1
|
Wang H, Fung NH, Aloe C, Miles M, Selemidis S, Lopez AF, Wilson N, Owczarek C, Bozinovski S. CD131 antagonism blocks inflammation, emphysema and fibrosis in an asthma-COPD overlap mouse model originating in early life. Respirology 2025; 30:303-316. [PMID: 39814691 DOI: 10.1111/resp.14877] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Accepted: 12/15/2024] [Indexed: 01/18/2025]
Abstract
BACKGROUND AND OBJECTIVE Asthma-COPD overlap (ACO) is characterized by patients exhibiting features of both asthma and COPD. Currently, there is no specific treatment for ACO. This study aimed to investigate the therapeutic potential of targeting CD131, a shared receptor subunit for IL-3, IL-5 and GM-CSF, in ACO development and in preventing acute viral exacerbations. METHODS A two-hit mouse model of ACO was established by house dust mite (HDM) allergen sensitization to model asthma, and elastase treatment to model emphysema. In a separate model, human rhinovirus 1b (RV1b) was used to induce an acute asthma exacerbation. A neutralizing antibody against CD131 was used to block CD131 in vivo signalling. RESULTS Mice exposed to HDM and elastase developed cardinal features for asthma and COPD, including airway hyperreactivity (AHR) and emphysema. A mixed granulocytic inflammatory profile was identified in the lungs, including expansion of monocyte-derived macrophages, neutrophils and eosinophils. RT-qPCR analysis detected heightened gene expression of Mmp12, Il5 and Il13. Transcriptomic analysis further revealed pathway enrichment for type 2 inflammation and macrophage activation. Blockade of CD131 effectively reduced the lung inflammation and prevented the development of AHR, airway fibrosis and emphysema. Interestingly, pathway enrichment for Th1 response and interferon production detected in the model was not affected by the treatment. Consistently, CD131 antagonism prevented RV1b-induced asthma exacerbation without compromising RV1b clearance. CONCLUSION CD131 signalling coordinates multiple pathological pathways that drive airway inflammation and lung remodelling in ACO. Hence, CD131 antagonism represents a novel approach to combating the immunopathology in the complex ACO setting.
Collapse
Affiliation(s)
- Hao Wang
- School of Health and Biomedical Sciences, RMIT University, Melbourne, Victoria, Australia
- Centre for Respiratory Science & Health, Melbourne, Victoria, Australia
| | - Nok Him Fung
- School of Health and Biomedical Sciences, RMIT University, Melbourne, Victoria, Australia
- Centre for Respiratory Science & Health, Melbourne, Victoria, Australia
| | - Christian Aloe
- School of Health and Biomedical Sciences, RMIT University, Melbourne, Victoria, Australia
- Centre for Respiratory Science & Health, Melbourne, Victoria, Australia
| | - Mark Miles
- School of Health and Biomedical Sciences, RMIT University, Melbourne, Victoria, Australia
- Centre for Respiratory Science & Health, Melbourne, Victoria, Australia
| | - Stavros Selemidis
- School of Health and Biomedical Sciences, RMIT University, Melbourne, Victoria, Australia
- Centre for Respiratory Science & Health, Melbourne, Victoria, Australia
| | - Angel F Lopez
- Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, South Australia, Australia
| | | | | | - Steven Bozinovski
- School of Health and Biomedical Sciences, RMIT University, Melbourne, Victoria, Australia
- Centre for Respiratory Science & Health, Melbourne, Victoria, Australia
| |
Collapse
|
2
|
Mirra D, Esposito R, Spaziano G, Rafaniello C, Panico F, Squillante A, Falciani M, Abrego-Guandique DM, Caiazzo E, Gallelli L, Cione E, D’Agostino B. miRNA Signatures in Alveolar Macrophages Related to Cigarette Smoke: Assessment and Bioinformatics Analysis. Int J Mol Sci 2025; 26:1277. [PMID: 39941045 PMCID: PMC11818525 DOI: 10.3390/ijms26031277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 01/23/2025] [Accepted: 01/30/2025] [Indexed: 02/16/2025] Open
Abstract
Cigarette smoke (CS) is a driver of many respiratory diseases, including chronic obstructive pulmonary disease (COPD) and non-small cell lung cancer (NSCLC). Tobacco causes oxidative stress, impaired phagocytosis of alveolar macrophages (AMs), and alterations in gene expression in the lungs of smokers. MicroRNAs (miRNAs) are small non-coding RNAs that influence several regulatory pathways. Previously, we monitored the expressions of hsa-miR-223-5p, 16-5p, 20a-5p, -17-5p, 34a-5p, and 106a-5p in AMs derived from the bronchoalveolar lavage (BAL) of subjects with NSCLC, COPD, and smoker and non-smoker control groups. Here, we investigated the capability of CS conditionate media to modulate the abovementioned miRNAs in primary AMs obtained in the same 43 sex-matched subjects. The expressions of has-miR-34a-5p, 17-5p, 16-5p, 106a-5p, 223-5p, and 20a-5p were assessed before and after in vitro CS exposure by RT-PCR. In addition, a comprehensive bioinformatic analysis of miRNAs KEGGS and PPI linked to inflammation was performed. Distinct and common miRNA expression profiles were identified in response to CS, suggesting their possible role in smoking-related diseases. It is worth noting that, following exposure to CS, the expression levels of hsa-miR-34a-5p and 17-5p in both smokers and non-smokers, 106a-5p in non-smokers, and 20a-5p in smokers, shifted towards those found in individuals with COPD, suggesting them as a risk factor in developing this lung condition. Moreover, CS-focused sub-analysis identified miRNA which exhibited CS-dependent pattern and modulated mRNA involved in the immune system or AMs property regulation. In conclusion, our study uncovered miRNA signatures in AMs exposed to CS, indicating that CS might modify epigenetic patterns that contribute to macrophage activation and lung disease onset and progression.
Collapse
Affiliation(s)
- Davida Mirra
- Department of Environmental Biological and Pharmaceutical Sciences and Technologies, University of Campania “Luigi Vanvitelli”, 81100 Caserta, Italy; (D.M.); (R.E.); (E.C.); (B.D.)
| | - Renata Esposito
- Department of Environmental Biological and Pharmaceutical Sciences and Technologies, University of Campania “Luigi Vanvitelli”, 81100 Caserta, Italy; (D.M.); (R.E.); (E.C.); (B.D.)
| | - Giuseppe Spaziano
- Department of Environmental Biological and Pharmaceutical Sciences and Technologies, University of Campania “Luigi Vanvitelli”, 81100 Caserta, Italy; (D.M.); (R.E.); (E.C.); (B.D.)
| | - Concetta Rafaniello
- Campania Regional Centre for Pharmacovigilance and Pharmacoepidemiology, 80138 Naples, Italy;
- Section of Pharmacology “L. Donatelli”, Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy
| | - Francesca Panico
- Science of Health Department, School of Medicine, University of Catanzaro, 88100 Catanzaro, Italy; (F.P.); (D.M.A.-G.)
| | | | - Maddalena Falciani
- Pulmonary and Critical Care Medicine, Ospedale Scarlato, 84018 Scafati, Italy;
| | | | - Eleonora Caiazzo
- Department of Environmental Biological and Pharmaceutical Sciences and Technologies, University of Campania “Luigi Vanvitelli”, 81100 Caserta, Italy; (D.M.); (R.E.); (E.C.); (B.D.)
| | - Luca Gallelli
- Clinical Pharmacology and Pharmacovigilance Unit, Department of Health Sciences, Mater Domini Hospital, University of “Magna Graecia”, 88100 Catanzaro, Italy;
| | - Erika Cione
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, Italy;
| | - Bruno D’Agostino
- Department of Environmental Biological and Pharmaceutical Sciences and Technologies, University of Campania “Luigi Vanvitelli”, 81100 Caserta, Italy; (D.M.); (R.E.); (E.C.); (B.D.)
| |
Collapse
|
3
|
Li L, Chen H, Shi J, Chai S, Yan L, Meng D, Cai Z, Guan J, Xin Y, Zhang X, Sun W, Lu X, He M, Li Q, Yan X. Exhaled breath analysis for the discrimination of asthma and chronic obstructive pulmonary disease. J Breath Res 2024; 18:046002. [PMID: 38834048 DOI: 10.1088/1752-7163/ad53f8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Accepted: 06/04/2024] [Indexed: 06/06/2024]
Abstract
Chronic obstructive pulmonary disease (COPD) and asthma are the most common chronic respiratory diseases. In middle-aged and elderly patients, it is difficult to distinguish between COPD and asthma based on clinical symptoms and pulmonary function examinations in clinical practice. Thus, an accurate and reliable inspection method is required. In this study, we aimed to identify breath biomarkers and evaluate the accuracy of breathomics-based methods for discriminating between COPD and asthma. In this multi-center cross-sectional study, exhaled breath samples were collected from 89 patients with COPD and 73 with asthma and detected on a high-pressure photon ionization time-of-flight mass spectrometry (HPPI-TOFMS) platform from 20 October 2022, to 20 May 2023, in four hospitals. Data analysis was performed from 15 June 2023 to 16 August 2023. The sensitivity, specificity, and accuracy were calculated to assess the overall performance of the volatile organic component (VOC)-based COPD and asthma discrimination models. Potential VOC markers related to COPD and asthma were also analyzed. The age of all participants ranged from to 18-86 years, and 54 (33.3%) were men. The age [median (minimum, maximum)] of COPD and asthma participants were 66.0 (46.0, 86.0), and 44.0 (17.0, 80.0). The male and female ratio of COPD and asthma participants were 14/75 and 40/33, respectively. Based on breathomics feature selection, ten VOCs were identified as COPD and asthma discrimination biomarkers via breath testing. The joint panel of these ten VOCs achieved an area under the curve of 0.843, sensitivity of 75.9%, specificity of 87.5%, and accuracy of 80.0% in COPD and asthma discrimination. Furthermore, the VOCs detected in the breath samples were closely related to the clinical characteristics of COPD and asthma. The VOC-based COPD and asthma discrimination model showed good accuracy, providing a new strategy for clinical diagnosis. Breathomics-based methods may play an important role in the diagnosis of COPD and asthma.
Collapse
Affiliation(s)
- Lan Li
- The First Department of Pulmonary and Critical Care Medicine, The Second Hospital of Hebei Medical University, Hebei Key Laboratory of Respiratory Critical Care Medicine, Hebei Institute of Respiratory Diseases, No. 215 Heping West Road, Shijiazhuang, Hebei 050000, People's Republic of China
- Shijiazhuang People's Hospital, No. 365 Jianhua Street, Shijiazhuang, Hebei 050000, People's Republic of China
| | - Haibin Chen
- Breax Laboratory, PCAB Research Center of Breath and Metabolism, Beijing 100071, People's Republic of China
- Digital Medicine Division, Guangzhou Sinohealth Digital Technology Co., Ltd, Guangzhou 510000, People's Republic of China
| | - Jinying Shi
- Shijiazhuang People's Hospital, No. 365 Jianhua Street, Shijiazhuang, Hebei 050000, People's Republic of China
| | - Shukun Chai
- Shijiazhuang People's Hospital, No. 365 Jianhua Street, Shijiazhuang, Hebei 050000, People's Republic of China
| | - Li Yan
- Hebei General Hospital, No. 348 Heping West Road, Shijiazhuang, Hebei 050000, People's Republic of China
| | - Deyang Meng
- Hebei General Hospital, No. 348 Heping West Road, Shijiazhuang, Hebei 050000, People's Republic of China
| | - Zhigang Cai
- The First Department of Pulmonary and Critical Care Medicine, The Second Hospital of Hebei Medical University, Hebei Key Laboratory of Respiratory Critical Care Medicine, Hebei Institute of Respiratory Diseases, No. 215 Heping West Road, Shijiazhuang, Hebei 050000, People's Republic of China
| | - Jitao Guan
- The First Department of Pulmonary and Critical Care Medicine, The Second Hospital of Hebei Medical University, Hebei Key Laboratory of Respiratory Critical Care Medicine, Hebei Institute of Respiratory Diseases, No. 215 Heping West Road, Shijiazhuang, Hebei 050000, People's Republic of China
| | - Yunwei Xin
- The First Department of Pulmonary and Critical Care Medicine, The Second Hospital of Hebei Medical University, Hebei Key Laboratory of Respiratory Critical Care Medicine, Hebei Institute of Respiratory Diseases, No. 215 Heping West Road, Shijiazhuang, Hebei 050000, People's Republic of China
| | - Xu Zhang
- The First Department of Pulmonary and Critical Care Medicine, The Second Hospital of Hebei Medical University, Hebei Key Laboratory of Respiratory Critical Care Medicine, Hebei Institute of Respiratory Diseases, No. 215 Heping West Road, Shijiazhuang, Hebei 050000, People's Republic of China
| | - Wuzhuang Sun
- The First Hospital of Hebei Medical University, No. 68 Donggang Road, Shijiazhuang, Hebei 050000, People's Republic of China
| | - Xi Lu
- The First Hospital of Hebei Medical University, No. 68 Donggang Road, Shijiazhuang, Hebei 050000, People's Republic of China
| | - Mengqi He
- Breax Laboratory, PCAB Research Center of Breath and Metabolism, Beijing 100071, People's Republic of China
| | - Qingyun Li
- Breax Laboratory, PCAB Research Center of Breath and Metabolism, Beijing 100071, People's Republic of China
| | - Xixin Yan
- The First Department of Pulmonary and Critical Care Medicine, The Second Hospital of Hebei Medical University, Hebei Key Laboratory of Respiratory Critical Care Medicine, Hebei Institute of Respiratory Diseases, No. 215 Heping West Road, Shijiazhuang, Hebei 050000, People's Republic of China
| |
Collapse
|
4
|
Kokkinis S, Singh M, Paudel KR, De Rubis G, Bani Saeid A, Jessamine V, Datsyuk J, Singh SK, Vishwas S, Adams J, Hansbro PM, Oliver B, Gupta G, Dureja H, Dua K. Plant-based therapeutics for chronic obstructive pulmonary diseases: Nanoformulation strategies to overcome delivery challenges. FOOD BIOSCI 2024; 58:103761. [DOI: 10.1016/j.fbio.2024.103761] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
5
|
Pellegrino D, Casas-Recasens S, Faner R, Palange P, Agusti A. When GETomics meets aging and exercise in COPD. Respir Med 2023:107294. [PMID: 37295536 DOI: 10.1016/j.rmed.2023.107294] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 05/17/2023] [Accepted: 05/23/2023] [Indexed: 06/12/2023]
Abstract
The term GETomics has been recently proposed to illustrate that human health and disease are actually the final outcome of many dynamic, interacting and cumulative gene (G) - environment (E) interactions that occur through the lifetime (T) of the individual. According to this new paradigm, the final outcome of any GxE interactions depends on both the age of the individual at which such GxE interaction occurs as well as on the previous, cumulative history of previous GxE interactions through the induction of epigenetic changes and immune memory (both lasting overtime). Following this conceptual approach, our understanding of the pathogenesis of chronic obstructive pulmonary disease (COPD) has changed dramatically. Traditionally believed to be a self-inflicted disease induced by tobacco smoking occurring in older men and characterized by an accelerated decline of lung function with age, now we understand that there are many other risk factors associated with COPD, that it occurs also in females and young individuals, that there are different lung function trajectories through life, and that COPD is not always characterized by accelerated lung function decline. In this paper we discuss how a GETomics approach to COPD may open new perspectives to better understand its relationship with exercise limitation and the ageing process.
Collapse
Affiliation(s)
- D Pellegrino
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Italy; Respiratory and Critical Care Unit, Policlinico Umberto I Hospital of Rome, Italy
| | - S Casas-Recasens
- Institut d'investigacions biomédiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain; Centro Investigacion Biomedica en Red de Enfermedades Respiratorias (CIBERES), Spain
| | - R Faner
- Institut d'investigacions biomédiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain; Centro Investigacion Biomedica en Red de Enfermedades Respiratorias (CIBERES), Spain; Cathedra Salut Respiratoria, University of Barcelona, Spain
| | - P Palange
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Italy; Respiratory and Critical Care Unit, Policlinico Umberto I Hospital of Rome, Italy
| | - A Agusti
- Institut d'investigacions biomédiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain; Centro Investigacion Biomedica en Red de Enfermedades Respiratorias (CIBERES), Spain; Cathedra Salut Respiratoria, University of Barcelona, Spain; Respiratory Institute, Clinic Barcelona, Spain.
| |
Collapse
|
6
|
Hanson C, Ponce J, Isaak M, Heires A, Nordgren T, Wichman C, Furtado JD, LeVan T, Romberger D. Fatty Acids, Amphiregulin Production, and Lung Function in a Cohort of Midwestern Veterans. FRONTIERS IN REHABILITATION SCIENCES 2022; 3:773835. [PMID: 36188926 PMCID: PMC9397678 DOI: 10.3389/fresc.2022.773835] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Accepted: 02/10/2022] [Indexed: 11/30/2022]
Abstract
Rationale The relationship between many fatty acids and respiratory outcomes remains unclear, especially with regard to mechanistic actions. Altered regulation of the process of lung repair is a key feature of chronic lung disease and may impact the potential for pulmonary rehabilitation, but underlying mechanisms of lung repair following injury or inflammation are not well-studied. The epidermal growth factor receptor agonist amphiregulin (AREG) has been demonstrated to promote lung repair following occupational dust exposure in animals. Studies suggest the polyunsaturated fatty acid (PUFA) docosahexaenoic acid (DHA) may enhance the production of AREG. The objective of this study was to determine the relationship between fatty acids and lung function in a population of veterans and determine if fatty acid status is associated with concentrations of AREG. Materials and Methods Data were collected from a cross-sectional study of veterans within the Nebraska-Western Iowa Health Care System. Whole blood assays were performed to quantify AREG concentrations via a commercially available ELISA kit. Fatty acids from plasma samples from the same patients were measured using gas-liquid chromatography. Intakes of fatty acids were quantified with a validated food frequency questionnaire. Linear regression models were used to determine whether plasma fatty acids or intakes of fatty acids predicted lung function or AREG concentrations. A p < 0.05 was considered statistically significant. Results Ninety participants were included in this analysis. In fully adjusted models, plasma fatty acids were associated with AREG production, including the PUFA eicosapentaenoic acid (EPA) (β = 0.33, p = 0.03) and the monounsaturated fatty acid octadecenoic acid: (β = −0.56, p = 0.02). The omega-3 PUFA docosapentaenoic acid (DPA) was positively associated with lung function (β = 0.28, p = 0.01; β = 26.5, p = 0.05 for FEV1/FVC ratio and FEV1 % predicted, respectively), as were the omega-6 PUFAs eicosadienoic acid (β = 1.13, p < 0.001; β = 91.2, p = 0.005 for FEV1/FVC ratio and FEV1 % predicted, respectively) and docosadienoic acid (β = 0.29, p = 0.01 for FEV1/FVC ratio). Plasma monounsaturated and saturated fatty acids were inversely associated with lung function. Conclusion Opposing anti- and pro-inflammatory properties of different fatty acids may be associated with lung function in this population, in part by regulating AREG induction.
Collapse
Affiliation(s)
- Corrine Hanson
- Department of Medical Sciences, Division of Medical Nutrition, College of Allied Health Professions, University of Nebraska Medical Center, Omaha, NE, United States
- *Correspondence: Corrine Hanson
| | - Jana Ponce
- Department of Medical Sciences, Division of Medical Nutrition, College of Allied Health Professions, University of Nebraska Medical Center, Omaha, NE, United States
| | - Mia Isaak
- Department of Medical Sciences, Division of Medical Nutrition, College of Allied Health Professions, University of Nebraska Medical Center, Omaha, NE, United States
| | - Art Heires
- Department of Internal Medicine, College of Medicine, University of Nebraska Medical Center, Omaha, NE, United States
- VA Nebraska Western Iowa Healthcare System, Omaha, NE, United States
| | - Tara Nordgren
- Colorado State University, Fort Collins, CO, United States
| | - Chris Wichman
- Department of Biostatistics, College of Public Health, University of Nebraska Medical Center, Omaha, NE, United States
| | - Jeremy D. Furtado
- Harvard T. H. Chan School of Public Health, Boston, MA, United States
| | - Tricia LeVan
- Department of Internal Medicine, College of Medicine, University of Nebraska Medical Center, Omaha, NE, United States
- VA Nebraska Western Iowa Healthcare System, Omaha, NE, United States
| | - Debra Romberger
- Department of Internal Medicine, College of Medicine, University of Nebraska Medical Center, Omaha, NE, United States
- VA Nebraska Western Iowa Healthcare System, Omaha, NE, United States
| |
Collapse
|
7
|
Avci E, Sarvari P, Savai R, Seeger W, Pullamsetti SS. Epigenetic Mechanisms in Parenchymal Lung Diseases: Bystanders or Therapeutic Targets? Int J Mol Sci 2022; 23:ijms23010546. [PMID: 35008971 PMCID: PMC8745712 DOI: 10.3390/ijms23010546] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 12/28/2021] [Accepted: 12/30/2021] [Indexed: 12/17/2022] Open
Abstract
Epigenetic responses due to environmental changes alter chromatin structure, which in turn modifies the phenotype, gene expression profile, and activity of each cell type that has a role in the pathophysiology of a disease. Pulmonary diseases are one of the major causes of death in the world, including lung cancer, idiopathic pulmonary fibrosis (IPF), chronic obstructive pulmonary disease (COPD), pulmonary hypertension (PH), lung tuberculosis, pulmonary embolism, and asthma. Several lines of evidence indicate that epigenetic modifications may be one of the main factors to explain the increasing incidence and prevalence of lung diseases including IPF and COPD. Interestingly, isolated fibroblasts and smooth muscle cells from patients with pulmonary diseases such as IPF and PH that were cultured ex vivo maintained the disease phenotype. The cells often show a hyper-proliferative, apoptosis-resistant phenotype with increased expression of extracellular matrix (ECM) and activated focal adhesions suggesting the presence of an epigenetically imprinted phenotype. Moreover, many abnormalities observed in molecular processes in IPF patients are shown to be epigenetically regulated, such as innate immunity, cellular senescence, and apoptotic cell death. DNA methylation, histone modification, and microRNA regulation constitute the most common epigenetic modification mechanisms.
Collapse
MESH Headings
- Animals
- Biomarkers
- Combined Modality Therapy
- DNA Methylation
- Diagnosis, Differential
- Disease Management
- Disease Susceptibility
- Epigenesis, Genetic
- Gene Expression Regulation
- Histones/metabolism
- Humans
- Idiopathic Pulmonary Fibrosis/diagnosis
- Idiopathic Pulmonary Fibrosis/etiology
- Idiopathic Pulmonary Fibrosis/metabolism
- Idiopathic Pulmonary Fibrosis/therapy
- Lung Diseases, Interstitial/diagnosis
- Lung Diseases, Interstitial/etiology
- Lung Diseases, Interstitial/metabolism
- Lung Diseases, Interstitial/therapy
- Pulmonary Disease, Chronic Obstructive/diagnosis
- Pulmonary Disease, Chronic Obstructive/etiology
- Pulmonary Disease, Chronic Obstructive/metabolism
- Pulmonary Disease, Chronic Obstructive/therapy
- Treatment Outcome
Collapse
Affiliation(s)
- Edibe Avci
- Department of Lung Development and Remodeling, Max-Planck Institute for Heart and Lung Research, Parkstrasse 1, 61231 Bad Nauheim, Germany; (E.A.); (P.S.); (R.S.); (W.S.)
| | - Pouya Sarvari
- Department of Lung Development and Remodeling, Max-Planck Institute for Heart and Lung Research, Parkstrasse 1, 61231 Bad Nauheim, Germany; (E.A.); (P.S.); (R.S.); (W.S.)
| | - Rajkumar Savai
- Department of Lung Development and Remodeling, Max-Planck Institute for Heart and Lung Research, Parkstrasse 1, 61231 Bad Nauheim, Germany; (E.A.); (P.S.); (R.S.); (W.S.)
- Department of Internal Medicine, Justus Liebig University, 35392 Giessen, Germany
- Institute for Lung Health (ILH), Justus Liebig University, 35392 Giessen, Germany
| | - Werner Seeger
- Department of Lung Development and Remodeling, Max-Planck Institute for Heart and Lung Research, Parkstrasse 1, 61231 Bad Nauheim, Germany; (E.A.); (P.S.); (R.S.); (W.S.)
- Department of Internal Medicine, Justus Liebig University, 35392 Giessen, Germany
- Institute for Lung Health (ILH), Justus Liebig University, 35392 Giessen, Germany
| | - Soni S. Pullamsetti
- Department of Lung Development and Remodeling, Max-Planck Institute for Heart and Lung Research, Parkstrasse 1, 61231 Bad Nauheim, Germany; (E.A.); (P.S.); (R.S.); (W.S.)
- Department of Internal Medicine, Justus Liebig University, 35392 Giessen, Germany
- Correspondence: ; Tel.: +49-603-270-5380; Fax: +49-603-270-5385
| |
Collapse
|
8
|
Wang M, Wang K, Liao X, Hu H, Chen L, Meng L, Gao W, Li Q. Carnitine Palmitoyltransferase System: A New Target for Anti-Inflammatory and Anticancer Therapy? Front Pharmacol 2021; 12:760581. [PMID: 34764874 PMCID: PMC8576433 DOI: 10.3389/fphar.2021.760581] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Accepted: 09/17/2021] [Indexed: 11/20/2022] Open
Abstract
Lipid metabolism involves multiple biological processes. As one of the most important lipid metabolic pathways, fatty acid oxidation (FAO) and its key rate-limiting enzyme, the carnitine palmitoyltransferase (CPT) system, regulate host immune responses and thus are of great clinical significance. The effect of the CPT system on different tissues or organs is complex: the deficiency or over-activation of CPT disrupts the immune homeostasis by causing energy metabolism disorder and inflammatory oxidative damage and therefore contributes to the development of various acute and chronic inflammatory disorders and cancer. Accordingly, agonists or antagonists targeting the CPT system may become novel approaches for the treatment of diseases. In this review, we first briefly describe the structure, distribution, and physiological action of the CPT system. We then summarize the pathophysiological role of the CPT system in chronic obstructive pulmonary disease, bronchial asthma, acute lung injury, chronic granulomatous disease, nonalcoholic fatty liver disease, hepatic ischemia–reperfusion injury, kidney fibrosis, acute kidney injury, cardiovascular disorders, and cancer. We are also concerned with the current knowledge in either preclinical or clinical studies of various CPT activators/inhibitors for the management of diseases. These compounds range from traditional Chinese medicines to novel nanodevices. Although great efforts have been made in studying the different kinds of CPT agonists/antagonists, only a few pharmaceuticals have been applied for clinical uses. Nevertheless, research on CPT activation or inhibition highlights the pharmacological modulation of CPT-dependent FAO, especially on different CPT isoforms, as a promising anti-inflammatory/antitumor therapeutic strategy for numerous disorders.
Collapse
Affiliation(s)
- Muyun Wang
- Department of Pulmonary and Critical Care Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Kun Wang
- Department of Pulmonary and Critical Care Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Ximing Liao
- Department of Pulmonary and Critical Care Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Haiyang Hu
- Department of Vascular Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Liangzhi Chen
- Department of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Linlin Meng
- Department of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Wei Gao
- Department of Pulmonary and Critical Care Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Qiang Li
- Department of Pulmonary and Critical Care Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
9
|
Huo X, Jin S, Wang Y, Ma L. DNA methylation in chronic obstructive pulmonary disease. Epigenomics 2021; 13:1145-1155. [PMID: 34142873 DOI: 10.2217/epi-2021-0111] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Chronic obstructive pulmonary disease (COPD), a complex disease with polygenetic tendency, is one of the most important health problems in the world. Recently, in the study of the pathogenesis of the COPD, epigenetic changes caused by environmental factors, such as DNA methylation, started to attract more attention than genetic factors. In this review, we discuss the main features of DNA methylation, such as DNA methyltransferases and the methylation sites that modulate the DNA methylation level, and their roles in COPD progression. Finally, to promote new ideas for the prevention and treatment of COPD, we focus on the potential of DNA methylation as a COPD therapeutic target.
Collapse
Affiliation(s)
- XinXin Huo
- School of Public Health, Lanzhou University, Lanzhou, China
| | - SiHui Jin
- School of Public Health, Lanzhou University, Lanzhou, China
| | - YiGe Wang
- School of Public Health, Lanzhou University, Lanzhou, China
| | - Li Ma
- School of Public Health, Lanzhou University, Lanzhou, China
| |
Collapse
|
10
|
Zheng L, Jiang YL, Fei J, Cao P, Zhang C, Xie GF, Wang LX, Cao W, Fu L, Zhao H. Circulatory cadmium positively correlates with epithelial-mesenchymal transition in patients with chronic obstructive pulmonary disease. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2021; 215:112164. [PMID: 33756289 DOI: 10.1016/j.ecoenv.2021.112164] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 03/02/2021] [Accepted: 03/15/2021] [Indexed: 06/12/2023]
Abstract
Environmental cadmium (Cd) exposure can cause several pulmonary diseases. Epithelial-mesenchymal transition (EMT) involved in the process of chronic obstructive pulmonary disease (COPD). However, the association between environmental Cd exposure and EMT was unclear in COPD patients. This study aimed to analyze the associations among circulatory Cd, EMT and COPD based on case-control study. Four hundred COPD patients and 400 control subjects were recruited. Circulatory Cd was detected using atomic adsorption spectrometer. MicroRNA-30 (miR-30) was measured by RT-PCR and the markers of pulmonary EMT were evaluated through western blotting. Circulatory Cd concentration was increased and serum miR-30 was decreased in COPD patients. Circulatory Cd was inversely associated with pulmonary function in COPD patients. Moreover, serum miR-30 was gradually decreased in parallel with FEV1 in COPD patients. Meanwhile, there was a negative association between serum miR-30 and circulatory Cd in COPD patients. Further analysis found that E-cadherin, one of epithelial biomarkers, was reduced in lung tissues of COPD patients with higher circulatory Cd. On the contrary, pulmonary N-cadherin, Vimentin and α-SMA, three of mesenchymal biomarkers, were increased in COPD patients with higher circulatory Cd. In vitro experiments revealed that Cd exposure repressed miR-30 levels and promoted EMT in BEAS-2B cells. Our results provide evidence that miR-30 reduction contributing to pulmonary EMT may involve in the process of Cd-induced COPD.
Collapse
Affiliation(s)
- Ling Zheng
- The Second Affiliated Hospital, Anhui Medical University, Hefei 230032, China
| | - Ya-Lin Jiang
- The Bozhou People's Hospital, Bozhou 236800, China
| | - Jun Fei
- The Second Affiliated Hospital, Anhui Medical University, Hefei 230032, China
| | - Peng Cao
- The Second Affiliated Hospital, Anhui Medical University, Hefei 230032, China
| | - Chen Zhang
- The Second Affiliated Hospital, Anhui Medical University, Hefei 230032, China
| | - Guo-Fang Xie
- The Second Affiliated Hospital, Anhui Medical University, Hefei 230032, China
| | - Li-Xiang Wang
- The Second Affiliated Hospital, Anhui Medical University, Hefei 230032, China
| | - Wei Cao
- The Second Affiliated Hospital, Anhui Medical University, Hefei 230032, China
| | - Lin Fu
- The Second Affiliated Hospital, Anhui Medical University, Hefei 230032, China; Department of Toxicology, Anhui Medical University, Hefei 230032, China.
| | - Hui Zhao
- The Second Affiliated Hospital, Anhui Medical University, Hefei 230032, China.
| |
Collapse
|
11
|
Li T, He X, Luo L, Zeng H, Ren S, Chen Y. F-Box Protein FBXW17-Mediated Proteasomal Degradation of Protein Methyltransferase PRMT6 Exaggerates CSE-Induced Lung Epithelial Inflammation and Apoptosis. Front Cell Dev Biol 2021; 9:599020. [PMID: 33959602 PMCID: PMC8095709 DOI: 10.3389/fcell.2021.599020] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Accepted: 02/02/2021] [Indexed: 11/13/2022] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a chronic debilitating lung disease, characterized by progressive airway inflammation and lung structural cell death. Cigarette smoke is considered the most common risk factor of COPD pathogenesis. Understanding the molecular mechanisms of persistent inflammation and epithelial apoptosis induced by cigarette smoke would be extremely beneficial for improving the treatment and prevention of COPD. A histone methyl modifier, protein arginine N-methyltransferase 6 (PRMT6), is reported to alleviate cigarette smoke extract (CSE)-induced emphysema through inhibiting inflammation and cell apoptosis. However, few studies have focused on the modulation of PRMT6 in regulating inflammation and cell apoptosis. In this study, we showed that protein expression of PRMT6 was aberrantly decreased in the lung tissue of COPD patients and CSE-treated epithelial cells. FBXW17, a member of the Skp1-Cullin-F-box (SCF) family of E3 ubiquitin ligases, selectively bound to PRMT6 in nuclei to modulate its elimination in the proteasome system. Proteasome inhibitor or silencing of FBXW17 abrogated CSE-induced PRMT6 protein degradation. Furthermore, negative alteration of FBXW17/PRMT6 signaling lessened the proapoptotic and proinflammatory effects of CSE in lung epithelial cells. Our study, therefore, provides a potential therapeutic target against the airway inflammation and cell death in CS-induced COPD.
Collapse
Affiliation(s)
- Tiao Li
- Department of Respiratory Medicine, The Second Xiangya Hospital of Central South University, Changsha, China.,Research Unit of Respiratory Disease, Central South University, Changsha, China.,Diagnosis and Treatment Center of Respiratory Disease, Central South University, Changsha, China
| | - Xue He
- Department of Respiratory Medicine, The Second Xiangya Hospital of Central South University, Changsha, China.,Research Unit of Respiratory Disease, Central South University, Changsha, China.,Diagnosis and Treatment Center of Respiratory Disease, Central South University, Changsha, China
| | - Lijuan Luo
- Department of Respiratory Medicine, The Second Xiangya Hospital of Central South University, Changsha, China.,Research Unit of Respiratory Disease, Central South University, Changsha, China.,Diagnosis and Treatment Center of Respiratory Disease, Central South University, Changsha, China
| | - Huihui Zeng
- Department of Respiratory Medicine, The Second Xiangya Hospital of Central South University, Changsha, China.,Research Unit of Respiratory Disease, Central South University, Changsha, China.,Diagnosis and Treatment Center of Respiratory Disease, Central South University, Changsha, China
| | - Siying Ren
- Department of Respiratory Medicine, The Second Xiangya Hospital of Central South University, Changsha, China.,Research Unit of Respiratory Disease, Central South University, Changsha, China.,Diagnosis and Treatment Center of Respiratory Disease, Central South University, Changsha, China
| | - Yan Chen
- Department of Respiratory Medicine, The Second Xiangya Hospital of Central South University, Changsha, China.,Research Unit of Respiratory Disease, Central South University, Changsha, China.,Diagnosis and Treatment Center of Respiratory Disease, Central South University, Changsha, China
| |
Collapse
|
12
|
Gondim FDL, Moura MF, Ferreira RM, Serra DS, Araújo RS, Oliveira MLMD, Cavalcante FSÁ. Exposure to total particulate matter obtained from combustion of diesel vehicles (EURO 3 and EURO 5): Effects on the respiratory systems of emphysematous mice. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2021; 83:103583. [PMID: 33434645 DOI: 10.1016/j.etap.2021.103583] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 10/16/2020] [Accepted: 01/06/2021] [Indexed: 06/12/2023]
Abstract
Air pollution has association with chronic obstructive pulmonary disease (COPD) and reduced life expectancy. This study investigated the deleterious effects caused by tobacco smoke and diesel exhaust particles (DEP) from vehicles operating under EURO 3 and EURO 5 standards. Experiments were carried out on C57BL/6 mice divided into six groups: control group, group exposed to cigarette smoke (CS), two groups exposed to DEP (AAE3 and AAE5), and two groups exposed to tobacco smoke and vehicle DEP (CSE3 and CSE5). Results showed that, when compared to AA, groups AAE3 and AAE5 showed changes in respiratory mechanics, and that DEP originating from EURO 5 diesel vehicles was less harmful when compared to DEP originating from EURO 3 diesel vehicles. Analyses of groups CSE3 and CSE5 revealed increased inspiratory capacity and decreased tissue elastance, when compared to their respective controls, suggesting an exacerbation of changes in respiratory system mechanics compatible with COPD development.
Collapse
|
13
|
Li N, Li S, Wu Y, Xiong L, Li T, Xing D, Li Q, Wu D. Dexmedetomidine targets miR-146a and participates in the progress of chronic obstructive pulmonary disease in vivo and in vitro. Genes Genomics 2021; 43:1371-1379. [PMID: 33945148 DOI: 10.1007/s13258-020-01019-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Accepted: 11/17/2020] [Indexed: 11/28/2022]
Abstract
BACKGROUND Chronic obstructive pulmonary disease (COPD) is a chronic lung disease and the third leading cause of death in the world. Dexmedetomidine has been reported to effectively inhibit histamine-induced bronchoconstriction. However, the molecular mechanism of dexmedetomidine in COPD has not been found. OBJECTIVE To explore the role and mechanism of dexmedetomidine in COPD, and to provide theoretical basis for clinical treatment of COPD. METHODS The expression of miR-146a was regulated by mimics or inhibitor and the relative expression of apoptotic proteins p53, Bax and Bcl-2 in human bronchial epithelial 16HBE cells was determined by real-time PCR and Western blot. Dexmedetomidine was treated for 16HBE cells and alveolar epithelial type II cells (AEC2), the cell apoptosis was detected by TUNEL and Hoechst33342 staining. A COPD rat model was established by smoking to test the effects of dexmedetomidine on the progression of COPD. The levels of IL-6, IL-1β and TNF-α in serum were measured by ELISA and the protein concentration of bronchoalveolar lavage fluid (BALF) was also detected in dexmedetomidine treated COPD rat model. RESULTS miR-146a promoted 16HBE cell apoptosis and reduced cell proliferation. Additionally, dexmedetomidine was showed to reduce the 16HBEL cell apoptosis through reducing the expression of miR-146a. Moreover, dexmedetomidine regulated cell apoptosis and cell apoptosis through miR-146a in AEC2 cells. More importantly, dexmedetomidine attenuated the morphology and pathology of COPD rat model. CONCLUSION Dexmedetomidine reduced 16HBE cells and AEC2 cell apoptosis and attenuated COPD by down-regulating miR-146a.
Collapse
Affiliation(s)
- Na Li
- Department of Anesthesiology, Hainan General Hospital, Road Xiuhua, Haikou, Hainan Province, China
| | - Shuangfeng Li
- Department of Anesthesiology, Hainan General Hospital, Road Xiuhua, Haikou, Hainan Province, China
| | - Yehua Wu
- Department of Anesthesiology, Hainan General Hospital, Road Xiuhua, Haikou, Hainan Province, China
| | - Lu Xiong
- Department of Anesthesiology, Hainan General Hospital, Road Xiuhua, Haikou, Hainan Province, China
| | - Tiejun Li
- Department of Anesthesiology, Hainan General Hospital, Road Xiuhua, Haikou, Hainan Province, China
| | - Dandan Xing
- Department of Anesthesiology, Hainan General Hospital, Road Xiuhua, Haikou, Hainan Province, China
| | - Qiuchang Li
- Department of Anesthesiology, Hainan General Hospital, Road Xiuhua, Haikou, Hainan Province, China
| | - Duozhi Wu
- Department of Anesthesiology, Hainan General Hospital, Road Xiuhua, Haikou, Hainan Province, China.
| |
Collapse
|
14
|
Analysis of Acrolein Exposure Induced Pulmonary Response in Seven Inbred Mouse Strains and Human Primary Bronchial Epithelial Cells Cultured at Air-Liquid Interface. BIOMED RESEARCH INTERNATIONAL 2020; 2020:3259723. [PMID: 33110918 PMCID: PMC7582059 DOI: 10.1155/2020/3259723] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 09/07/2020] [Accepted: 09/15/2020] [Indexed: 11/23/2022]
Abstract
Background Acrolein is a major component of environmental pollutants, cigarette smoke, and is also formed by heating cooking oil. We evaluated the interstrain variability of response to subchronic inhalation exposure to acrolein among inbred mouse strains for inflammation, oxidative stress, and tissue injury responses. Furthermore, we studied the response to acrolein vapor in the lung mucosa model using human primary bronchial epithelial cells (PBEC) cultured at an air-liquid interface (ALI) to evaluate the findings of mouse studies. Methods Female 129S1/SvlmJ, A/J, BALB/cByJ, C3H/HeJ, C57BL/6J, DBA/2J, and FVB/NJ mice were exposed to 1 part per million (ppm) acrolein or filtered air for 11 weeks. Total cell counts and protein concentrations were measured in bronchoalveolar lavage (BAL) fluid to assess airway inflammation and membrane integrity. PBEC-ALI models were exposed to acrolein vapor (0.1 and 0.2 ppm) for 30 minutes. Gene expression of proinflammatory, oxidative stress, and tissue injury-repair markers was assessed (cut off: ≥2 folds; p < 0.05) in the lung models. Results Total BAL cell numbers and protein concentrations remained unchanged following acrolein exposure in all mouse strains. BALB/cByJ, C57BL/6J, and 129S1/SvlmJ strains were the most affected with an increased expression of proinflammatory, oxidative stress, and/or tissue injury markers. DBA/2J, C3H/HeJ, A/J, and FVB/NJ were affected to a lesser extent. Both matrix metalloproteinase 9 (Mmp9) and tissue inhibitor of metalloproteinase 1 (Timp1) were upregulated in the strains DBA/2J, C3H/HeJ, and FVB/NJ indicating altered protease/antiprotease balance. Upregulation of lung interleukin- (IL-) 17b transcript in the susceptible strains led us to investigate the IL-17 pathway genes in the PBEC-ALI model. Acrolein exposure resulted in an increased expression of IL-17A, C, and D; IL-1B; IL-22; and RAR-related orphan receptor A in the PBEC-ALI model. Conclusion The interstrain differences in response to subchronic acrolein exposure in mouse suggest a genetic predisposition. Altered expression of IL-17 pathway genes following acrolein exposure in the PBEC-ALI models indicates that it has a central role in chemical irritant toxicity. The findings also indicate that genetically determined differences in IL-17 signaling pathway genes in the different mouse strains may explain their susceptibility to different chemical irritants.
Collapse
|
15
|
Easter M, Bollenbecker S, Barnes JW, Krick S. Targeting Aging Pathways in Chronic Obstructive Pulmonary Disease. Int J Mol Sci 2020; 21:E6924. [PMID: 32967225 PMCID: PMC7555616 DOI: 10.3390/ijms21186924] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 09/05/2020] [Accepted: 09/15/2020] [Indexed: 12/17/2022] Open
Abstract
Chronic obstructive pulmonary disease (COPD) has become a global epidemic and is the third leading cause of death worldwide. COPD is characterized by chronic airway inflammation, loss of alveolar-capillary units, and progressive decline in lung function. Major risk factors for COPD are cigarette smoking and aging. COPD-associated pathomechanisms include multiple aging pathways such as telomere attrition, epigenetic alterations, altered nutrient sensing, mitochondrial dysfunction, cell senescence, stem cell exhaustion and chronic inflammation. In this review, we will highlight the current literature that focuses on the role of age and aging-associated signaling pathways as well as their impact on current treatment strategies in the pathogenesis of COPD. Furthermore, we will discuss established and experimental COPD treatments including senolytic and anti-aging therapies and their potential use as novel treatment strategies in COPD.
Collapse
Affiliation(s)
- Molly Easter
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL 35294, USA; (M.E.); (S.B.); (J.W.B.)
| | - Seth Bollenbecker
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL 35294, USA; (M.E.); (S.B.); (J.W.B.)
| | - Jarrod W. Barnes
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL 35294, USA; (M.E.); (S.B.); (J.W.B.)
- Gregory Fleming James Cystic Fibrosis Center, The University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Stefanie Krick
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL 35294, USA; (M.E.); (S.B.); (J.W.B.)
- Gregory Fleming James Cystic Fibrosis Center, The University of Alabama at Birmingham, Birmingham, AL 35294, USA
| |
Collapse
|
16
|
Zhang L, Valizadeh H, Alipourfard I, Bidares R, Aghebati-Maleki L, Ahmadi M. Epigenetic Modifications and Therapy in Chronic Obstructive Pulmonary Disease (COPD): An Update Review. COPD 2020; 17:333-342. [PMID: 32558592 DOI: 10.1080/15412555.2020.1780576] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Chronic obstructive pulmonary disease (COPD) that is one of the most prevalent chronic adult diseases and the third leading cause of fatality until 2020. Elastase/anti-elastase hypothesis, chronic inflammation, apoptosis, oxidant-antioxidant balance and infective repair cause pathogenesis of COPD are among the factors at play. Epigenetic changes are post-translational modifications in histone proteins and DNA such as methylation and acetylation as well as dysregulation of miRNAs expression. In this update review, we have examined recent studies on the upregulation or downregulation of methylation in different genes associated with COPD. Dysregulation of HDAC activity which is caused by some factors and miRNAs plays a key role in the suppression and reduction of COPD development. Also, some therapeutic approaches are proposed against COPD by targeting HDAC2 and miRNAs, which have therapeutic effects.
Collapse
Affiliation(s)
- Lingzhi Zhang
- Department of Emergency Medicine, Shengjing Hospital of China Medical University, Shenyang, China
| | - Hamed Valizadeh
- Department of Internal Medicine and Pulmonology, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran.,Tuberculosis and Lung Disease Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Iraj Alipourfard
- Faculty of Life Sciences, Center of pharmaceutical sciences, University of Vienna, Vienna, Austria.,Faculty of Sciences, School of Pharmacy, University of Rome Tor Vergata, Roma, Italy
| | - Ramtin Bidares
- Department of Experimental Medicine, Sapienza University, Rome, Italy
| | | | - Majid Ahmadi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
17
|
Moll M, Regan EA, Hokanson JE, Lutz SM, Silverman EK, Crapo JD, Make BJ, DeMeo DL. The Association of Multiparity with Lung Function and Chronic Obstructive Pulmonary Disease-Related Phenotypes. CHRONIC OBSTRUCTIVE PULMONARY DISEASES (MIAMI, FLA.) 2020; 7:86-98. [PMID: 32324980 PMCID: PMC7454017 DOI: 10.15326/jcopdf.7.2.2019.0166] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
BACKGROUND Apparent increased female susceptibility to chronic obstructive pulmonary disease (COPD) suggests sex hormones modulate disease pathogenesis. Little is known about associations between multiparity and lung function in smokers. RESEARCH QUESTION We hypothesized that multiparity is associated with lung function and measures of emphysema and airway disease. STUDY DESIGN AND METHODS Utilizing female participants from the 5-year follow up of the COPD Genetic Epidemiology (COPDGene®) study we performed multivariable linear regressions to assess the effect of multiparity and number of pregnancies on forced expiratory volume in 1 second (FEV1) percentage of predicted (% predicted), FEV1/forced vital capacity (FVC), percent emphysema on computed tomography (CT) scans, and Pi10, a measure of airway thickening. We sampled never smokers and those with lower smoking exposure from the National Health and Nutrition Examination Survey (NHANES) 2011-2012 dataset. RESULTS We included 1820 participants from COPDGene® and 418 participants from NHANES (321 never smokers, 97 ever smokers). In COPDGene®, multiparity (beta coefficient [β] = -3.8, 95% confidence interval [CI]: [-6.5, -1.1], p = 0.005) and higher number of pregnancies were associated with lower FEV1 % predicted. Multiparity was not associated with percent emphysema or Pi10. In individuals with no or mild obstruction, multiparity was associated with lower FEV1 % predicted. There was an interaction with multiparity and age on FEV1 % predicted (p = 0.025). In NHANES, there was no association between multiparity and FEV1 % predicted in never smokers or the lower smoking exposure group. INTERPRETATION Multiparity was associated with lower FEV1 % predicted in current and former smokers in COPDGene® study participants. These preliminary results emphasize the importance of smoking abstinence in women of child-bearing age.
Collapse
Affiliation(s)
- Matthew Moll
- Channing Division of Network Medicine, Brigham and Women’s Hospital, Boston, Massachusetts,Division of Pulmonary and Critical Care Medicine, Brigham and Women’s Hospital, Boston, Massachusetts
| | | | - John E. Hokanson
- Department of Epidemiology, Colorado School of Public Health, University of Colorado Anschutz Medical Campus, Aurora
| | - Sharon M. Lutz
- PRecisiOn Medicine Translational Research (PROMoTeR) Center, Department of Population Medicine, Harvard Medical School and Harvard Pilgrim Health Care, Boston, Massachusetts
| | - Edwin K. Silverman
- Channing Division of Network Medicine, Brigham and Women’s Hospital, Boston, Massachusetts,Division of Pulmonary and Critical Care Medicine, Brigham and Women’s Hospital, Boston, Massachusetts,Harvard Medical School, Boston, Massachusetts
| | - James D. Crapo
- Division of Pulmonary, Critical Care and Sleep Medicine, National Jewish Health, Denver, Colorado
| | - Barry J. Make
- Division of Pulmonary, Critical Care and Sleep Medicine, National Jewish Health, Denver, Colorado
| | - Dawn L. DeMeo
- Channing Division of Network Medicine, Brigham and Women’s Hospital, Boston, Massachusetts,Division of Pulmonary and Critical Care Medicine, Brigham and Women’s Hospital, Boston, Massachusetts
| |
Collapse
|
18
|
Poole JA, Nordgren TM, Heires AJ, Nelson AJ, Katafiasz D, Bailey KL, Romberger DJ. Amphiregulin modulates murine lung recovery and fibroblast function following exposure to agriculture organic dust. Am J Physiol Lung Cell Mol Physiol 2020; 318:L180-L191. [PMID: 31693392 PMCID: PMC6985879 DOI: 10.1152/ajplung.00039.2019] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Revised: 10/22/2019] [Accepted: 10/29/2019] [Indexed: 01/10/2023] Open
Abstract
Exposure to agricultural bioaerosols can lead to chronic inflammatory lung diseases. Amphiregulin (AREG) can promote the lung repair process but can also lead to fibrotic remodeling. The objective of this study was to determine the role of AREG in altering recovery from environmental dust exposure in a murine in vivo model and in vitro using cultured human and murine lung fibroblasts. C57BL/6 mice were intranasally exposed to swine confinement facility dust extract (DE) or saline daily for 1 wk or allowed to recover for 3-7 days while being treated with an AREG-neutralizing antibody or recombinant AREG. Treatment with the anti-AREG antibody prevented resolution of DE exposure-induced airway influx of total cells, neutrophils, and macrophages and increased levels of TNF-α, IL-6, and CXCL1. Neutrophils and activated macrophages (CD11c+CD11bhi) persisted after recovery in lung tissues of anti-AREG-treated mice. In murine and human lung fibroblasts, DE induced the release of AREG and inflammatory cytokines. Fibroblast recellularization of primary human lung mesenchymal matrix scaffolds and wound closure was inhibited by DE and enhanced with recombinant AREG alone. AREG treatment rescued the DE-induced inhibitory fibroblast effects. AREG intranasal treatment for 3 days during recovery phase reduced repetitive DE-induced airway inflammatory cell influx and cytokine release. Collectively, these studies demonstrate that inhibition of AREG reduced, whereas AREG supplementation promoted, the airway inflammatory recovery response following environmental bioaerosol exposure, and AREG enhanced fibroblast function, suggesting that AREG could be targeted in agricultural workers repetitively exposed to organic dust environments to potentially prevent and/or reduce disease.
Collapse
Affiliation(s)
- Jill A Poole
- Pulmonary, Critical Care, Sleep & Allergy Division, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, Nebraska
| | - Tara M Nordgren
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, California
| | - Art J Heires
- Pulmonary, Critical Care, Sleep & Allergy Division, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, Nebraska
| | - Amy J Nelson
- Pulmonary, Critical Care, Sleep & Allergy Division, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, Nebraska
| | - Dawn Katafiasz
- Pulmonary, Critical Care, Sleep & Allergy Division, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, Nebraska
| | - Kristina L Bailey
- Pulmonary, Critical Care, Sleep & Allergy Division, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, Nebraska
- Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, Nebraska
| | - Debra J Romberger
- Pulmonary, Critical Care, Sleep & Allergy Division, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, Nebraska
- Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, Nebraska
| |
Collapse
|
19
|
Nader CP, Cidem A, Verrills NM, Ammit AJ. Protein phosphatase 2A (PP2A): a key phosphatase in the progression of chronic obstructive pulmonary disease (COPD) to lung cancer. Respir Res 2019; 20:222. [PMID: 31623614 PMCID: PMC6798356 DOI: 10.1186/s12931-019-1192-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2019] [Accepted: 09/20/2019] [Indexed: 02/06/2023] Open
Abstract
Lung cancer (LC) has the highest relative risk of development as a comorbidity of chronic obstructive pulmonary disease (COPD). The molecular mechanisms that mediate chronic inflammation and lung function impairment in COPD have been identified in LC. This suggests the two diseases are more linked than once thought. Emerging data in relation to a key phosphatase, protein phosphatase 2A (PP2A), and its regulatory role in inflammatory and tumour suppression in both disease settings suggests that it may be critical in the progression of COPD to LC. In this review, we uncover the importance of the functional and active PP2A holoenzyme in the context of both diseases. We describe PP2A inactivation via direct and indirect means and explore the actions of two key PP2A endogenous inhibitors, cancerous inhibitor of PP2A (CIP2A) and inhibitor 2 of PP2A (SET), and the role they play in COPD and LC. We explain how dysregulation of PP2A in COPD creates a favourable inflammatory micro-environment and promotes the initiation and progression of tumour pathogenesis. Finally, we highlight PP2A as a druggable target in the treatment of COPD and LC and demonstrate the potential of PP2A re-activation as a strategy to halt COPD disease progression to LC. Although further studies are required to elucidate if PP2A activity in COPD is a causal link for LC progression, studies focused on the potential of PP2A reactivating agents to reduce the risk of LC formation in COPD patients will be pivotal in improving clinical outcomes for both COPD and LC patients in the future.
Collapse
Affiliation(s)
- Cassandra P Nader
- Woolcock Emphysema Centre, Woolcock Institute of Medical Research, University of Sydney, Sydney, NSW, Australia
| | - Aylin Cidem
- Woolcock Emphysema Centre, Woolcock Institute of Medical Research, University of Sydney, Sydney, NSW, Australia
| | - Nicole M Verrills
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, University of Newcastle, Callaghan, NSW, 2308, Australia
- Priority Research Centre for Cancer Research, Innovation & Translation, Faculty of Health & Medicine, Hunter Medical Research Institute, New Lambton Heights, NSW, 2305, Australia
| | - Alaina J Ammit
- Woolcock Emphysema Centre, Woolcock Institute of Medical Research, University of Sydney, Sydney, NSW, Australia.
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW, Australia.
| |
Collapse
|
20
|
Eskandari M, Nordgren TM, O'Connell GD. Mechanics of pulmonary airways: Linking structure to function through constitutive modeling, biochemistry, and histology. Acta Biomater 2019; 97:513-523. [PMID: 31330329 DOI: 10.1016/j.actbio.2019.07.020] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Revised: 07/07/2019] [Accepted: 07/11/2019] [Indexed: 12/24/2022]
Abstract
Breathing involves fluid-solid interactions in the lung; however, the lack of experimental data inhibits combining the mechanics of air flow to airway deformation, challenging the understanding of how biomaterial constituents contribute to tissue response. As such, lung mechanics research is increasingly focused on exploring the relationship between structure and function. To address these needs, we characterize mechanical properties of porcine airways using uniaxial tensile experiments, accounting for bronchial orientation- and location- dependency. Structurally-reinforced constitutive models are developed to incorporate the role of collagen and elastin fibers embedded within the extrafibrillar matrix. The strain-energy function combines a matrix description (evaluating six models: compressible NeoHookean, unconstrained Ogden, uncoupled Mooney-Rivlin, incompressible Ogden, incompressible Demiray and incompressible NeoHookean), superimposed with non-linear fibers (evaluating two models: exponential and polynomial). The best constitutive formulation representative of all bronchial regions is determined based on curve-fit results to experimental data, accounting for uniqueness and sensitivity. Glycosaminoglycan and collagen composition, alongside tissue architecture, indicate fiber form to be primarily responsible for observed airway anisotropy and heterogeneous mechanical behavior. To the authors' best knowledge, this study is the first to formulate a structurally-motivated constitutive model, augmented with biochemical analysis and microstructural observations, to investigate the mechanical function of proximal and distal bronchi. Our systematic pulmonary tissue characterization provides a necessary foundation for understanding pulmonary mechanics; furthermore, these results enable clinical translation through simulations of airway obstruction in disease, fluid-structure interaction insights during breathing, and potentially, predictive capabilities for medical interventions. STATEMENT OF SIGNIFICANCE: The advancement of pulmonary research relies on investigating the biomechanical response of the bronchial tree. Experiments demonstrating the non-linear, heterogeneous, and anisotropic material behavior of porcine airways are used to develop a structural constitutive model representative of proximal and distal bronchial behavior. Calibrated material parameters exhibit regional variation in biomaterial properties, initially hypothesized to originate from tissue constituents. Further exploration through biochemical and histological analysis indicates mechanical function is primarily governed by microstructural form. The results of this study can be directly used in finite element and fluid-structure interaction models to enable physiologically relevant and more accurate computational simulations aimed to help diagnose and monitor pulmonary disease.
Collapse
Affiliation(s)
- Mona Eskandari
- Department of Mechanical Engineering, University of California at Riverside, Riverside, CA 92521, USA; Department of Bioengineering, University of California at Riverside, Riverside, CA 92521, USA; BREATHE Center School of Medicine, University of California at Riverside, Riverside, CA 92521, USA; Department of Mechanical Engineering, University of California at Berkeley, Berkeley, CA 94720, USA.
| | - Tara M Nordgren
- Division of Biomedical Sciences, University of California at Riverside, Riverside, CA 92521, USA; BREATHE Center School of Medicine, University of California at Riverside, Riverside, CA 92521, USA
| | - Grace D O'Connell
- Department of Mechanical Engineering, University of California at Berkeley, Berkeley, CA 94720, USA; Department of Orthopaedic Surgery, University of California at San Francisco, San Francisco, CA 94143, USA
| |
Collapse
|
21
|
Abstract
Introduction: Neutrophils are the most abundant inflammatory cells in the lungs of patients with chronic lung diseases, especially COPD, yet despite this, patients often experience repeated chest infections. Neutrophil function may be altered in disease, but the reasons are unclear. In chronic disease, sequential pro-inflammatory and pro-repair responses appear distorted. As understanding of neutrophil heterogeneity has expanded, it is suggested that different neutrophil phenotypes may impact on health and disease. Areas covered: In this review, the definition of cellular phenotype, the implication of neutrophil surface markers and functions in chronic lung disease and the complex influences of external, local and genetic factors on these changes are discussed. Literature was accessed up to the 19 July 2019 using: PubMed, US National Library of Medicine National Institutes of Health and the National Centre for Biotechnology Information. Expert opinion: As more is learned about neutrophils, the further we step from the classical view of neutrophils being unrefined killing machines to highly complex and finely tuned cells. Future therapeutics may aim to normalize neutrophil function, but to achieve this, knowledge of phenotypes in humans and how these relate to observed pathology and disease processes is required.
Collapse
Affiliation(s)
- Michael J Hughes
- Birmingham Acute Care Research Group, Institute of Inflammation and Ageing, University of Birmingham , Birmingham , UK
| | - Elizabeth Sapey
- Birmingham Acute Care Research Group, Institute of Inflammation and Ageing, University of Birmingham , Birmingham , UK
| | - Robert Stockley
- Birmingham Acute Care Research Group, Institute of Inflammation and Ageing, University of Birmingham , Birmingham , UK
| |
Collapse
|
22
|
Respiratory Sound Based Classification of Chronic Obstructive Pulmonary Disease: a Risk Stratification Approach in Machine Learning Paradigm. J Med Syst 2019; 43:255. [PMID: 31254141 DOI: 10.1007/s10916-019-1388-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Accepted: 06/14/2019] [Indexed: 10/26/2022]
Abstract
This article investigates the classification of normal and COPD subjects on the basis of respiratory sound analysis using machine learning techniques. Thirty COPD and 25 healthy subject data are recorded. Total of 39 lung sound features and 3 spirometry features are extracted and evaluated. Various parametric and nonparametric tests are conducted to evaluate the relevance of extracted features. Classifiers such as support vector machine (SVM), k-nearest neighbor (KNN), logistic regression (LR), decision tree and discriminant analysis (DA) are used to categorize normal and COPD breath sounds. Classification based on spirometry parameters as well as respiratory sound parameters are assessed. Maximum classification accuracy of 83.6% is achieved by the SVM classifier while using the most relevant lung sound parameters i.e. median frequency and linear predictive coefficients. Further, SVM classifier and LR classifier achieved classification accuracy of 100% when relevant lung sound parameters, i.e. median frequency and linear predictive coefficient are combined with the spirometry parameters, i.e. forced vital capacity (FVC) and forced expiratory volume in 1 s (FEV1). It is concluded that combining lung sound based features with spirometry data can improve the accuracy of COPD diagnosis and hence the clinician's performance in routine clinical practice. The proposed approach is of great significance in a clinical scenario wherein it can be used to assist clinicians for automated COPD diagnosis. A complete handheld medical system can be developed in the future incorporating lung sounds for COPD diagnosis using machine learning techniques.
Collapse
|
23
|
Cigarette Smoke Induced Lung Barrier Dysfunction, EMT, and Tissue Remodeling: A Possible Link between COPD and Lung Cancer. BIOMED RESEARCH INTERNATIONAL 2019; 2019:2025636. [PMID: 31341890 PMCID: PMC6613007 DOI: 10.1155/2019/2025636] [Citation(s) in RCA: 101] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Accepted: 06/02/2019] [Indexed: 12/13/2022]
Abstract
Chronic obstructive pulmonary disease (COPD) and lung cancer, closely related to smoking, are major lung diseases affecting millions of individuals worldwide. The generated gas mixture of smoking is proved to contain about 4,500 components such as carbon monoxide, nicotine, oxidants, fine particulate matter, and aldehydes. These components were considered to be the principle factor driving the pathogenesis and progression of pulmonary disease. A large proportion of lung cancer patients showed a history of COPD, which demonstrated that there might be a close relationship between COPD and lung cancer. In the early stages of smoking, lung barrier provoked protective response and DNA repair are likely to suppress these changes to a certain extent. In the presence of long-term smoking exposure, these mechanisms seem to be malfunctioned and lead to disease progression. The infiltration of inflammatory cells to mucosa, submucosa, and glandular tissue caused by inhaled cigarette smoke is responsible for the destruction of matrix, blood supply shortage, and epithelial cell death. Conversely, cancer cells have the capacity to modulate the proliferation of epithelial cells and produce of new vascular networks. Comprehension understanding of mechanisms responsible for both pathologies is necessary for the prevention and treatment of COPD and lung cancer. In this review, we will summarize related articles and give a glance of possible mechanism between cigarette smoking induced COPD and lung cancer.
Collapse
|
24
|
Muhammed A, Moiz JA, Singla D, Ali MS, Talwar D. Postural abnormalities in phenotypes of chronic obstructive pulmonary disease. Braz J Phys Ther 2019; 24:325-332. [PMID: 31151892 DOI: 10.1016/j.bjpt.2019.05.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Revised: 03/16/2019] [Accepted: 05/14/2019] [Indexed: 10/26/2022] Open
Abstract
BACKGROUND Few studies have demonstrated postural abnormalities in patients with chronic obstructive pulmonary disease - when compared with healthy individuals. However, none of these studies have compared postural abnormalities in different phenotypes of chronic obstructive pulmonary disease. OBJECTIVE To compare the thoracic posture between two phenotypes of chronic obstructive pulmonary disease (emphysema and chronic bronchitis) with healthy individuals. METHODS Forty individuals with chronic obstructive pulmonary disease (20 with chronic bronchitis, 67±3.5 years, 20 with emphysema, 67.7±4 years) and 20 age-matched healthy individuals (67.3±3.9 years) underwent postural assessment which was performed using photogrammetric measurements of head protraction, shoulder protraction, thoracic kyphosis angle, coronal shoulder angle, and scapular elevation. RESULTS Significant differences were found amongst the groups in protraction of head (emphysema vs. chronic bronchitis, mean difference=7.63°, 95% confidence interval [CI]=2.10, 13.15°; emphysema vs. healthy, 7.91°, 95% CI=2.38, 13.43°), protraction of shoulder (emphysema vs. healthy, 13.69°, 95% CI=6.96, 20.43°; chronic bronchitis vs. healthy, 8.11°, 95% CI=1.38, 14.85°), thoracic kyphosis (emphysema vs. healthy, -11.59°, 95% CI=-17.26, -5.92°; chronic bronchitis vs. healthy, -6.75°, 95% CI=-12.41, -1.08°), coronal shoulder angle (emphysema vs. chronic bronchitis, 1.01°, 95% CI=.22, 1.80°; emphysema vs. healthy, 1.59°, 95% CI=.80, 2.38°) and scapular elevation (emphysema vs. chronic bronchitis, =.74cm, 95% CI=.34, 1.15cm; emphysema vs. healthy, .99cm, 95% CI=.59, 1.40cm). CONCLUSION People with emphysema show greater degree of postural malalignments in terms of head and shoulder protraction, thoracic kyphosis, symmetry of shoulders and scapular elevation than patients with chronic bronchitis and age-matched healthy individuals. These observations emphasize the importance of postural assessment in individuals with chronic obstructive pulmonary disease, particularly if they are emphysematous.
Collapse
Affiliation(s)
- Ansari Muhammed
- Centre for Physiotherapy and Rehabilitation Sciences, Jamia Millia Islamia, New Delhi, India
| | - Jamal Ali Moiz
- Centre for Physiotherapy and Rehabilitation Sciences, Jamia Millia Islamia, New Delhi, India.
| | - Deepika Singla
- Centre for Physiotherapy and Rehabilitation Sciences, Jamia Millia Islamia, New Delhi, India
| | - Mir Shad Ali
- Department of Pulmonary Rehabilitation, Metro Centre for Respiratory Diseases, Metro Hospital and Multispecialty Institute, Noida, Uttar Pradesh, India
| | - Deepak Talwar
- Department of Pulmonology Allergy Sleep and Critical Care Medicine, Metro Centre for Respiratory Diseases, Metro Hospital and Multispecialty Institute, Noida, Uttar Pradesh, India
| |
Collapse
|
25
|
Voisin MB, Leoni G, Woodfin A, Loumagne L, Patel NS, Di Paola R, Cuzzocrea S, Thiemermann C, Perretti M, Nourshargh S. Neutrophil elastase plays a non-redundant role in remodeling the venular basement membrane and neutrophil diapedesis post-ischemia/reperfusion injury. J Pathol 2019; 248:88-102. [PMID: 30632166 PMCID: PMC6850085 DOI: 10.1002/path.5234] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Revised: 11/09/2018] [Accepted: 12/23/2018] [Indexed: 12/20/2022]
Abstract
Ischemia/reperfusion (I/R) injury is a severe inflammatory insult associated with numerous pathologies, such as myocardial infarction, stroke and acute kidney injury. I/R injury is characterized by a rapid influx of activated neutrophils secreting toxic free radical species and degrading enzymes that can irreversibly damage the tissue, thus impairing organ functions. Significant efforts have been invested in identifying therapeutic targets to suppress neutrophil recruitment and activation post‐I/R injury. In this context, pharmacological targeting of neutrophil elastase (NE) has shown promising anti‐inflammatory efficacy in a number of experimental and clinical settings of I/R injury and is considered a plausible clinical strategy for organ care. However, the mechanisms of action of NE, and hence its inhibitors, in this process are not fully understood. Here we conducted a comprehensive analysis of the impact of NE genetic deletion on neutrophil infiltration in four murine models of I/R injury as induced in the heart, kidneys, intestine and cremaster muscle. In all models, neutrophil migration into ischemic regions was significantly suppressed in NE−/− mice as compared with wild‐type controls. Analysis of inflamed cremaster muscle and mesenteric microvessels by intravital and confocal microscopy revealed a selective entrapment of neutrophils within venular walls, most notably at the level of the venular basement membrane (BM) following NE deletion/pharmacological blockade. This effect was associated with the suppression of NE‐mediated remodeling of the low matrix protein expressing regions within the venular BM used by transmigrating neutrophils as exit portals. Furthermore, whilst NE deficiency led to reduced neutrophil activation and vascular leakage, levels of monocytes and prohealing M2 macrophages were reduced in tissues of NE−/− mice subjected to I/R. Collectively our results identify a vital and non‐redundant role for NE in supporting neutrophil breaching of the venular BM post‐I/R injury but also suggest a protective role for NE in promoting tissue repair. © 2019 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Mathieu-Benoit Voisin
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Giovanna Leoni
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK.,Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilian University (LMU), Munich, Germany
| | - Abigail Woodfin
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Laure Loumagne
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Nimesh Sa Patel
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Rosanna Di Paola
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Messina, Italy
| | - Salvatore Cuzzocrea
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Messina, Italy
| | - Christoph Thiemermann
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Mauro Perretti
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Sussan Nourshargh
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| |
Collapse
|
26
|
Cho J, Lee CH, Hwang SS, Kim KU, Lee SH, Park HY, Park SJ, Min KH, Oh YM, Yoo KH, Jung KS. Risk of acute exacerbations in chronic obstructive pulmonary disease associated with biomass smoke compared with tobacco smoke. BMC Pulm Med 2019; 19:68. [PMID: 30902075 PMCID: PMC6429752 DOI: 10.1186/s12890-019-0833-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Accepted: 03/13/2019] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Risk of exacerbations in chronic obstructive pulmonary disease (COPD) associated with biomass smoke has not been well addressed, although biomass smoke is similar in composition to tobacco smoke. METHODS To investigate whether the risk of exacerbations in COPD associated with biomass smoke differs from that in COPD associated with tobacco smoke, we recruited patients with COPD from two Korean multicenter prospective cohorts. In a multiple linear regression model, the standardized regression coefficient (β) of biomass smoke exposure ≥25 years was most similar to that (β') of tobacco smoke exposure ≥10 pack-years (β = - 0.13 and β' = - 0.14). We grouped patients with COPD into four categories based on the above cut-offs: Less Tobacco-Less Biomass, Less Tobacco-More Biomass, More Tobacco-Less Biomass, and More Tobacco-More Biomass. The main outcome was the incidence of moderate or severe exacerbations. RESULTS Among 1033 patients with COPD, 107 were included in Less Tobacco-Less Biomass (mean age: 67 years, men: 67%), 40 in Less Tobacco-More Biomass (mean age: 70 years, men: 35%), 631 in More Tobacco-Less Biomass (mean age: 68 years, men: 98%), and 255 in More Tobacco-More Biomass (mean age: 69 years, men: 97%). The incidence rates of exacerbations were not significantly different between Less Tobacco-More Biomass and More Tobacco-Less Biomass (adjusted incidence rate ratio, 1.03; 95% confidence interval, 0.56-1.89; P = 0.921). No interaction between sex and tobacco and biomass smoke was observed. When propensity score matching with available covariates including age and sex was applied, a similar result was observed. CONCLUSIONS Patients with COPD associated with biomass smoke and those with COPD associated with tobacco smoke had a similar risk of exacerbations. This suggests that patients with COPD associated with biomass smoke should be treated actively.
Collapse
Affiliation(s)
- Jaeyoung Cho
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea
| | - Chang-Hoon Lee
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea.
| | - Seung-Sik Hwang
- Department of Public Health Science, Graduate School of Public Health, Seoul National University, Seoul, Republic of Korea
| | - Ki Uk Kim
- Department of Internal Medicine, Pusan National University School of Medicine, Busan, Republic of Korea
| | - Sang Haak Lee
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, St. Paul's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Hye Yun Park
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Seoung Ju Park
- Department of Internal Medicine, Chonbuk National University Medical School, Jeonju, Republic of Korea
| | - Kyung Hoon Min
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Internal Medicine, Korea University Guro Hospital, Korea University College of Medicine, Seoul, Republic of Korea
| | - Yeon-Mok Oh
- Department of Pulmonary and Critical Care Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Kwang Ha Yoo
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Konkuk University Medical Center, Konkuk University School of Medicine, Seoul, Republic of Korea
| | - Ki-Suck Jung
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Internal Medicine, Hallym University Sacred Heart Hospital, Hallym University College of Medicine, Anyang, Republic of Korea
| |
Collapse
|
27
|
Chino K, Ganzberg S, Mendoza K. Office-Based Sedation/General Anesthesia for COPD Patients, Part II. Anesth Prog 2019; 66:44-51. [PMID: 30883229 PMCID: PMC6424168 DOI: 10.2344/anpr-66-02-05] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Accepted: 10/10/2018] [Indexed: 11/11/2022] Open
Abstract
The safe treatment of patients with chronic obstructive pulmonary disease (COPD) in dental office office-based settings can be quite complex without a current understanding of the etiology, course, severity, and current treatment modalities of the disease. The additional concerns of providing sedation and/or general anesthesia to patients with COPD in settings outside of a hospital demand thorough investigation of individual patient presentation and realistic development of planned treatment that patients suffering from this respiratory condition can tolerate. Along with other co-morbidities, such as advanced age and potential significant cardiovascular compromise, the dental practitioner providing sedation or general anesthesia must tailor any treatment plan to address multiple organ systems and mitigate risks of precipitating acute respiratory failure from inadequate pain and/or anxiety control. Part I of this article covered the epidemiology, etiology, and pathophysiology of COPD. Patient considerations in the preoperative period were also reviewed. Part II will cover which patients are acceptable for sedation/general anesthesia in the dental office-based setting as well as sedation/general anesthesia techniques that may be considered. Postoperative care will also be reviewed.
Collapse
Affiliation(s)
- Kristin Chino
- Private Practice, Anesthesia for Dentistry, Las Vegas, Nevada
| | - Steven Ganzberg
- Clinical Professor of Anesthesiology, UCLA School of Dentistry, Los Angeles, California
| | | |
Collapse
|
28
|
Burgess JK, Heijink IH. Chronic Lung Pathologies That Require Repair and Regeneration. STEM CELL-BASED THERAPY FOR LUNG DISEASE 2019. [PMCID: PMC7122311 DOI: 10.1007/978-3-030-29403-8_1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
Chronic lung diseases, such as chronic obstructive pulmonary disease (COPD) and pulmonary fibrosis, are a major cause of mortality worldwide. With the increasing incidence with ageing, the full impact of these diseases is yet to be realised. For most chronic lung diseases there are limited treatments options, with the existing approaches mainly addressing symptom relief. Little progress has been made, in recent years, in the development of new therapeutic strategies for managing these burdensome pathologies. There is an urgent need to increase our understanding of the mechanisms underlying these diseases. Endogenous progenitor cells (stem cells) have been recognised in many organs, including the lungs where they are suggested to maintain a population of cells that are able to facilitate the endogenous repair processes. Emerging knowledge of how these repair processes are disrupted in chronic lung diseases and the potential to capitalise upon the regenerative capacity of stem cell populations raise the hopes of the field worldwide for innovative treatment approaches for these devastating diseases in the future. This chapter outlines the series of diseases that may benefit from these emerging new therapeutic outlooks.
Collapse
Affiliation(s)
- Janette K. Burgess
- The University of Groningen, University Medical Center Groningen, Department of Pathology and Medical Biology, Groningen, The Netherlands
| | - Irene H. Heijink
- The University of Groningen, University Medical Center Groningen, Department of Pathology and Medical Biology, Groningen, The Netherlands
| |
Collapse
|
29
|
Tuder RM. Bringing Light to Chronic Obstructive Pulmonary Disease Pathogenesis and Resilience. Ann Am Thorac Soc 2018; 15:S227-S233. [PMID: 30759011 PMCID: PMC6944393 DOI: 10.1513/annalsats.201808-583mg] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Accepted: 10/03/2018] [Indexed: 12/15/2022] Open
Abstract
The pathogenesis of chronic obstructive pulmonary disease remains elusive; investigators in the field have struggled to decipher the cellular and molecular processes underlying chronic bronchitis and emphysema. Studies in the past 20 years have underscored that the tissue destruction, notably in emphysema, involves a multitude of injurious stresses, with progressive engagement of endogenous destructive processes triggered by decades of exposure to cigarette smoke and/or pollutants. These lead to an aged lung, with evidence of macromolecular damage that is unlikely to repair. Here we discuss these key pathogenetic elements in the context of organismal evolution as this concept may best capture the challenges facing chronic obstructive pulmonary disease.
Collapse
Affiliation(s)
- Rubin M Tuder
- Program in Translational Lung Research and Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado
| |
Collapse
|
30
|
Chino K, Ganzberg S, Mendoza K. Office-Based Sedation/General Anesthesia for COPD Patients, Part I. Anesth Prog 2018; 65:261-268. [PMID: 30715953 PMCID: PMC6318726 DOI: 10.2344/anpr-65-04-12] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Accepted: 10/10/2018] [Indexed: 11/11/2022] Open
Abstract
The safe treatment of patients with chronic obstructive pulmonary disease (COPD) in dental office-based settings can be quite complex without a current understanding of the etiology, course, severity, and treatment modalities of the disease. The additional concerns of providing sedation and/or general anesthesia to patients with COPD in settings outside of a hospital demand thorough investigation of individual patient presentation and realistic development of planned treatment that patients suffering from this respiratory condition can tolerate. Along with other comorbidities, such as advanced age and potential significant cardiovascular compromise, the dental practitioner providing sedation or general anesthesia must tailor any treatment plan to address multiple organ systems and mitigate risks of precipitating acute respiratory failure from inadequate pain and/or anxiety control. Part I of this article will cover the epidemiology, etiology, and pathophysiology of COPD. Patient evaluation in the preoperative period will also be reviewed. Part II will cover which patients are acceptable for sedation/general anesthesia in the dental office-based setting as well as sedation/general anesthesia techniques that may be considered.
Collapse
Affiliation(s)
- Kristin Chino
- Private Practice, Anesthesia for Dentistry, Las Vegas, Nevada
| | - Steven Ganzberg
- Clinical Professor of Anesthesiology, UCLA School of Dentistry, Los Angeles, California
| | | |
Collapse
|
31
|
Nevárez-Sida A, Castro-Bucio AJ, García-Contreras F, Cisneros-González N. Costos medicos directos en pacientes con enfermedad pulmonar obstructiva Crónica en Mexico. Value Health Reg Issues 2017; 14:9-14. [PMID: 29254548 DOI: 10.1016/j.vhri.2017.03.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2016] [Revised: 09/05/2016] [Accepted: 03/01/2017] [Indexed: 11/26/2022]
Abstract
INTRODUCTION chronic obstructive pulmonary disease (COPD) is a progressive, incurable and potentially mortal. COPD generates a high burden of illness and decreased quality of life in patients. The aim of this study was to determine the direct medical cost of COPD and the primary variables associated. METHODOLOGY We conducted a multicenter clinical study, based in a retrospective cohort as base of a partial economic evaluation in patients diagnosed with moderate to severe COPD. It was considered an institutional point of view to determine medical costs, with an annual time horizon. For analysis of associations between explanatory and end point variables, a generalized lineal regression model was developed. RESULTS We analyzed data from 283 patients, Fifty-nine percent were women, the average age was 72 years ± 11, Sixty-five percent of patients had a history of smoking and 57.6 % were exposed to wood smoke. The annual direct medical costs (MXN 2016) was 20,754 and 41,887 for patients with moderate and severe COPD, respectively, this difference is mainly due to the use of oxygen as well as longer hospital stay (12.9 vs. 24.7 days) of patients with severe COPD. CONCLUSIONS Although the severity level is associated with greater health care costs, the quality of life of the patients should be considered carefully because it is inversely associated with the cost of care for patients with COPD.
Collapse
Affiliation(s)
- Armando Nevárez-Sida
- Unidad de Investigación en Epidemiología y Servicios de Salud Área de Envejecimiento. Coordinación de Investigación en Salud, IMSS. México D.F.
| | | | | | | |
Collapse
|
32
|
Nordgren TM, Heires AJ, Bailey KL, Katafiasz DM, Toews ML, Wichman CS, Romberger DJ. Docosahexaenoic acid enhances amphiregulin-mediated bronchial epithelial cell repair processes following organic dust exposure. Am J Physiol Lung Cell Mol Physiol 2017; 314:L421-L431. [PMID: 29097425 DOI: 10.1152/ajplung.00273.2017] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Injurious dust exposures in the agricultural workplace involve the release of inflammatory mediators and activation of epidermal growth factor receptor (EGFR) in the respiratory epithelium. Amphiregulin (AREG), an EGFR ligand, mediates tissue repair and wound healing in the lung epithelium. Omega-3 fatty acids such as docosahexaenoic acid (DHA) are also known modulators of repair and resolution of inflammatory injury. This study investigated how AREG, DHA, and EGFR modulate lung repair processes following dust-induced injury. Primary human bronchial epithelial (BEC) and BEAS-2B cells were treated with an aqueous extract of swine confinement facility dust (DE) in the presence of DHA and AREG or EGFR inhibitors. Mice were exposed to DE intranasally with or without EGFR inhibition and DHA. Using a decellularized lung scaffolding tissue repair model, BEC recolonization of human lung scaffolds was analyzed in the context of DE, DHA, and AREG treatments. Through these investigations, we identified an important role for AREG in mediating BEC repair processes. DE-induced AREG release from BEC, and DHA treatment following DE exposure, enhanced this release. Both DHA and AREG also enhanced BEC repair capacities and rescued DE-induced recellularization deficits. In vivo, DHA treatment enhanced AREG production following DE exposure, whereas EGFR inhibitor-treated mice exhibited reduced AREG in their lung homogenates. These data indicate a role for AREG in the process of tissue repair after inflammatory lung injury caused by environmental dust exposure and implicate a role for DHA in regulating AREG-mediated repair signaling in BEC.
Collapse
Affiliation(s)
- Tara M Nordgren
- Pulmonary, Critical Care, Sleep and Allergy Division, Department of Internal Medicine, University of Nebraska Medical Center , Omaha, Nebraska.,Division of Biomedical Sciences, School of Medicine, University of California Riverside , Riverside, California
| | - Art J Heires
- Pulmonary, Critical Care, Sleep and Allergy Division, Department of Internal Medicine, University of Nebraska Medical Center , Omaha, Nebraska
| | - Kristina L Bailey
- Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, Nebraska.,Pulmonary, Critical Care, Sleep and Allergy Division, Department of Internal Medicine, University of Nebraska Medical Center , Omaha, Nebraska
| | - Dawn M Katafiasz
- Pulmonary, Critical Care, Sleep and Allergy Division, Department of Internal Medicine, University of Nebraska Medical Center , Omaha, Nebraska
| | - Myron L Toews
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center , Omaha, Nebraska
| | - Christopher S Wichman
- Department of Biostatistics, University of Nebraska Medical Center , Omaha, Nebraska
| | - Debra J Romberger
- Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, Nebraska.,Pulmonary, Critical Care, Sleep and Allergy Division, Department of Internal Medicine, University of Nebraska Medical Center , Omaha, Nebraska
| |
Collapse
|
33
|
Ambrosino P, Lupoli R, Iervolino S, De Felice A, Pappone N, Storino A, Di Minno MND. Clinical assessment of endothelial function in patients with chronic obstructive pulmonary disease: a systematic review with meta-analysis. Intern Emerg Med 2017; 12:877-885. [PMID: 28593450 DOI: 10.1007/s11739-017-1690-0] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Accepted: 05/24/2017] [Indexed: 12/21/2022]
Abstract
Patients with chronic obstructive pulmonary disease (COPD) have an increased cardiovascular morbidity and mortality. Flow-mediated (FMD) and nitrate-mediated dilatation (NMD) are considered non-invasive methods to assess endothelial function and surrogate markers of subclinical atherosclerosis. We performed a systematic review with meta-analysis and meta-regression to evaluate the impact of COPD on FMD and NMD. Studies were systematically searched in the PubMed, Web of Science, Scopus and EMBASE databases. The random-effect method was used to take into account the variability among included studies. A total of eight studies were included in the final analysis, eight with data on FMD (334 COPD patients) and two on NMD (104 COPD patients). Compared to controls, COPD patients show a significantly lower FMD (MD -3.15%; 95% CI -4.89, -1.40; P < 0.001) and NMD (MD -3.53%; 95% CI -7.04, -0.02; P = 0.049). Sensitivity analyses substantially confirms the results. Meta-regression models show that a more severe degree of airway obstruction is associated with a more severe FMD impairment in COPD patients than in controls. Regression analyses confirm that the association between COPD and endothelial dysfunction is independent of baseline smoking status and most traditional cardiovascular risk factors. In conclusion, COPD is significantly and independently associated with endothelial dysfunction. These findings may be useful to plan adequate cardiovascular prevention strategies in this clinical setting, with particular regard to patients with a more severe disease.
Collapse
Affiliation(s)
- Pasquale Ambrosino
- Department of Clinical Medicine and Surgery, Federico II University, Naples, Italy
- ICS Maugeri SpA SB, Scientific Institute of Telese Terme, IRCCS, Telese Terme, BN, Italy
| | - Roberta Lupoli
- Department of Clinical Medicine and Surgery, Federico II University, Naples, Italy
| | - Salvatore Iervolino
- ICS Maugeri SpA SB, Scientific Institute of Telese Terme, IRCCS, Telese Terme, BN, Italy
| | - Alberto De Felice
- ICS Maugeri SpA SB, Scientific Institute of Telese Terme, IRCCS, Telese Terme, BN, Italy
| | - Nicola Pappone
- ICS Maugeri SpA SB, Scientific Institute of Telese Terme, IRCCS, Telese Terme, BN, Italy
| | - Antonio Storino
- ICS Maugeri SpA SB, Scientific Institute of Telese Terme, IRCCS, Telese Terme, BN, Italy
- Department of Public Health, Federico II University, Naples, Italy
| | - Matteo Nicola Dario Di Minno
- Division of Cardiology, Department of Advanced Biomedical Sciences, Federico II University, Via S. Pansini 5, 80131, Naples, Italy.
| |
Collapse
|
34
|
Ascher K, Elliot SJ, Rubio GA, Glassberg MK. Lung Diseases of the Elderly: Cellular Mechanisms. Clin Geriatr Med 2017; 33:473-490. [PMID: 28991645 DOI: 10.1016/j.cger.2017.07.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Natural lung aging is characterized by molecular and cellular changes in multiple lung cell populations. These changes include shorter telomeres, increased expression of cellular senescence markers, increased DNA damage, oxidative stress, apoptosis, and stem cell exhaustion. Aging, combined with the loss of protective repair processes, correlates with the development and incidence of chronic respiratory diseases, including idiopathic pulmonary fibrosis and chronic obstructive pulmonary disease. Ultimately, it is the interplay of age-related changes in biology and the subsequent responses to environmental exposures that largely define the physiology and clinical course of the aging lung.
Collapse
Affiliation(s)
- Kori Ascher
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, University of Miami Leonard M. Miller School of Medicine, 1600 Northwest 10th Avenue RMSB 7056 (D-60), Miami, FL 33136, USA
| | - Sharon J Elliot
- DeWitt Daughtry Family Department of Surgery, University of Miami Leonard M. Miller School of Medicine, 1600 NW 10th Avenue, Miami, FL 33136, USA
| | - Gustavo A Rubio
- DeWitt Daughtry Family Department of Surgery, University of Miami Leonard M. Miller School of Medicine, 1600 NW 10th Avenue, Miami, FL 33136, USA
| | - Marilyn K Glassberg
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, University of Miami Leonard M. Miller School of Medicine, 1600 Northwest 10th Avenue RMSB 7056 (D-60), Miami, FL 33136, USA; DeWitt Daughtry Family Department of Surgery, University of Miami Leonard M. Miller School of Medicine, 1600 NW 10th Avenue, Miami, FL 33136, USA; Division of Pediatric Pulmonology, Department of Pediatrics, University of Miami Leonard M. Miller School of Medicine, 1600 NW 10th Avenue, Miami, FL 33136, USA.
| |
Collapse
|
35
|
Radder JE, Zhang Y, Gregory AD, Yu S, Kelly NJ, Leader JK, Kaminski N, Sciurba FC, Shapiro SD. Extreme Trait Whole-Genome Sequencing Identifies PTPRO as a Novel Candidate Gene in Emphysema with Severe Airflow Obstruction. Am J Respir Crit Care Med 2017; 196:159-171. [PMID: 28199135 DOI: 10.1164/rccm.201606-1147oc] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
RATIONALE Genetic association studies in chronic obstructive pulmonary disease have primarily tested for association with common variants, the results of which explain only a portion of disease heritability. Because rare variation is also likely to contribute to susceptibility, we used whole-genome sequencing of subjects with clinically extreme phenotypes to identify genomic regions enriched for rare variation contributing to chronic obstructive pulmonary disease susceptibility. OBJECTIVES To identify regions of rare genetic variation contributing to emphysema with severe airflow obstruction. METHODS We identified heavy smokers that were resistant (n = 65) or susceptible (n = 64) to emphysema with severe airflow obstruction in the Pittsburgh Specialized Center of Clinically Oriented Research cohort. We filtered whole-genome sequencing results to include only rare variants and conducted single variant tests, region-based tests across the genome, gene-based tests, and exome-wide tests. MEASUREMENTS AND MAIN RESULTS We identified several suggestive associations with emphysema with severe airflow obstruction, including a suggestive association of all rare variation in a region within the gene ZNF816 (19q13.41; P = 4.5 × 10-6), and a suggestive association of nonsynonymous coding rare variation in the gene PTPRO (P = 4.0 × 10-5). Association of rs61754411, a rare nonsynonymous variant in PTPRO, with emphysema and obstruction was demonstrated in all non-Hispanic white individuals in the Pittsburgh Specialized Center of Clinically Oriented Research cohort. We found that cells containing this variant have decreased signaling in cellular pathways necessary for survival and proliferation. CONCLUSIONS PTPRO is a novel candidate gene in emphysema with severe airflow obstruction, and rs61754411 is a previously unreported rare variant contributing to emphysema susceptibility. Other suggestive candidate genes, such as ZNF816, are of interest for future studies.
Collapse
Affiliation(s)
- Josiah E Radder
- 1 Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, and
| | - Yingze Zhang
- 1 Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, and
| | - Alyssa D Gregory
- 1 Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, and
| | - Shibing Yu
- 1 Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, and
| | - Neil J Kelly
- 1 Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, and
| | - Joseph K Leader
- 2 Department of Radiology, University of Pittsburgh, Pittsburgh, Pennsylvania; and
| | - Naftali Kaminski
- 3 Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, Yale University, New Haven, Connecticut
| | - Frank C Sciurba
- 1 Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, and
| | - Steven D Shapiro
- 1 Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, and
| |
Collapse
|
36
|
Nam HS, Izumchenko E, Dasgupta S, Hoque MO. Mitochondria in chronic obstructive pulmonary disease and lung cancer: where are we now? Biomark Med 2017; 11:475-489. [PMID: 28598223 DOI: 10.2217/bmm-2016-0373] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Recent advances in mitochondrial biogenesis have provided the emerging recognition that mitochondria do much more than 'simply providing energy for cellular function'. Currently, a constantly improving understanding of the mitochondrial structure and function has been providing valuable insights into the contribution of defects in mitochondrial metabolism to various human diseases, including chronic obstructive pulmonary disease and lung cancer. The growing interest in mitochondria research led to development of new biomedical fields in the two main smoking-related lung diseases. However, there is considerable paucity in our understanding of mechanisms by which mitochondrial dynamics regulate lung diseases. In this review, we will discuss our current knowledge on the role of mitochondrial dysfunction in the pathogenesis of chronic obstructive pulmonary disease and non-small-cell lung cancer.
Collapse
Affiliation(s)
- Hae-Seong Nam
- Department of Otolaryngology & Head & Neck Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA.,Division of Pulmonology, Department of Internal Medicine, Inha University Hospital, Inha University School of Medicine, Incheon 22332, South Korea
| | - Evgeny Izumchenko
- Department of Otolaryngology & Head & Neck Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Santanu Dasgupta
- Department of Cellular & Molecular Biology, The University of Texas Health Science Center at Tyler, Tyler, TX 75708, USA
| | - Mohammad O Hoque
- Department of Otolaryngology & Head & Neck Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA.,Department of Urology, The Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA.,Department of Oncology, The Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| |
Collapse
|
37
|
Jiang Z, Knudsen NH, Wang G, Qiu W, Naing ZZC, Bai Y, Ai X, Lee CH, Zhou X. Genetic Control of Fatty Acid β-Oxidation in Chronic Obstructive Pulmonary Disease. Am J Respir Cell Mol Biol 2017; 56:738-748. [PMID: 28199134 PMCID: PMC5516290 DOI: 10.1165/rcmb.2016-0282oc] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Accepted: 01/31/2017] [Indexed: 12/17/2022] Open
Abstract
Bioenergetics homeostasis is important for cells to sustain normal functions and defend against injury. The genetic controls of bioenergetics homeostasis, especially lipid metabolism, remain poorly understood in chronic obstructive pulmonary disease (COPD), the third leading cause of death in the world. Additionally, the biological function of most of the susceptibility genes identified from genome-wide association studies (GWASs) in COPD remains unclear. Here, we aimed to address (1) how fatty acid oxidation (FAO), specifically β-oxidation, a key lipid metabolism pathway that provides energy to cells, contributes to cigarette smoke (CS)-induced COPD; and (2) whether-and if so, how-FAM13A (family with sequence similarity 13 member A), a well-replicated COPD GWAS gene, modulates the FAO pathway. We demonstrated that CS induced expression of carnitine palmitoyltransferase 1A (CPT1A), a key mitochondrial enzyme for the FAO pathway, thereby enhancing FAO. Pharmacological inhibition of FAO by etomoxir blunted CS-induced reactive oxygen species accumulation and cell death in lung epithelial cells. FAM13A promoted FAO, possibly by interacting with and activating sirutin 1, and increasing expression of CPT1A. Furthermore, CS-induced cell death was reduced in lungs from Fam13a-/- mice. Our results suggest that FAM13A, the COPD GWAS gene, shapes the cellular metabolic response to CS exposure by promoting the FAO pathway, which may contribute to COPD development.
Collapse
Affiliation(s)
| | - Nelson H. Knudsen
- Departments of Genetics and Complex Diseases, and
- Nutrition, Division of Biological Sciences, Harvard T. H. Chan School of Public Health, Boston, Massachusetts, and
| | - Gang Wang
- Department of Cardiology, Boston Children’s Hospital, Boston, Massachusetts
| | | | | | - Yan Bai
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School
| | - Xingbin Ai
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School
| | - Chih-Hao Lee
- Departments of Genetics and Complex Diseases, and
- Nutrition, Division of Biological Sciences, Harvard T. H. Chan School of Public Health, Boston, Massachusetts, and
| | - Xiaobo Zhou
- Channing Division of Network Medicine and
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School
| |
Collapse
|
38
|
Jiang Y, Wang X, Hu D. Mitochondrial alterations during oxidative stress in chronic obstructive pulmonary disease. Int J Chron Obstruct Pulmon Dis 2017; 12:1153-1162. [PMID: 28458526 PMCID: PMC5402882 DOI: 10.2147/copd.s130168] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The high incidence of chronic obstructive pulmonary disease (COPD), one of the most prevalent diseases worldwide, has attracted growing attention. Cigarette smoking is considered a major contributory factor in the pathogenesis and progression of COPD due to the tremendous oxidative burden that it causes, which induces an oxidant/antioxidant imbalance. Excessive oxidation induced by the excessive generation of mitochondrial reactive oxygen species disturbs the antioxidant systems and plays an important role in triggering and promoting chronic inflammation of airways. Given that mitochondria is one of the main sites of reactive oxygen species production by the oxidative phosphorylation process, oxidative stress may affect mitochondrial function by changing its structure and morphology and by affecting a series of mitochondrial proteins. In particular, PTEN-induced putative kinase 1/Parkin and p62 play critical roles in mitophagy. During the process, the Akt ubiquitin E3 ligase is an important mediator associated with cigarette smoke exposure-induced pulmonary endothelial cell death and dysfunction. Thus, understanding the underlying mechanisms of the signaling pathway may provide important information regarding the therapeutic treatment of COPD by application of alternative PTEN-induced putative kinase 1 targets or ubiquitin E3 ligase.
Collapse
Affiliation(s)
- Ying Jiang
- Department of Clinical Pharmacology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Xiaoqin Wang
- Department of Clinical Pharmacology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Daode Hu
- Department of Clinical Pharmacology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| |
Collapse
|
39
|
Zhao XK, Che P, Cheng ML, Zhang Q, Mu M, Li H, Luo Y, Liang YD, Luo XH, Gao CQ, Jackson PL, Wells JM, Zhou Y, Hu M, Cai G, Thannickal VJ, Steele C, Blalock JE, Han X, Chen CY, Ding Q. Tristetraprolin Down-Regulation Contributes to Persistent TNF-Alpha Expression Induced by Cigarette Smoke Extract through a Post-Transcriptional Mechanism. PLoS One 2016; 11:e0167451. [PMID: 27911957 PMCID: PMC5135108 DOI: 10.1371/journal.pone.0167451] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2016] [Accepted: 11/14/2016] [Indexed: 12/19/2022] Open
Abstract
Rationale Tumor necrosis factor-alpha (TNF-α) is a potent pro-inflammatory mediator and its expression is up-regulated in chronic obstructive pulmonary disease (COPD). Tristetraprolin (TTP) is implicated in regulation of TNF-α expression; however, whether TTP is involved in cigarette smoke-induced TNF-α expression has not been determined. Methods TTP expression was examined by western blot analysis in murine alveolar macrophages and alveolar epithelial cells challenged without or with cigarette smoke extract (CSE). TNF-α mRNA stability, and the decay of TNF-α mRNA, were determined by real-time quantitative RT-PCR. TNF-α protein levels were examined at the same time in these cells. To identify the molecular mechanism involved, a construct expressing the human beta-globin reporter mRNA containing the TNF-α 3’-untranslated region was generated to characterize the TTP targeted site within TNF-α mRNA. Results CSE induced TTP down-regulation in alveolar macrophages and alveolar epithelial cells. Reduced TTP expression resulted in significantly increased TNF-α mRNA stability. Importantly, increased TNF-α mRNA stability due to impaired TTP function resulted in significantly increased TNF-α levels in these cells. Forced TTP expression abrogated the increased TNF-α mRNA stability and expression induced by CSE. By using the globin reporter construct containing TNF-α mRNA 3’-untranslated region, the data indicate that TTP directly targets the adenine- and uridine-rich region (ARE) of TNF-α mRNA and negatively regulates TNF-α expression at the post-transcriptional level. Conclusion The data demonstrate that cigarette smoke exposure reduces TTP expression and impairs TTP function, resulting in significantly increased TNF-α mRNA stability and excessive TNF-α expression in alveolar macrophages and epithelial cells. The data suggest that TTP is a novel post-transcriptional regulator and limits excessive TNF-α expression and inflammatory response induced by cigarette smoke.
Collapse
Affiliation(s)
- Xue-Ke Zhao
- Department of Infectious Diseases, The Hospital Affiliated to Guizhou Medical University, Guiyang, Guizhou, China
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Pulin Che
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Ming-Liang Cheng
- Department of Infectious Diseases, The Hospital Affiliated to Guizhou Medical University, Guiyang, Guizhou, China
- * E-mail: (MLC); (QD)
| | - Quan Zhang
- Department of Infectious Diseases, The Hospital Affiliated to Guizhou Medical University, Guiyang, Guizhou, China
| | - Mao Mu
- Department of Infectious Diseases, The Hospital Affiliated to Guizhou Medical University, Guiyang, Guizhou, China
| | - Hong Li
- Department of Infectious Diseases, The Hospital Affiliated to Guizhou Medical University, Guiyang, Guizhou, China
| | - Yuan Luo
- Department of Oral Surgery, Shanghai Stomatology Hospital, Fudan University, Shanghai, China
| | - Yue-Dong Liang
- Department of Infectious Diseases, Public Health Center of Guiyang, Guiyang, Guizhou, China
| | - Xin-Hua Luo
- Department of Infectious Diseases, People's Hospital of Guizhou Province, Guiyang, Guizhou, China
| | - Chang-Qing Gao
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Patricia L. Jackson
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - J. Michael Wells
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Yong Zhou
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Meng Hu
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Guoqiang Cai
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Victor J. Thannickal
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Chad Steele
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - J. Edwin Blalock
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Xiaosi Han
- Neurology, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Ching-Yi Chen
- Department of Biochemistry, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Qiang Ding
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
- * E-mail: (MLC); (QD)
| |
Collapse
|
40
|
Deeb RS, Hajjar DP. Repair Mechanisms in Oxidant-Driven Chronic Inflammatory Disease. THE AMERICAN JOURNAL OF PATHOLOGY 2016; 186:1736-1749. [PMID: 27171899 DOI: 10.1016/j.ajpath.2016.03.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/30/2015] [Revised: 02/24/2016] [Accepted: 03/04/2016] [Indexed: 12/19/2022]
Abstract
The interplay that governs chronic diseases through pathways specifically associated with chronic inflammation remains undefined. Many metabolic events have been identified during the injury and repair process. Nonetheless, the cellular events that control the pathogenesis of inflammation-induced disease have not been fully characterized. We and others reason that chronic inflammatory diseases associated with a cascade of complex network mediators, such as nitric oxide, arachidonic acid metabolites, cytokines, and reactive oxygen species, play a significant role in the governance of alterations in homeostasis, oxidative stress, and thromboatherosclerosis. In this context, we discuss lipid mediators associated with the maintenance of health, including the specialized proresolving mediators that help drive cellular repair. Emphasis is placed on the pathophysiology of chronic metabolic insults involving both the airways and the cardiovascular system during oxidant-driven inflammatory disease. In this review, we highlight new pathways of inquiry that show promise for the identification of those metabolic targets that can improve therapy for chronic inflammation.
Collapse
Affiliation(s)
- Ruba S Deeb
- Department of Bioengineering, University of Bridgeport, Bridgeport, Connecticut.
| | - David P Hajjar
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, Cornell University, New York, New York.
| |
Collapse
|
41
|
Tiwari N, Marudamuthu AS, Tsukasaki Y, Ikebe M, Fu J, Shetty S. p53- and PAI-1-mediated induction of C-X-C chemokines and CXCR2: importance in pulmonary inflammation due to cigarette smoke exposure. Am J Physiol Lung Cell Mol Physiol 2016; 310:L496-506. [PMID: 26747783 PMCID: PMC4888555 DOI: 10.1152/ajplung.00290.2015] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Accepted: 01/02/2016] [Indexed: 11/22/2022] Open
Abstract
We previously demonstrated that tumor suppressor protein p53 augments plasminogen activator inhibitor-1 (PAI-1) expression in alveolar epithelial cells (AECs) during chronic cigarette smoke (CS) exposure-induced lung injury. Chronic lung inflammation with elevated p53 and PAI-1 expression in AECs and increased susceptibility to and exacerbation of respiratory infections are all associated with chronic obstructive pulmonary disease (COPD). We recently demonstrated that preventing p53 from binding to the endogenous PAI-1 mRNA in AECs by either suppressing p53 expression or blockading p53 interactions with the PAI-1 mRNA mitigates apoptosis and lung injury. Within this context, we now show increased expression of the C-X-C chemokines (CXCL1 and CXCL2) and their receptor CXCR2, and the intercellular cellular adhesion molecule-1 (ICAM-1), in the lung tissues of patients with COPD. We also found a similar increase in lung tissues and AECs from wild-type (WT) mice exposed to passive CS for 20 wk and in primary AECs treated with CS extract in vitro. Interestingly, passive CS exposure of mice lacking either p53 or PAI-1 expression resisted an increase in CXCL1, CXCL2, CXCR2, and ICAM-1. Furthermore, inhibition of p53-mediated induction of PAI-1 expression by treatment of WT mice exposed to passive CS with caveolin-1 scaffolding domain peptide reduced CXCL1, CXCL2, and CXCR2 levels and lung inflammation. Our study reveals that p53-mediated induction of PAI-1 expression due to chronic CS exposure exacerbates lung inflammation through elaboration of CXCL1, CXCL2, and CXCR2. We further provide evidence that targeting this pathway mitigates lung injury associated with chronic CS exposure.
Collapse
Affiliation(s)
- Nivedita Tiwari
- The Texas Lung Injury Institute, The University of Texas Health Science Center at Tyler, Tyler, Texas; and
| | - Amarnath S Marudamuthu
- The Texas Lung Injury Institute, The University of Texas Health Science Center at Tyler, Tyler, Texas; and
| | - Yoshikazu Tsukasaki
- The Texas Lung Injury Institute, The University of Texas Health Science Center at Tyler, Tyler, Texas; and
| | - Mitsuo Ikebe
- The Texas Lung Injury Institute, The University of Texas Health Science Center at Tyler, Tyler, Texas; and
| | - Jian Fu
- Center for Research on Environmental Disease and Toxicology, College of Medicine, University of Kentucky, Lexington, Kentucky
| | - Sreerama Shetty
- The Texas Lung Injury Institute, The University of Texas Health Science Center at Tyler, Tyler, Texas; and
| |
Collapse
|
42
|
Yeager RP, Kushman M, Chemerynski S, Weil R, Fu X, White M, Callahan-Lyon P, Rosenfeldt H. Proposed Mode of Action for Acrolein Respiratory Toxicity Associated with Inhaled Tobacco Smoke. Toxicol Sci 2016; 151:347-64. [DOI: 10.1093/toxsci/kfw051] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
|
43
|
Margelidon-Cozzolino V, Chbini K, Freymond N, Devouassoux G, Belaaouaj A, Pacheco Y. [COPD: An early disease]. REVUE DE PNEUMOLOGIE CLINIQUE 2016; 72:49-60. [PMID: 26657351 PMCID: PMC7126852 DOI: 10.1016/j.pneumo.2015.08.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/02/2014] [Accepted: 08/16/2015] [Indexed: 05/04/2023]
Abstract
This general review deals with the mechanisms which underlie the genetic factors in COPD. Many cellular and biochemical mechanisms occur in bronchial inflammation. We present the experimental models of COPD, insisting on the importance of oxydative stress, and on recent knowledge about the lung microbiome. Starting from this pathophysiology basis, we show how various genetic targets are able to interfere with the disease model. Thanks to these genetic targets, new markers in exhaled breath condensates and new drug targets are rising.
Collapse
Affiliation(s)
- V Margelidon-Cozzolino
- Service de pneumologie A, centre hospitalier de Lyon Sud, hospices civils de Lyon, faculté de médecine, université Claude-Bernard Lyon 1, 69310 Pierre-Bénite, France.
| | - K Chbini
- Service de cardiologie, CHU Mohammed VI, faculté de médecine et de pharmacie, université Cadi Ayyad, Marrakech, Maroc
| | - N Freymond
- Service de pneumologie A, centre hospitalier de Lyon Sud, hospices civils de Lyon, 69310 Pierre-Bénite, France
| | - G Devouassoux
- Service de pneumologie, hôpital de la Croix Rousse, hospices civils de Lyon, faculté de médecine Lyon Sud, université Claude-Bernard Lyon 1, 69005 Lyon, France
| | - A Belaaouaj
- Inserm 1111, faculté de médecine Lyon Sud, chemin du Grand-Revoyet, 69310 Pierre-Bénite, France
| | - Y Pacheco
- Service de pneumologie A, centre hospitalier de Lyon Sud, hospices civils de Lyon, faculté de médecine Lyon Sud, université Claude-Bernard Lyon 1, 69310 Pierre-Bénite, France
| |
Collapse
|
44
|
Kenche H, Ye ZW, Vedagiri K, Richards DM, Gao XH, Tew KD, Townsend DM, Blumental-Perry A. Adverse Outcomes Associated with Cigarette Smoke Radicals Related to Damage to Protein-disulfide Isomerase. J Biol Chem 2016; 291:4763-78. [PMID: 26728460 DOI: 10.1074/jbc.m115.712331] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Indexed: 12/19/2022] Open
Abstract
Identification of factors contributing to the development of chronic obstructive pulmonary disease (COPD) is crucial for developing new treatments. An increase in the levels of protein-disulfide isomerase (PDI), a multifaceted endoplasmic reticulum resident chaperone, has been demonstrated in human smokers, presumably as a protective adaptation to cigarette smoke (CS) exposure. We found a similar increase in the levels of PDI in the murine model of COPD. We also found abnormally high levels (4-6 times) of oxidized and sulfenilated forms of PDI in the lungs of murine smokers compared with non-smokers. PDI oxidation progressively increases with age. We begin to delineate the possible role of an increased ratio of oxidized PDI in the age-related onset of COPD by investigating the impact of exposure to CS radicals, such as acrolein (AC), hydroxyquinones (HQ), peroxynitrites (PN), and hydrogen peroxide, on their ability to induce unfolded protein response (UPR) and their effects on the structure and function of PDIs. Exposure to AC, HQ, PN, and CS resulted in cysteine and tyrosine nitrosylation leading to an altered three-dimensional structure of the PDI due to a decrease in helical content and formation of a more random coil structure, resulting in protein unfolding, inhibition of PDI reductase and isomerase activity in vitro and in vivo, and subsequent induction of endoplasmic reticulum stress response. Addition of glutathione prevented the induction of UPR, and AC and HQ induced structural changes in PDI. Exposure to PN and glutathione resulted in conjugation of PDI possibly at active site tyrosine residues. The findings presented here propose a new role of PDI in the pathogenesis of COPD and its age-dependent onset.
Collapse
Affiliation(s)
- Harshavardhan Kenche
- From the Anderson Cancer Institute, Memorial Health University Medical Center, Savannah, Georgia 31404
| | - Zhi-Wei Ye
- the College of Pharmacy, Medical University of South Carolina, Charleston, South Carolina 29425
| | - Kokilavani Vedagiri
- From the Anderson Cancer Institute, Memorial Health University Medical Center, Savannah, Georgia 31404
| | - Dylan M Richards
- From the Anderson Cancer Institute, Memorial Health University Medical Center, Savannah, Georgia 31404
| | - Xing-Huang Gao
- Genetics, Case Western Reserve University, Cleveland, Ohio 44106, and
| | - Kenneth D Tew
- the College of Pharmacy, Medical University of South Carolina, Charleston, South Carolina 29425
| | - Danyelle M Townsend
- the College of Pharmacy, Medical University of South Carolina, Charleston, South Carolina 29425
| | - Anna Blumental-Perry
- From the Anderson Cancer Institute, Memorial Health University Medical Center, Savannah, Georgia 31404, the Department of Biomedical Sciences, Mercer University School of Medicine, Savannah, Georgia 31404, the Departments of Surgery and
| |
Collapse
|
45
|
Oliva LV, Almeida-Reis R, Theodoro-Junior O, Oliveira BM, Leick EA, Prado CM, Brito MV, Correia MTDS, Paiva PM, Martins MA, Oliva MLV, Tibério IF. A plant proteinase inhibitor from Crataeva tapia (CrataBL) attenuates elastase-induced pulmonary inflammatory, remodeling, and mechanical alterations in mice. Process Biochem 2015. [DOI: 10.1016/j.procbio.2015.06.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
46
|
Ko HK, Lee HF, Lin AH, Liu MH, Liu CI, Lee TS, Kou YR. Regulation of Cigarette Smoke Induction of IL-8 in Macrophages by AMP-activated Protein Kinase Signaling. J Cell Physiol 2015; 230:1781-93. [PMID: 25503516 DOI: 10.1002/jcp.24881] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2013] [Accepted: 12/05/2014] [Indexed: 12/20/2022]
Abstract
Inhaled cigarette smoke (CS) causes persistent lung inflammation in smokers. Interleukin 8 (IL-8) released from macrophages is a key chemokine during initiation and progression of CS-induced lung inflammation, yet its regulation is largely unknown. AMP-activated protein kinase (AMPK), a crucial energy homeostasis regulator, may modulate inflammation. Here we report that CS extract (CSE) increased the level of intracellular reactive oxygen species (ROS), activating AMPK, mitogen-activated protein kinases (MAPKs), and NF-κB, as well as inducing IL-8, in human macrophages. N-acetyl-cysteine (ROS scavenger) or hexamethonium [nicotinic acetylcholine receptor (nAChR) antagonist] attenuated the CSE-induced increase in intracellular ROS, activation of AMPK and NF-κB, as well as IL-8 induction, which suggests that nAChRs and ROS are important. Prevention of AMPK activation by compound C or AMPK siRNA reduced CSE-induced IL-8 production, confirming the role of AMPK. Compound C or AMPK siRNA also inhibited the activation of MAPKs and NF-κB by CSE, which suggests that these molecules are downstream of AMPK. Additionally, exposure of human macrophages to nicotine activated AMPK and induced IL-8 and that these effects were lessened by hexamethonium or compound C, implying that nicotine in CS may be causative. Furthermore, chronic CS exposure in mice promoted AMPK phosphorylation and expression of MIP-2 (an IL-8 homolog) in infiltrated macrophages and in lung tissues, as well as induced lung inflammation, all of which were reduced by compound C treatment. Thus, we identified a novel nAChRs-dependent, ROS-sensitive, AMPK/MAPKs/NF-κB signaling pathway, which seems to be important to CS-induced macrophage IL-8 production and possibly to lung inflammation.
Collapse
Affiliation(s)
- Hsin-Kuo Ko
- Department of Physiology, School of Medicine, National Yang-Ming University, Taipei, Taiwan; Department of Respiratory Therapy, Taipei Veterans General Hospital, Taipei, Taiwan
| | | | | | | | | | | | | |
Collapse
|
47
|
McGarry Houghton A. Matrix metalloproteinases in destructive lung disease. Matrix Biol 2015; 44-46:167-74. [DOI: 10.1016/j.matbio.2015.02.002] [Citation(s) in RCA: 113] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2015] [Revised: 02/05/2015] [Accepted: 02/06/2015] [Indexed: 01/09/2023]
|
48
|
D'Anna C, Cigna D, Costanzo G, Ferraro M, Siena L, Vitulo P, Gjomarkaj M, Pace E. Cigarette smoke alters cell cycle and induces inflammation in lung fibroblasts. Life Sci 2015; 126:10-8. [DOI: 10.1016/j.lfs.2015.01.017] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2014] [Revised: 01/14/2015] [Accepted: 01/16/2015] [Indexed: 12/11/2022]
|
49
|
Liu F, Zou Y, Huang Q, Zheng L, Wang W. Electronic health records and improved nursing management of chronic obstructive pulmonary disease. Patient Prefer Adherence 2015; 9:495-500. [PMID: 25848228 PMCID: PMC4376268 DOI: 10.2147/ppa.s76562] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
This paper identifies evolving trends in the diagnosis and treatment of chronic obstructive pulmonary disease (COPD), and recommends the integration of nursing strategies in COPD management via widespread implementation of electronic health records. COPD is a complex lung disease with diverse origins, both physical and behavioral, manifested in a wide range of symptoms that further increase the patient's risk for comorbidities. Early diagnosis and effective management of COPD require monitoring of a dizzying array of COPD symptoms over extended periods of time, and nurses are especially well positioned to manage potential progressions of COPD, as frontline health care providers who obtain, record, and organize patient data. Developments in medical technology greatly aid nursing management of COPD, from the deployment of spirometry as a diagnostic tool at the family practice level to newly approved treatment options, including non-nicotine pharmacotherapies that reduce the cravings associated with tobacco withdrawal. Among new medical technologies, electronic health records have proven particularly advantageous in the management of COPD, enabling providers to gather, maintain, and reference more patient data than has ever been possible before. Thus, consistent and widespread implementation of electronic health records facilitates the coordination of diverse treatment strategies, resulting in increased positive health outcomes for patients with COPD.
Collapse
Affiliation(s)
- Fengping Liu
- Yancheng Medical College, Yancheng, Jiangsu Province, People’s Republic of China
| | - Yeqing Zou
- Yancheng Medical College, Yancheng, Jiangsu Province, People’s Republic of China
| | - Qingmei Huang
- The First Affiliated College of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, People’s Republic of China
| | - Li Zheng
- The First Affiliated College of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, People’s Republic of China
| | - Wei Wang
- The First Affiliated College of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, People’s Republic of China
| |
Collapse
|
50
|
Yan H, Zhao L, Wu X, Liu H, Wu C, Li Y, Zheng W, Jiang H. Inflammation and pathological damage to the lungs of mice are only partially reversed following smoking cessation on subacute exposure to cigarette smoke. Mol Med Rep 2015; 11:4246-54. [PMID: 25672547 PMCID: PMC4394953 DOI: 10.3892/mmr.2015.3337] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2014] [Accepted: 01/21/2015] [Indexed: 02/07/2023] Open
Abstract
The present study aimed to observe the level of inflammation and the number of lesions in the airways and parenchyma of mouse lungs subsequent to smoking cessation following 4 weeks exposure to cigarette smoke. Enlargement of the regional airspaces, deposition of peribronchial collagen fibers and macrophage infiltration were assessed. In addition, the expression levels of matrix metalloproteinase (MMP)‑12 and transforming growth factor (TGF)‑β1 were detected in the airways and lung parenchyma of C57BL/6 J mice. Mice, which were exposed to filtered air for 4 weeks or cigarette smoke for 8 weeks were used as control groups. A 4 week duration of smoke exposure induced the expansion of alveolar spaces ~100 µm from the terminal bronchioles, but without increased deposition of collagen around the small airways, which was not reversed following smoking cessation. Pulmonary infiltration of macrophages and the protein expression levels of MMP‑12 and TGF‑β1 increased in the airways following 4 weeks smoke exposure, however, there was no further increase at 8 weeks, and the expression levels of TGF‑β1 in the lung parenchyma decreased. At 4 weeks post‑smoking cessation, the expression levels of TGF‑β1 in the airways and lung parenchyma returned to normal; whereas, 1 week after smoking cessation, the expression levels of MMP‑12 were higher compared with the normal control group. Subacute exposure to cigarette smoke induced an inflammatory response and regional damage to the lung parenchyma, prior to deposition of collagen around the airways. Following smoking cessation, the pulmonary inflammatory reaction was partially reversed, however, macrophage infiltration and the expression levels of MMP‑12 remained significantly higher compared with the control mice. These results suggested that regulation of the expression of MMP‑12 and TGF‑β1, particularly in the distribution in the airways and lung parenchyma, may be a strategy for the early treatment of chronic obstructive pulmonary disease.
Collapse
Affiliation(s)
- Hengyi Yan
- Department of First Respiratory Medicine, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| | - Li Zhao
- Department of First Respiratory Medicine, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| | - Xiaojie Wu
- Department of First Respiratory Medicine, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| | - Hongbo Liu
- Department of First Respiratory Medicine, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| | - Cen Wu
- Department of First Respiratory Medicine, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| | - Yu Li
- Department of First Respiratory Medicine, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| | - Wei Zheng
- Department of First Respiratory Medicine, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| | - Hongfang Jiang
- Department of First Respiratory Medicine, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| |
Collapse
|