis and dermatomyositis complicated by rapidly progressive or chronic interstitial lung disease. Rheumatology (Oxford) 2014; 53:2196-203. [DOI: 10.1093/rheumatology/keu258] [Citation(s) in RCA: 113] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
|
75
|
Atzeni F, Boiardi L, Sallì S, Benucci M, Sarzi-Puttini P. Lung involvement and drug-induced lung disease in patients with rheumatoid arthritis. Expert Rev Clin Immunol 2014; 9:649-57. [PMID: 23899235 DOI: 10.1586/1744666x.2013.811173] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Interstitial lung disease (ILD) is a common extra-articular manifestation of rheumatoid arthritis (RA) and a significant cause of morbidity and mortality. Usual interstitial pneumonia and nonspecific interstitial pneumonia seem to be the most frequent patterns in RA patients with ILD, although the proportion of patients with usual interstitial pneumonia is higher than among patients with other systemic rheumatic autoimmune diseases. RA patients with ILD most frequently present with chronic symptoms of cough and dyspnea when climbing stairs or walking uphill. A physical examination may reveal inhalatory crackles and a pulmonary function test demonstrates restrictive physiology, often with reduced diffusing capacity. High-resolution computed tomography is generally sufficient to confirm a diagnosis of ILD, although a minority of cases may require a surgical lung biopsy. Conventional disease-modifying antirheumatic drugs such as methotrexate (MTX) or leflunomide (LEF) and biological agents such as TNF-blocking agents or rituximab may trigger or aggravate ILD in RA patients, and infections may contribute to increased mortality in such patients. LEF should not be used in patients with a history of MTX pneumonitis. The prevalence of interstitial pneumonia among RA patients treated with anti-TNF agents ranges from 0.5 to 3%; however, as the evidence that anti-TNF increases or decreases the risk of ILD is controversial, it is not clear whether this indicates more severe RA requiring biological therapy or the effect of exposure to potentially toxic drugs such as MTX or LEF. The development of treatment-related ILD is a paradoxical adverse event, and patients should be warned about this rare but serious complication of biological or disease-modifying antirheumatic drug therapy.
Collapse
Affiliation(s)
- Fabiola Atzeni
- Rheumatology Unit, L Sacco University Hospital, Milan, Italy.
| | | | | | | | | |
Collapse
|
76
|
Kayhan S, Guzel A, Duran L, Tutuncu S, Guzel A, Gunaydın M, Salis O, Okuyucu A, Selcuk MY. Effects of leflunomide on inflamation and fibrosis in bleomycine induced pulmonary fibrosis in wistar albino rats. J Thorac Dis 2013; 5:641-9. [PMID: 24255778 DOI: 10.3978/j.issn.2072-1439.2013.09.20] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2013] [Accepted: 09/24/2013] [Indexed: 11/14/2022]
Abstract
PURPOSES Pulmonary fibrosis is a rare and progressive lung disease with a high mortality rate. The treatment regimens still fail to recover the disease. Leflunomide (LEF) is an immunomodulatory agent with antiproliferative activity that is used for the treatment of rheumatoid arthritis. The purpose of the study is to investigate the potential therapeutic efficacy of LEF in bleomycin (BLM) induced pulmonary fibrosis. METHODS A total of 21 male, adult wistar albino rats were used. The animals were divided into three groups as control, BLM and BLM plus LEF groups (n=7). In BLM group, mice were treated with intratracheal instillation of BLM (2.5 U/kg). Control group received the same volume of saline instead of BLM. In LEF group, in addition to BLM, LEF (10 mg/kg, daily) was administrated by oral gavage. The effect of LEF on pulmonary inflammation and fibrosis was studied by measurements of serum clara cell protein-16 (CC-16), thiobarbituric acid reactive substance levels (TBARS), superoxide dismutase (SOD) and advanced oxidation protein products (AOPP) levels and lung tissue contents of IL-6, TNF-α and NF-κB by immunhistochemical examinations. RESULTS LEF significantly increased the level of CC-16 and decreased the level of AOPP (P=0.042 and P=0.003 respectively). Lung tissue contents of IL-6, TNF-α and NF-κB significantly decreased in LEF group compared to BLM group by immunhistochemical examinations (P<0.001). CONCLUSIONS LEF reduces oxidative stress factors, alveolar inflammation and attenuates lung injury and fibrosis.
Collapse
Affiliation(s)
- Servet Kayhan
- Department of Chest Disease, Ondokuz Mayis University, Faculty of Medicine, Samsun, Turkey
| | | | | | | | | | | | | | | | | |
Collapse
|
77
|
Aoshiba K, Tsuji T, Kameyama S, Itoh M, Semba S, Yamaguchi K, Nakamura H. Senescence-associated secretory phenotype in a mouse model of bleomycin-induced lung injury. EXPERIMENTAL AND TOXICOLOGIC PATHOLOGY : OFFICIAL JOURNAL OF THE GESELLSCHAFT FUR TOXIKOLOGISCHE PATHOLOGIE 2013; 65:1053-1062. [PMID: 23688655 DOI: 10.1016/j.etp.2013.04.001] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/15/2012] [Revised: 04/01/2013] [Accepted: 04/09/2013] [Indexed: 02/07/2023]
Abstract
Bleomycin produces DNA damage, apoptosis and senescence, all of which play crucial roles in the development of pulmonary fibrosis. Recently, close attention has been paid to a DNA damage-induced phenotypic change (senescence-associated secretory phenotype; SASP) as a trigger for the secretion of various mediators which modify the processes of tissue injury, inflammation, repair and fibrosis. We characterized the SASP in a murine model of bleomycin-induced lung injury. Mice were intratracheally administered bleomycin or control saline, and the lungs were obtained on days 7, 14 and 21. The occurrence of DNA damage and the SASP in the lungs was examined by immunostaining. γH2AX immunostaining of the bleomycin-treated lungs revealed double-strand breaks (DSBs), largely within E-cadherin-positive, β4-integirn-positive alveolar epithelial cells. The DSBs were associated with phosphorylation of ATM/ATR, a central signal transducer mediating the DNA damage response, and upregulation of the cyclin-dependent kinase inhibitor p21(CIP1). The DSBs persisted for at least 21 days after the bleomycin exposure, although it began to wane after 7 days. A subpopulation of the γH2AX-positive, DNA-damaged cells exhibited the SASP, characterized by overexpression of IL-6, TNFα, MMP-2 and MMP-9, in association with the phosphorylation of IKKα/β and p38 MAPK. Persistent DNA damage and the SASP are induced in the process of bleomycin-induced lung injury and repair, suggesting that these events play an important role in the regulation of inflammation and tissue remodeling in bleomycin-induced pneumopathy.
Collapse
Affiliation(s)
- Kazutetsu Aoshiba
- Department of Respiratory Medicine, Tokyo Medical University Ibaraki Medical Center, Japan.
| | | | | | | | | | | | | |
Collapse
|
78
|
Wada T, Akiyama Y, Yokota K, Sato K, Funakubo Y, Mimura T. [A case of rheumatoid arthritis complicated with deteriorated interstitial pneumonia after the administration of abatacept]. ACTA ACUST UNITED AC 2013; 35:433-8. [PMID: 23124086 DOI: 10.2177/jsci.35.433] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
We report a case of rheumatoid arthritis (RA) complicated with interstitial pneumonia that deteriorated after the administration of abatacept. A 55-year-old man developed RA and interstitial pneumonia. Although interstitial pneumonia was improved by high-dose glucocorticoids, various disease-modifying antirheumatic drugs including infliximab were ineffective for his arthritis. Tacrolimus was effective but was discontinued due to refractory itching and diarrhea. After 2 months, he was registered on the Phase III trial of abatacept in Japan because of worsening of arthritis. From 2 days after the abatacept administration, frothy sputum frequently appeared, but sputum culture was negative. On 13 days after the administration, the interstitial shadow was deteriorated by chest CT as compared with that of 2 months before, and he was dropped out from the trial. On 27 days after the administration, the dose of prednisolone was increased from 2 to 10 mg/day for his arthritis. On 44 days after the administration, the interstitial pneumonia improved. Abatacpet might be the cause of the deterioration of the interstitial pneumonia, but other possibilities such as discontinuation of tacrolimus, flare-up of RA itself or viral infection should be considered. This is the first report of deteriorated interstitial pneumonia after the abatacept administration in the literature. Further cases are needed to identify the relation between abatacept and interstitial pneumonia, however this possibility should be always kept in mind when we use abatacept.
Collapse
Affiliation(s)
- Takuma Wada
- Department of Rheumatology and Applied Immunology, Saitama Medical University
| | | | | | | | | | | |
Collapse
|
79
|
Roubille C, Haraoui B. Interstitial lung diseases induced or exacerbated by DMARDS and biologic agents in rheumatoid arthritis: a systematic literature review. Semin Arthritis Rheum 2013; 43:613-26. [PMID: 24231065 DOI: 10.1016/j.semarthrit.2013.09.005] [Citation(s) in RCA: 196] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2013] [Revised: 09/25/2013] [Accepted: 09/29/2013] [Indexed: 12/23/2022]
Abstract
OBJECTIVE To review published cases of induced or exacerbated interstitial lung disease (ILD) in rheumatoid arthritis (RA) associated with non-biologic disease-modifying antirheumatic drugs (nbDMARDs) and biologics and to discuss clinical implications in daily practice. METHODS We performed a systematic literature review from 1975 to July 2013 using Medline, Embase, Cochrane, and abstracts from the ACR 2010-2012 and EULAR 2010-2013 annual meetings. Case reports and series that suggest a causative role of nbDMARDs (methotrexate [MTX], leflunomide [LEF], gold, azathioprine [AZA], sulfasalazine [SSZ], and hydroxychloroquine [HCQ]) and biologic agents (TNF inhibitors [TNFi], rituximab [RTX], tocilizumab [TCZ], abatacept [ABA], and anakinra) in causing ILD or worsening a pre-existing ILD in RA patients were included. Results from observational and postmarketing studies as well as reviews on this topic were excluded from the qualitative analysis but still considered to discuss the implication of such drugs in generating or worsening ILD in RA patients. Comparisons were made between MTX-induced ILD in RA and the cases reported with other agents, in terms of clinical presentation, radiological features, and therapeutic management and outcomes. RESULTS The literature search identified 32 articles for MTX, 12 for LEF (resulting in 34 case reports), 3 for gold, 1 for AZA, 4 for SSZ, 27 for TNFi (resulting in 31 case reports), 3 for RTX, 5 for TCZ (resulting in 8 case reports), and 1 for ABA. No case was found for HCQ or anakinra. Common points are noted between LEF- and TNFi-related ILD in RA: ILD is a rare severe adverse event, mostly occurs within the first 20 weeks after initiation of therapy, causes dyspnea mostly in older patients, and can be fatal. Although no definitive causative relationship can be drawn from case reports and observational studies, these data argue for a pulmonary follow-up in RA patients with pre-existing ILD, while receiving biologic therapy or nbDMARDs. CONCLUSION As previously described for MTX, growing evidence highlights that LEF, TNFi, RTX, and TCZ may induce pneumonitis or worsen RA-related pre-existing ILD. Nonetheless, identifying a causal relationship between RA therapy and ILD-induced toxicity clearly appears difficult, partly because it is a rare condition.
Collapse
Affiliation(s)
- Camille Roubille
- Osteoarthritis Research Unit, University of Montreal Hospital Research Center (CRCHUM), Notre-Dame Hospital, Montreal, Quebec, Canada
| | - Boulos Haraoui
- Department of Medicine, Rheumatic Disease Unit, Centre Hospitalier de l'Université de Montréal (CHUM), Montreal, Quebec, Canada; Institut de rhumatologie de Montréal, 1551 Ontario St East, Montreal, Quebec, Canada H2L 1S6.
| |
Collapse
|
80
|
Della Latta V, Cabiati M, Rocchiccioli S, Del Ry S, Morales MA. The role of the adenosinergic system in lung fibrosis. Pharmacol Res 2013; 76:182-9. [PMID: 23994158 DOI: 10.1016/j.phrs.2013.08.004] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2013] [Revised: 08/05/2013] [Accepted: 08/14/2013] [Indexed: 12/15/2022]
Abstract
Adenosine (ADO) is a retaliatory metabolite that is expressed in conditions of injury or stress. During these conditions ATP is released at the extracellular level and is metabolized to adenosine. For this reason, adenosine is defined as a "danger signal" for cells and organs, in addition to its important role as homeostatic regulator. Its physiological functions are mediated through interaction with four specific transmembrane receptors called ADORA1, ADORA2A, ADORA2B and ADORA3. In the lungs of mice and humans all four adenosine receptors are expressed with different roles, having pro- and anti-inflammatory roles, determining bronchoconstriction and regulating lung inflammation and airway remodeling. Adenosine receptors can also promote differentiation of lung fibroblasts into myofibroblasts, typical of the fibrotic event. This last function suggests a potential involvement of adenosine in the fibrotic lung disease processes, which are characterized by different degrees of inflammation and fibrosis. Idiopathic pulmonary fibrosis (IPF) is the pathology with the highest degree of fibrosis and is of unknown etiology and burdened by lack of effective treatments in humans.
Collapse
Key Words
- 1-deoxy-1,6[[(3-iodophenyl)methyl]amino]-9H-purin-9yl-N-methyl-B-d-ribofuronamide
- 1-propyl-8-p-sulfophenulxanthine
- 2 hexynyl-5′-N ethylcarboxamidoadenosine
- 2-(2-phenyl)ethynyl-N-ethylcarboxamido-adenosine
- 2-CI-IB MECA
- 2-chloro-N6-cyclopentyladenosine
- 2-cloro-N6-(3-iodobenzyl)-adenosine-50-N methyluronamide
- 2-methyl-6-phenyl-4-phenylethynyl-1,4-dihydro-pyridine-3,5-dicarboxylicacid-3-ethyl ester-5-(4-nitro-benzyl)ester
- 2-p-(2-carboxyethyl) phenethylamino-50-N-ethyl-carboxamidoadenosine
- 2-phenyl hydroxypropynyl-5′-N-ethylcarboxamido adenosine phosphoinositide 3
- 3-ethyl-1-propyl-8-(1-(3-(trifluoromethyl) benzyl)-1H-pyrazol-4-yl)-1H-purine-2,6(3H,7H)-dione
- 3-ethyl-5-benzyl-2-methyl-4-phenylethynyl-6-phenyl-1,4-(±)-dihydropyridine-3,5-dicarboxylate
- 3-propyl-6-ethyl-5-[(ethylthio)carbonyl]-2-phenyl-4-propyl-3-pyridinecarboxylate
- 4-(2-[7-amino-2-(2-furyl)-{1,2,4}-triazolo{2,3-a}{1,3,5}triazin-5-ylamino]ethyl)pieno
- 5-[[(4-methoxyphenyl)amino]carbonyl]amino-8-methyl-2-(2-furyl)pyra-zolo[4,3-e]1,2,4-triazolo[1,5-c]pyrimidine
- 7-methyl-[11C]-(E)-8-(3-bromostyryl)-3,7-dimethyl-1-propargylxanthin
- 8-[4-[[[[(2-aminoethyl)amino]carbonyl]methyl]oxy]phenyl]-l,3-dipropylxanthine
- 8-cyclopentyl-1,3-dipropylxanthine
- 9-chloro-2-(2-furanyl)-5-[(phenylacetyl) amino] [1,2,4]-triazolo[1,5-c]quinazoline
- 9-chloro-2-(2-furanyl)-[1,2,4]triazolo[1,5-c]quinazolin-5-amine
- A(1)R
- A(2A)R
- A(2B)R
- A(3)R
- AB-MECA
- ADA
- ADO
- ADORA 1 receptor
- ADORA 2A receptor
- ADORA 2B receptor
- ADORA 3 receptor
- ADP
- AIP
- AK
- AMP
- ARs
- ATP
- Adenosine
- Adenosine receptors
- Bleomycin
- CCPA
- CD39
- CD73
- CGS 15943
- CGS21680
- CHA
- CNS
- CNT-1
- CNT-2
- COP
- COPD
- CPA
- CVT6883
- DAG
- DIP
- DPCPX
- E-8-(3,4-dimethoxystyryl)-1,3-dipropyl-7-methylxanthine
- ECM
- ENT-1
- ENT-2
- ET-1
- FITC
- HE-NECA
- IB-MECA
- IIPs
- ILD
- INO
- IPF
- Idiopathic pulmonary fibrosis
- KF17837
- LIP
- Lung disease
- MAP
- MRE3008-F207
- MRS 1191
- MRS 1220
- MRS 1334
- MRS 1523
- MRS 1754
- N-(4-cyanophenyl)-2-[4-(2,3,6,7-tetrahydro-2,6-dioxo-1,3-dipropyl-1H-purin-8-yl)-phenoxy]acetamide
- N-ethylcarboxamido-adenosine
- N6-(2-phenylisopropyl)adenosine
- N6-(4-aminobenzyl)-adenosine-5′-N-methyluronamidedihydrochloride
- N6-cyclohexyl adenosine
- N6-cyclopentyladenosine
- NECA
- NSPI
- PAH
- PENECA
- PHPNECA
- PIA
- PKC
- PLA2
- PLC
- PLD
- PSB1115
- RB-ILD
- ROS
- SCH-58261
- UIP
- XAC
- ZM 241385
- [11C]BS-DMPX
- [7-(2-phenylethyl)-5-amino-2-(2-furyl)-pyrazolo-(4,3-e)-1,2,4-triazolo(1,5-c) pyrimidine]
- acute interstitial pneumonia
- adenosine
- adenosine deaminase
- adenosine diphosphate
- adenosine kinase
- adenosine monophosphate
- adenosine receptors
- adenosine triphosphate
- cAMP
- central nervous system
- chronic obstructive pulmonary diseases
- concentrative nucleoside transporters-1
- concentrative nucleoside transporters-2
- cryptogenic organizing pneumonia
- cyclic adenosine monophosphate
- desquamative interstitial pneumonia
- diacylglycerol
- ecto-5′-nucleotidase
- ectonucleoside triphosphate diphosphohydrolase
- endothelin 1
- equilibrative nucleoside transporters-1
- equilibrative nucleoside transporters-2
- extracellular matrix
- fluorescein isothiocyanate
- idiopathic interstitial pneumonias
- idiopathic pulmonary fibrosis
- inosine
- interstitial lung disease
- lymphocytic interstitial pneumonia
- mitogen-activated protein
- nonspecific interstitial pneumonia
- phospholipase A2
- phospholipase C
- phospholipase D
- protein kinase C
- pulmonary arterial hypertension
- reactive oxygen specie
- respiratory bronchiolitis-associated interstitial lung disease
- usual interstitial pneumonia
Collapse
|
81
|
Borthwick LA, Wynn TA, Fisher AJ. Cytokine mediated tissue fibrosis. BIOCHIMICA ET BIOPHYSICA ACTA 2013; 1832:1049-60. [PMID: 23046809 PMCID: PMC3787896 DOI: 10.1016/j.bbadis.2012.09.014] [Citation(s) in RCA: 284] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2012] [Revised: 09/28/2012] [Accepted: 09/29/2012] [Indexed: 12/20/2022]
Abstract
Acute inflammation is a recognised part of normal wound healing. However, when inflammation fails to resolve and a chronic inflammatory response is established this process can become dysregulated resulting in pathological wound repair, accumulation of permanent fibrotic scar tissue at the site of injury and the failure to return the tissue to normal function. Fibrosis can affect any organ including the lung, skin, heart, kidney and liver and it is estimated that 45% of deaths in the western world can now be attributed to diseases where fibrosis plays a major aetiological role. In this review we examine the evidence that cytokines play a vital role in the acute and chronic inflammatory responses that drive fibrosis in injured tissues. This article is part of a Special Issue entitled: Fibrosis: Translation of basic research to human disease.
Collapse
Affiliation(s)
- Lee A Borthwick
- Tissue Fibrosis and Repair Group, Institute of Cellular Medicine, Medical School, Newcastle University, Newcastle Upon Tyne, NE2 4HH, UK; Immunopathogenesis Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA.
| | | | | |
Collapse
|
82
|
Tang K, Murano G, Wagner H, Nogueira L, Wagner PD, Tang A, Dalton ND, Gu Y, Peterson KL, Breen EC. Impaired exercise capacity and skeletal muscle function in a mouse model of pulmonary inflammation. J Appl Physiol (1985) 2013; 114:1340-50. [PMID: 23449936 DOI: 10.1152/japplphysiol.00607.2012] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Pulmonary TNFα has been linked to reduced exercise capacity in a subset of patients with moderate to severe chronic obstructive pulmonary disease (COPD). We hypothesized that prolonged, high expression of pulmonary TNFα impairs cardiac and skeletal muscle function, and both contribute to exercise limitation. Using a surfactant protein C promoter-TNFα construct, TNFα was overexpressed throughout life in mouse lungs (SP-C/TNFα+). TNFα levels in wild-type (WT) female serum and lung were two- and threefold higher than in WT male mice. In SP-C/TNFα+ mice, TNFα increased similarly in both sexes. Treadmill exercise was impaired only in male SP-C/TNFα+ mice. While increases in lung volume and airspace size induced by TNFα were comparable in both sexes, pulmonary hypertension along with lower body and muscle mass were evident only in male mice. Left ventricular (LV) function (cardiac output, stroke volume, LV maximal pressure, and LV maximal pressure dP/dt) was not altered by TNFα overexpression. Fatigue measured in isolated soleus and EDL was more rapid only in soleus of male SP-C/TNFα+ mice and accompanied by a loss of oxidative IIa fibers, citrate synthase activity, and PGC-1α mRNA and increase in atrogin-1 and MuRF1 expression also only in male mice. In situ gastrocnemius fatigue resistance, reflecting both oxygen availability and contractility, was decreased similarly in female and male SP-C/TNFα+ mice. These data indicate that male, but not female, mice overexpressing pulmonary TNFα are susceptible to exercise limitation, possibly due to muscle wasting and loss of the oxidative muscle phenotype, with protection in females possibly due to estrogen.
Collapse
Affiliation(s)
- Kechun Tang
- Department of Medicine, University of California, San Diego, La Jolla, California 92093-0623, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
83
|
Rafii R, Juarez MM, Albertson TE, Chan AL. A review of current and novel therapies for idiopathic pulmonary fibrosis. J Thorac Dis 2013; 5:48-73. [PMID: 23372951 DOI: 10.3978/j.issn.2072-1439.2012.12.07] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2012] [Accepted: 12/25/2012] [Indexed: 12/30/2022]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a progressively fibrotic interstitial lung disease that is associated with a median survival of 2-3 years from initial diagnosis. To date, there is no treatment approved for IPF in the United States, and only one pharmacological agent has been approved outside of the United States. Nevertheless, research over the past 10 years has provided us with a wealth of information on its histopathology, diagnostic work-up, and a greater understanding of its pathophysiology. Specifically, IPF is no longer thought to be a predominantly pro-inflammatory disorder. Rather, the fibrosis in IPF is increasingly understood to be the result of a fibroproliferative and aberrant wound healing cascade. The development of therapeutic targets has shifted in accord with this paradigm change. This review highlights the current understanding of IPF, and the recent as well as novel therapeutics being explored in clinical trials for the treatment of this devastating disease.
Collapse
Affiliation(s)
- Rokhsara Rafii
- Division of Pulmonary, Critical Care, and Sleep Medicine, University of California, Davis, School of Medicine and VA Northern California Health Care System, Sacramento, California, USA
| | | | | | | |
Collapse
|
84
|
Abstract
Fibrosis is a pathological feature of most chronic inflammatory diseases. Fibrosis, or scarring, is defined by the accumulation of excess extracellular matrix components. If highly progressive, the fibrotic process eventually leads to organ malfunction and death. Fibrosis affects nearly every tissue in the body. Here we discuss how key components of the innate and adaptive immune response contribute to the pathogenesis of fibrosis. We also describe how cell-intrinsic changes in important structural cells can perpetuate the fibrotic response by regulating the differentiation, recruitment, proliferation and activation of extracellular matrix-producing myofibroblasts. Finally, we highlight some of the key mechanisms and pathways of fibrosis that are being targeted as potential therapies for a variety of important human diseases.
Collapse
|
85
|
Abstract
Fibrosis is a pathological feature of most chronic inflammatory diseases. Fibrosis, or scarring, is defined by the accumulation of excess extracellular matrix components. If highly progressive, the fibrotic process eventually leads to organ malfunction and death. Fibrosis affects nearly every tissue in the body. Here we discuss how key components of the innate and adaptive immune response contribute to the pathogenesis of fibrosis. We also describe how cell-intrinsic changes in important structural cells can perpetuate the fibrotic response by regulating the differentiation, recruitment, proliferation and activation of extracellular matrix-producing myofibroblasts. Finally, we highlight some of the key mechanisms and pathways of fibrosis that are being targeted as potential therapies for a variety of important human diseases.
Collapse
Affiliation(s)
- Thomas A Wynn
- Immunopathogenesis Section, Program in Barrier Immunity and Repair, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA.
| | | |
Collapse
|
86
|
McGrath EE, Lawrie A, Marriott HM, Mercer P, Cross SS, Arnold N, Singleton V, Thompson AAR, Walmsley SR, Renshaw SA, Sabroe I, Chambers RC, Dockrell DH, Whyte MKB. Deficiency of tumour necrosis factor-related apoptosis-inducing ligand exacerbates lung injury and fibrosis. Thorax 2012; 67:796-803. [PMID: 22496351 PMCID: PMC3426075 DOI: 10.1136/thoraxjnl-2011-200863] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
BACKGROUND The death receptor ligand tumour necrosis factor-related apoptosis-inducing ligand (TRAIL) shows considerable clinical promise as a therapeutic agent. TRAIL induces leukocyte apoptosis, reducing acute inflammatory responses in the lung. It is not known whether TRAIL modifies chronic lung injury or whether TRAIL has a role in human idiopathic pulmonary fibrosis (IPF). We therefore explored the capacity of TRAIL to modify chronic inflammatory lung injury and studied TRAIL expression in patients with IPF. METHODS TRAIL(-/-) and wild-type mice were instilled with bleomycin and inflammation assessed at various time points by bronchoalveolar lavage and histology. Collagen deposition was measured by tissue hydroxyproline content. TRAIL expression in human IPF lung samples was assessed by immunohistochemistry and peripheral blood TRAIL measured by ELISA. RESULTS TRAIL(-/-) mice had an exaggerated delayed inflammatory response to bleomycin, with increased neutrophil numbers (mean 3.19±0.8 wild type vs 11.5±5.4×10(4) TRAIL(-/-), p<0.0001), reduced neutrophil apoptosis (5.42±1.6% wild type vs 2.47±0.5% TRAIL(-/-), p=0.0003) and increased collagen (3.45±0.2 wild type vs 5.8±1.3 mg TRAIL(-/-), p=0.005). Immunohistochemical analysis showed induction of TRAIL in bleomycin-treated wild-type mice. Patients with IPF demonstrated lower levels of TRAIL expression than in control lung biopsies and their serum levels of TRAIL were significantly lower compared with matched controls (38.1±9.6 controls vs 32.3±7.2 pg/ml patients with IPF, p=0.002). CONCLUSION These data suggest TRAIL may exert beneficial, anti-inflammatory actions in chronic pulmonary inflammation in murine models and that these mechanisms may be compromised in human IPF.
Collapse
Affiliation(s)
- Emmet E McGrath
- Department of Infection and Immunity, University of Sheffield, Sheffield, UK
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
87
|
Magnone M, Sturla L, Jacchetti E, Scarfì S, Bruzzone S, Usai C, Guida L, Salis A, Damonte G, De Flora A, Zocchi E. Autocrine abscisic acid plays a key role in quartz-induced macrophage activation. FASEB J 2012; 26:1261-1271. [PMID: 22042223 DOI: 10.1096/fj.11-187351] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/04/2024]
Abstract
Inhalation of quartz induces silicosis, a lung disease where alveolar macrophages release inflammatory mediators, including prostaglandin-E(2) (PGE(2)) and tumor necrosis factor α (TNF-α). Here we report the pivotal role of abscisic acid (ABA), a recently discovered human inflammatory hormone, in silica-induced activation of murine RAW264.7 macrophages and of rat alveolar macrophages (AMs). Stimulation of both RAW264.7 cells and AMs with quartz induced a significant increase of ABA release (5- and 10-fold, respectively), compared to untreated cells. In RAW264.7 cells, autocrine ABA released after quartz stimulation sequentially activates the plasma membrane receptor LANCL2 and NADPH oxidase, generating a Ca(2+) influx resulting in NFκ B nuclear translocation and PGE(2) and TNF-α release (3-, 2-, and 3.5-fold increase, respectively, compared to control, unstimulated cells). Quartz-stimulated RAW264.7 cells silenced for LANCL2 or preincubated with a monoclonal antibody against ABA show an almost complete inhibition of NFκ B nuclear translocation and PGE(2) and TNF-α release compared to controls electroporated with a scramble oligonucleotide or preincubated with an unrelated antibody. AMs showed similar early and late ABA-induced responses as RAW264.7 cells. These findings identify ABA and LANCL2 as key mediators in quartz-induced inflammation, providing possible new targets for antisilicotic therapy.
Collapse
Affiliation(s)
- Mirko Magnone
- DIMES, Section of Biochemistry, University of Genova, Viale Benedetto XV, 1 16132 Genova, Italy.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
88
|
Thomson EM, Williams A, Yauk CL, Vincent R. Overexpression of tumor necrosis factor-α in the lungs alters immune response, matrix remodeling, and repair and maintenance pathways. THE AMERICAN JOURNAL OF PATHOLOGY 2012; 180:1413-30. [PMID: 22322299 DOI: 10.1016/j.ajpath.2011.12.020] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/19/2011] [Revised: 12/03/2011] [Accepted: 12/09/2011] [Indexed: 11/25/2022]
Abstract
Increased production of tumor necrosis factor (TNF)-α and matrix metalloproteinases (MMPs) is a feature of inflammatory lung diseases, including emphysema and fibrosis, but the divergent pathological characteristics that result indicate involvement of other processes in disease pathogenesis. Transgenic mice overexpressing TNF-α in type II alveolar epithelial cells under the control of the surfactant protein (SP)-C promoter develop pulmonary inflammation and emphysema but are resistant to induction of fibrosis by administration of bleomycin or transforming growth factor-β. To study the molecular mechanisms underlying the development of this phenotype, we used a microarray approach to characterize the pulmonary transcriptome of SP-C/TNF-α mice and wild-type littermates. Four-month-old SP-C/TNF-α mice displayed pronounced pulmonary inflammation, airspace enlargement, increased MMP-2 and MMP-9 levels, and altered expression of 2332 probes. The functional assessment of genes with increased expression revealed enrichment of inflammatory/immune responses and proteases, whereas genes involved in protease inhibition, angiogenesis, cross-linking of basement membrane proteins, and myofibroblast differentiation were predominantly decreased. Comparison with multiple lung disease models identified a set of genes unique to the SP-C/TNF-α model and revealed that lack of extracellular matrix production distinguished SP-C/TNF-α mice from fibrosis models. Activation of inflammatory and proteolytic pathways and disruption of maintenance and repair processes are central features of emphysema in this TNF-overexpression model. Impairment of myofibroblast differentiation and extracellular matrix production may underlie resistance to induction of fibrosis.
Collapse
Affiliation(s)
- Errol M Thomson
- Hazard Identification Division, Environmental Health Science and Research Bureau, Health Canada, Ottawa, Ontario, Canada.
| | | | | | | |
Collapse
|
89
|
Panopoulos ST, Sfikakis PP. Biological treatments and connective tissue disease associated interstitial lung disease. Curr Opin Pulm Med 2012; 17:362-7. [PMID: 21597375 DOI: 10.1097/mcp.0b013e3283483ea5] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
PURPOSE OF REVIEW There is no specific therapy for interstitial lung disease associated with connective tissue diseases (CTDs-ILD), a potentially fatal condition for some of these patients. This article reviews currently available information on the effects on CTDs-ILD of biological treatments that are increasingly used with considerable success in various systemic diseases. RECENT FINDINGS A beneficial effect of antitumor necrosis factor (TNF) agents on CTDs-ILD has been described in sporadic patients with rheumatoid arthritis (RA), systemic sclerosis (SSc) and systemic lupus erythematosus (SLE). However, and despite the fact that there was no clear evidence of pulmonary toxicity of these agents in randomized-controlled trials comprising thousands of patients with RA and spondylarthropathies, new onset or exacerbation of preexisting ILD with high mortality rates has so far been observed in 144 RA patients following anti-TNF treatment in clinical practice. Likewise, administration of the B-cell depleting anti-CD20 antibody rituximab was beneficial for ILD in SSc patients but associated with new-onset ILD in isolated patients with RA and SLE. Pertinent information on other biological treatments is currently lacking. SUMMARY Data on the therapeutic role of biological agents in CTDs-ILD is preliminary and controversial. Although preexisting ILD is not a contraindication for these agents, until more information is available their administration should be stopped when new pulmonary symptoms occur.
Collapse
Affiliation(s)
- Stylianos T Panopoulos
- First Department of Propedeutic and Internal Medicine, Laikon Hospital, Athens University Medical School, Greece
| | | |
Collapse
|
90
|
Interstitial lung disease in rheumatoid arthritis in the era of biologics. Pulm Med 2011; 2011:931342. [PMID: 22229089 PMCID: PMC3250642 DOI: 10.1155/2011/931342] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2011] [Revised: 08/13/2011] [Accepted: 11/10/2011] [Indexed: 02/02/2023] Open
Abstract
Interstitial lung disease (ILD) represents a severe manifestation in connective tissue diseases (CTD), with an overall incidence of 15%, and it is still a challenge for clinicians evaluation and management. ILD is the most common manifestation of lung involvement in Rheumatoid Arthritis (RA), observed in up to 80% of biopsies, 50% of chest Computed Tomography (CT) and only 5% of chest radiographs. Histopatological patterns of ILD in RA may present with different patterns, such as: usual interstitial pneumonia, non specific interstitial pneumonia, desquamative interstitial pneumonia, organizing pneumonia, and eosinophilic infiltration. The incidence of ILD in RA patients is not only related to the disease itself, many drugs may be in fact associated with the development of pulmonary damage. Some reports suggest a causative role for TNFα inhibitors in RA-ILD development/worsening, anyway, no definitive statement can be drawn thus data are incomplete and affected by several variables. A tight control (pulmonary function tests and/or HRCT) is mandatory in patients with preexisting ILD, but it should be also performed in those presenting risk factors for ILD and mild respiratory symptoms. Biologic therapy should be interrupted, and, after excluding triggering infections, corticosteroids should be administered.
Collapse
|
91
|
Soluble tumor necrosis factor receptor: enbrel (etanercept) for subacute pulmonary dysfunction following allogeneic stem cell transplantation. Biol Blood Marrow Transplant 2011; 18:1044-54. [PMID: 22155140 DOI: 10.1016/j.bbmt.2011.11.031] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2011] [Accepted: 11/27/2011] [Indexed: 12/23/2022]
Abstract
Subacute lung disease, manifested as either obstructive (OLD) or restrictive (RLD) lung dysfunction, is a common complication following allogeneic stem cell transplantation. In each case, therapeutic options are limited, morbidity remains high, and long-term survival is poor. Between 2001 and 2008, 34 patients with noninfectious, obstructive (25) or RLD restrictive lung dysfunction (nine) received etanercept (Enbrel®, Amgen Inc.) 0.4 mg/kg/dose, subcutaneously, twice weekly, for 4 (group A) or 12 weeks (group B). Corticosteroids (if present at study entry) were kept constant for the initial 4 weeks of therapy and then tapered as tolerated. Thirty-one of 34 (91%) subjects were evaluable for response, and 10 (32%) met primary response criteria. There was no difference in response based on the duration of treatment (29% group A versus 35% group B; P = .99), the presence of RLD or OLD (33% versus 32%; P = .73), or the severity of pulmonary disease at study onset. Estimated 5-year overall survival rates following therapy were 61% (95% confidence interval, 46%-80%) for all subjects and 90% (95% confidence level, 73%-100%) for the 10 who met the primary response criteria. Five-year survival estimates for subjects treated with RLD was 44%, compared with 67% for those treated for OLD (P = .19). Etanercept was well tolerated, with no bacteremia or viremia observed. Pathogens were noted on posttherapy bronchoalveolar lavage in two cases. These data support the development of expanded clinical trials to study etanercept as a therapeutic agent for subacute lung injury after allogeneic stem cell transplantation.
Collapse
|
92
|
Jiang D, Liang J, Guo R, Xie T, Kelly FL, Martinu T, Yang T, Lovgren AK, Chia J, Liu N, Jung Y, Palmer SM, Noble PW. Long-term exposure of chemokine CXCL10 causes bronchiolitis-like inflammation. Am J Respir Cell Mol Biol 2011; 46:592-8. [PMID: 22162905 DOI: 10.1165/rcmb.2011-0116oc] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Chemokines and chemokine receptors have been implicated in the pathogenesis of bronchiolitis. CXCR3 ligands (CXCL10, CXCL9, and CXCL11) were elevated in patients with bronchiolitis obliterans syndrome (BOS) and chronic allorejection. Studies also suggested that blockage of CXCR3 or its ligands changed the outcome of T-cell recruitment and airway obliteration. We wanted to determine the role of the chemokine CXCL10 in the pathogenesis of bronchiolitis and BOS. In this study, we found that CXCL10 mRNA levels were significantly increased in patients with BOS. We generated transgenic mice expressing a mouse CXCL10 cDNA under control of the rat CC10 promoter. Six-month-old CC10-CXCL10 transgenic mice developed bronchiolitis characterized by airway epithelial hyperplasia and developed peribronchiolar and perivascular lymphocyte infiltration. The airway hyperplasia and T-cell inflammation were dependent on the presence of CXCR3. Therefore, long-term exposure of the chemokine CXCL10 in the lung causes bronchiolitis-like inflammation in mice.
Collapse
Affiliation(s)
- Dianhua Jiang
- Division of Pulmonary, Duke University School of Medicine, 106 Research Drive, Durham, NC 27710, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
93
|
Nieder ML, McDonald GB, Kida A, Hingorani S, Armenian SH, Cooke KR, Pulsipher MA, Baker KS. National Cancer Institute-National Heart, Lung and Blood Institute/pediatric Blood and Marrow Transplant Consortium First International Consensus Conference on late effects after pediatric hematopoietic cell transplantation: long-term organ damage and dysfunction. Biol Blood Marrow Transplant 2011; 17:1573-84. [PMID: 21963877 PMCID: PMC3215762 DOI: 10.1016/j.bbmt.2011.09.013] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2011] [Accepted: 09/27/2011] [Indexed: 01/19/2023]
Abstract
Long-term complications after hematopoietic cell transplantation (HCT) have been studied in detail. Although virtually every organ system can be adversely affected after HCT, the underlying pathophysiology of these late effects remain incompletely understood. This article describes our current understanding of the pathophysiology of late effects involving the gastrointestinal, renal, cardiac, and pulmonary systems, and discusses post-HCT metabolic syndrome studies. Underlying diseases, pretransplantation exposures, transplantation conditioning regimens, graft-versus-host disease, and other treatments contribute to these problems. Because organ systems are interdependent, long-term complications with similar pathophysiologic mechanisms often involve multiple organ systems. Current data suggest that post-HCT organ complications result from cellular damage that leads to a cascade of complex events. The interplay between inflammatory processes and dysregulated cellular repair likely contributes to end-organ fibrosis and dysfunction. Although many long-term problems cannot be prevented, appropriate monitoring can enable detection and organ-preserving medical management at earlier stages. Current management strategies are aimed at minimizing symptoms and optimizing function. There remain significant gaps in our knowledge of the pathophysiology of therapy-related organ toxicities disease after HCT. These gaps can be addressed by closely examining disease biology and identifying those patients at greatest risk for adverse outcomes. In addition, strategies are needed for targeted disease prevention and health promotion efforts for individuals deemed at high risk because of their genetic makeup or specific exposure profile.
Collapse
Affiliation(s)
| | - George B. McDonald
- University of Washington, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Aiko Kida
- University of Washington, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Sangeeta Hingorani
- University of Washington, Seattle Children’s Hospital, Seattle, WA
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA
| | | | - Kenneth R. Cooke
- Case Western Reserve University, Rainbow Babies & Children’s Hospital, Cleveland, OH
| | - Michael A. Pulsipher
- Primary Children’s Medical Center, Division of Hematology/BMT, University of Utah School of Medicine/Huntsman Cancer Institute, Salt Lake City, UT
| | - K. Scott Baker
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA
| |
Collapse
|
94
|
McGrath EE, Marriott HM, Lawrie A, Francis SE, Sabroe I, Renshaw SA, Dockrell DH, Whyte MKB. TNF-related apoptosis-inducing ligand (TRAIL) regulates inflammatory neutrophil apoptosis and enhances resolution of inflammation. J Leukoc Biol 2011; 90:855-65. [PMID: 21562052 PMCID: PMC3644175 DOI: 10.1189/jlb.0211062] [Citation(s) in RCA: 113] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2011] [Revised: 03/25/2011] [Accepted: 04/08/2011] [Indexed: 01/20/2023] Open
Abstract
Novel therapeutics targeting neutrophilic inflammation are a major unmet clinical need in acute and chronic inflammation. The timely induction of neutrophil apoptosis is critical for inflammation resolution, and it is thought that acceleration of apoptosis may facilitate resolution at inflammatory sites. We previously demonstrated that a death receptor ligand, TRAIL, accelerates neutrophil apoptosis in vitro. We examined the role of TRAIL in neutrophil-dominant inflammation in WT and TRAIL-deficient mice. TRAIL deficiency did not alter constitutive neutrophil apoptosis, whereas exogenous TRAIL accelerated apoptosis of murine peripheral blood neutrophils. We compared TRAIL-deficient and WT mice in two independent models of neutrophilic inflammation: bacterial LPS-induced acute lung injury and zymosan-induced peritonitis. In both models, TRAIL-deficient mice had an enhanced inflammatory response with increased neutrophil numbers and reduced neutrophil apoptosis. Correction of TRAIL deficiency and supraphysiological TRAIL signaling using exogenous protein enhanced neutrophil apoptosis and reduced neutrophil numbers in both inflammatory models with no evidence of effects on other cell types. These data indicate the potential therapeutic benefit of TRAIL in neutrophilic inflammation.
Collapse
Affiliation(s)
| | | | - Allan Lawrie
- Department of Cardiovascular Science, University of Sheffield, Sheffield, United Kingdom
| | - Sheila E. Francis
- Department of Cardiovascular Science, University of Sheffield, Sheffield, United Kingdom
| | - Ian Sabroe
- Academic Unit of Respiratory Medicine and
| | | | - David H. Dockrell
- Immunology and Infectious Disease, Department of Infection and Immunity, and
| | | |
Collapse
|
95
|
Wang Y, Xu SQ, Xu JH, Ding C. Treatment with etanercept in a patient with rheumatoid arthritis-associated interstitial lung disease. CLINICAL MEDICINE INSIGHTS-CASE REPORTS 2011; 4:49-52. [PMID: 22084614 PMCID: PMC3201102 DOI: 10.4137/ccrep.s8150] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
We report a case of a 52-year-old woman with a 1-year history of rheumatoid arthritis-associated interstitial lung disease referred to hospital because of aggravated pulmonary symptoms in spite of intensive treatment including prednisone, azathioprine and triptergium glycoside. We subsequently initiated treatment with 25 mg of etanercept, subcutaneously injected twice weekly. Following 6 months of therapy with this agent, sustained improvement in dyspnea, cough was reported by the patient and respiratory function test showed marked improvement. The improvement was confirmed by reduced middle and lower lung markings on chest radiography and high-resolution CT scan. This report suggests etanercept may be effective in the treatment of rheumatoid arthritis-associated interstitial lung disease.
Collapse
Affiliation(s)
- Yu Wang
- Department of Rheumatology and Immunology, No. 1 Affiliated Hospital, Anhui Medical University, Hefei 230022, Anhui, China
| | | | | | | |
Collapse
|
96
|
Abstract
Pulmonary fibrosis is a highly heterogeneous and lethal pathological process with limited therapeutic options. Although research on the pathogenesis of pulmonary fibrosis has frequently focused on the mechanisms that regulate the proliferation, activation, and differentiation of collagen-secreting myofibroblasts, recent studies have identified new pathogenic mechanisms that are critically involved in the initiation and progression of fibrosis in a variety of settings. A more detailed and integrated understanding of the cellular and molecular mechanisms of pulmonary fibrosis could help pave the way for effective therapeutics for this devastating and complex disease.
Collapse
Affiliation(s)
- Thomas A Wynn
- Program in Barrier Immunity and Repair and the Immunopathogenesis Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
97
|
Abstract
Idiopathic pulmonary fibrosis (IPF) remains a disease with an unknown cause and a poor prognosis. Among attempts to define disease pathogenesis, animal models of experimental lung fibrosis have a prominent role. Commonly used models include exposure to bleomycin, silica, fluorescein isothiocyanate; irradiation; or expression of specific genes through a viral vector or transgenic system. These all have been instrumental in the study of lung fibrosis, but all have limitations and fall short of recapitulating a pattern of usual interstitial pneumonia, the pathologic correlate to IPF. A model of repetitive bleomycin lung injury has recently been reported that results in marked lung fibrosis, prominent alveolar epithelial cell hyperplasia, a pattern of temporal heterogeneity and persistence of aberrant remodeling well after stimulus removal, representing a significant addition to the collection of animal lung fibrosis models. Taken together, animal models remain a key component in research strategies to better define IPF pathogenesis.
Collapse
|
98
|
Zuo L, Nogueira L, Hogan MC. Effect of pulmonary TNF-α overexpression on mouse isolated skeletal muscle function. Am J Physiol Regul Integr Comp Physiol 2011; 301:R1025-31. [PMID: 21697519 DOI: 10.1152/ajpregu.00126.2011] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
TNF-α is a proinflammatory cytokine that is involved in numerous pathological processes including chronic obstructive pulmonary disease (COPD). In the present study, we used a transgenic mouse model that overexpresses TNF-α in the lung (Tg(+)) to test the hypothesis that chronic exposure to TNF-α (as seen in COPD) reduces skeletal muscle force production and fatigue resistance, particularly under low Po(2) conditions. At 7-12 mo, body and muscle weight of both extensor digitorum longus (EDL) and soleus were significantly smaller in Tg(+) compared with littermate wild-type (WT) mice; however, the body-to-muscle weight ratio was not different between groups. EDL and soleus muscles were subjected to in vitro fatiguing contractile periods under high (∼550 Torr) and low Po(2) (∼40 Torr). Although all muscles were less fatigue-resistant during low Po(2) compared with high Po(2), only the soleus fatigued more rapidly in Tg(+) mice (∼12%) compared with WT at high Po(2). The maximal tension of EDL was equally reduced in Tg(+) mice (28-34% decrease from WT under both Po(2) conditions); but for soleus this parameter was smaller only under low Po(2) in Tg(+) mice (∼31% decrease from WT). The peak rate of relaxation and the peak rate of contraction were both significantly reduced in Tg(+) EDL muscles compared with WT EDL under low Po(2) conditions, but not in soleus. These results demonstrate that TNF-α upregulation in the lung impairs peripheral skeletal muscle function but affects fast- and slow-twitch muscles differentially at high and low Po(2).
Collapse
Affiliation(s)
- Li Zuo
- Dept. of Medicine, Univ. of California, San Diego, 9500 Gilman Dr., La Jolla, CA 92093-0623, USA.
| | | | | |
Collapse
|
99
|
Soluble TNF-α Receptor I Encoded on Plasmid Vector and Its Application in Experimental Gene Therapy of Radiation-Induced Lung Fibrosis. Arch Immunol Ther Exp (Warsz) 2011; 59:315-26. [DOI: 10.1007/s00005-011-0133-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2010] [Accepted: 03/07/2011] [Indexed: 01/10/2023]
|
100
|
Ramos-Casals M, Perez-Alvarez R, Perez-de-Lis M, Xaubet A, Bosch X. Pulmonary disorders induced by monoclonal antibodies in patients with rheumatologic autoimmune diseases. Am J Med 2011; 124:386-94. [PMID: 21531225 DOI: 10.1016/j.amjmed.2010.11.028] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2010] [Revised: 10/29/2010] [Accepted: 11/01/2010] [Indexed: 12/11/2022]
Abstract
Monoclonal antibodies have emerged as a new class of agents causing drug-related pulmonary involvement in patients with systemic rheumatologic autoimmune diseases. The most frequently associated noninfectious pulmonary diseases are interstitial pneumonia (118 cases reported by August 2010), sarcoid-like disease and vasculitis (40 cases), and 97% of cases are associated with agents blocking tumor necrosis factor (TNF), a cytokine implicated in pulmonary fibrosis, granuloma formation, and maintenance. Drug-induced interstitial pneumonia has a poor prognosis, with an overall mortality rate of around one-third, rising to two-thirds in patients with pre-existing interstitial disease. Sarcoid-like disease has a better prognosis, with resolution or improvement in 90% of cases. Although the evidence comes overwhelmingly from case reports and case series, suggested recommendations for patient management include a detailed pre-therapeutic evaluation, early identification of symptoms suggestive of pulmonary disease, and tailored therapy. Mycobacterial infection should be exhaustively investigated, especially after anti-TNF administration. Large, prospective, postmarketing studies including nonbiological agents as controls may help elucidate the real risk of pulmonary disease in patients with rheumatologic autoimmune diseases receiving monoclonal antibodies.
Collapse
Affiliation(s)
- Manuel Ramos-Casals
- Department of Autoimmune Diseases, Laboratory of Autoimmune Diseases Josep Font, Hospital Clínic, Institut d'Investigacions Biomèdiques August Pi i Sunyer, University of Barcelona, Spain
| | | | | | | | | | | |
Collapse
|