1
|
Lee HJ, Lee HY. Characterization of lung function impairment and pathological changes induced by chronic lead and cadmium inhalation: Insights from a mouse model study. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 283:116776. [PMID: 39059344 DOI: 10.1016/j.ecoenv.2024.116776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 07/14/2024] [Accepted: 07/21/2024] [Indexed: 07/28/2024]
Abstract
Exposure to environmental heavy metals such as lead (Pb) and cadmium (Cd) is a global concern due to their widespread presence. However, the specific pulmonary effects of inhaled exposure, especially related to long-term effects, remain poorly understood. In this study, we developed a novel mouse model of Pb and Cd inhalation to mimic real-world conditions and investigate pulmonary effects. Mice were exposed to Pb and Cd inhalation for 6 months using a whole-body exposure system, resulting in decreased lung compliance and progression from emphysematous changes to fibrosis. In addition, the blood Pb/Cd levels of mice exposed to Pb/Cd for 6 months are like those of humans occupationally exposed to heavy metals. Histology revealed inflammation and collagen deposition. Transcriptomic analysis highlighted immune responses and macrophage activity in developing fibrosis. These results confirm an association between Pb/Cd exposure and emphysema and fibrosis, reflecting clinical findings. The study highlights the importance of long-term exposure assessment and time-course analysis for understanding Pb/Cd-induced lung disease. The relevance of the mouse model in replicating human exposure scenarios underscores its value in studying fibrosis and emphysema simultaneously. These findings provide a basis for targeted therapeutic interventions against heavy metal-induced lung injury.
Collapse
Affiliation(s)
- Ho Jin Lee
- College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - Ho-Young Lee
- College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea; Natural Products Research Institute, Seoul National University, Seoul 08826, Republic of Korea; Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Republic of Korea.
| |
Collapse
|
2
|
Balzano E, De Cunto G, Goracci C, Bartalesi B, Cavarra E, Lungarella G, Lucattelli M. Immunohistochemical Study of Airways Fibrous Remodeling in Smoking Mice. J Histochem Cytochem 2023; 71:577-599. [PMID: 37818941 PMCID: PMC10617442 DOI: 10.1369/00221554231204926] [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: 02/08/2023] [Accepted: 09/11/2023] [Indexed: 10/13/2023] Open
Abstract
The fibrotic remodeling in chronic obstructive pulmonary disease (COPD) is held responsible for narrowing of small airways and thus for disease progression. Oxidant damage and cell senescence factors are recently involved in airways fibrotic remodeling. Unfortunately, we have no indications on their sequential expression at anatomical sites in which fibrotic remodeling develops in smoking subjects. Using immunohistochemical techniques, we investigated in two strains of mice after cigarette smoke (CS) exposure what happens at various times in airway areas where fibrotic remodeling occurs, and if there also exists correspondence among DNA damage induced by oxidants, cellular senescence, the presence of senescence-secreted factors involved in processes that affect transcription, metabolism as well as apoptosis, and the onset of fibrous remodeling that appears at later times in mice exposed to CS. A clear positivity for fibrogenic cytokines TGF-β, PDGF-B, and CTGF, and for proliferation marker PCNA around airways that will be remodeled is observed in both strains. Increased expression of p16ink4A senescence marker and MyoD is also seen in the same areas. p16ink4A and MyoD can promote cell cycle arrest, terminal differentiation of myofibroblasts, and can oppose their dedifferentiation. Of interest, an early progressive attenuation of SIRT-1 is observed after CS exposure. This intracellular regulatory protein can reduce premature cell senescence. These findings suggest that novel agents, which promote myofibroblast dedifferentiation and/or the apoptosis of senescent cells, may dampen progression of airway changes in smoking COPD subjects.
Collapse
Affiliation(s)
- Emilia Balzano
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - Giovanna De Cunto
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - Chiara Goracci
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - Barbara Bartalesi
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - Eleonora Cavarra
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - Giuseppe Lungarella
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - Monica Lucattelli
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| |
Collapse
|
3
|
Akbasheva OE, Spirina LV, Dyakov DA, Masunova NV. Proteolysis and Deficiency of α1-Proteinase Inhibitor in SARS-CoV-2 Infection. BIOCHEMISTRY (MOSCOW) SUPPLEMENT. SERIES B, BIOMEDICAL CHEMISTRY 2022; 16:271-291. [PMID: 36407837 PMCID: PMC9668222 DOI: 10.1134/s1990750822040035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 03/30/2022] [Accepted: 04/11/2022] [Indexed: 11/17/2022]
Abstract
The SARS-CoV-2 pandemic had stimulated the emergence of numerous publications on the α1-proteinase inhibitor (α1-PI, α1-antitrypsin), especially when it was found that the regions of high mortality corresponded to the regions with deficient α1-PI alleles. By analogy with the data obtained in the last century, when the first cause of the genetic deficiency of α1-antitrypsin leading to elastase activation in pulmonary emphysema was proven, it can be supposed that proteolysis hyperactivation in COVID-19 may be associated with the impaired functions of α1-PI. The purpose of this review was to systematize the scientific data and critical directions for translational studies on the role of α1-PI in SARS-CoV-2-induced proteolysis hyperactivation as a diagnostic marker and a therapeutic target. This review describes the proteinase-dependent stages of viral infection: the reception and penetration of the virus into a cell and the imbalance of the plasma aldosterone-angiotensin-renin, kinin, and blood clotting systems. The role of ACE2, TMPRSS, ADAM17, furin, cathepsins, trypsin- and elastase-like serine proteinases in the virus tropism, the activation of proteolytic cascades in blood, and the COVID-19-dependent complications is considered. The scientific reports on α1-PI involvement in the SARS-CoV-2-induced inflammation, the relationship with the severity of infection and comorbidities were analyzed. Particular attention is paid to the acquired α1-PI deficiency in assessing the state of patients with proteolysis overactivation and chronic non-inflammatory diseases, which are accompanied by the risk factors for comorbidity progression and the long-term consequences of COVID-19. Essential data on the search and application of protease inhibitor drugs in the therapy for bronchopulmonary and cardiovascular pathologies were analyzed. The evidence of antiviral, anti-inflammatory, anticoagulant, and anti-apoptotic effects of α1-PI, as well as the prominent data and prospects for its application as a targeted drug in the SARS-CoV-2 acquired pneumonia and related disorders, are presented.
Collapse
Affiliation(s)
| | - L. V. Spirina
- Siberian State Medical University, 634050 Tomsk, Russia
- Cancer Research Institute, Tomsk National Research Medical Center, 634009 Tomsk, Russia
| | - D. A. Dyakov
- Siberian State Medical University, 634050 Tomsk, Russia
| | | |
Collapse
|
4
|
Akbasheva OE, Spirina LV, Dyakov DA, Masunova NV. [Proteolysis and deficiency of α1-proteinase inhibitor in SARS-CoV-2 infection]. BIOMEDITSINSKAIA KHIMIIA 2022; 68:157-176. [PMID: 35717581 DOI: 10.18097/pbmc20226803157] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
The SARS-CoV-2 pandemia had stimulated the numerous publications emergence on the α1-proteinase inhibitor (α1-PI, α1-antitrypsin), primarily when it was found that high mortality in some regions corresponded to the regions with deficient α1-PI alleles. By analogy with the last century's data, when the root cause of the α1-antitrypsin, genetic deficiency leading to the elastase activation in pulmonary emphysema, was proven. It is evident that proteolysis hyperactivation in COVID-19 may be associated with α1-PI impaired functions. The purpose of this review is to systematize scientific data, critical directions for translational studies on the role of α1-PI in SARS-CoV-2-induced proteolysis hyperactivation as a diagnostic marker and a target in therapy. This review describes the proteinase-dependent stages of a viral infection: the reception and virus penetration into the cell, the plasma aldosterone-angiotensin-renin, kinins, blood clotting systems imbalance. The ACE2, TMPRSS, ADAM17, furin, cathepsins, trypsin- and elastase-like serine proteinases role in the virus tropism, proteolytic cascades activation in blood, and the COVID-19-dependent complications is presented. The analysis of scientific reports on the α1-PI implementation in the SARS-CoV-2-induced inflammation, the links with the infection severity, and comorbidities were carried out. Particular attention is paid to the acquired α1-PI deficiency in assessing the patients with the proteolysis overactivation and chronic non-inflammatory diseases that are accompanied by the risk factors for the comorbidities progression, and the long-term consequences of COVID-19 initiation. Analyzed data on the search and proteases inhibitory drugs usage in the bronchopulmonary cardiovascular pathologies therapy are essential. It becomes evident the antiviral, anti-inflammatory, anticoagulant, anti-apoptotic effect of α1-PI. The prominent data and prospects for its application as a targeted drug in the SARS-CoV-2 acquired pneumonia and related disorders are presented.
Collapse
Affiliation(s)
| | - L V Spirina
- Siberian State Medical University, Tomsk, Russia; Cancer Research Institute, Tomsk National Research Medical Center, Tomsk, Russia
| | - D A Dyakov
- Siberian State Medical University, Tomsk, Russia
| | - N V Masunova
- Siberian State Medical University, Tomsk, Russia
| |
Collapse
|
5
|
Sangani R, Ghio A, Culp S, Patel Z, Sharma S. Combined Pulmonary Fibrosis Emphysema: Role of Cigarette Smoking and Pulmonary Hypertension in a Rural Cohort. Int J Chron Obstruct Pulmon Dis 2021; 16:1873-1885. [PMID: 34188464 PMCID: PMC8232869 DOI: 10.2147/copd.s307192] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Accepted: 05/20/2021] [Indexed: 12/24/2022] Open
Abstract
Background Disease heterogeneity in idiopathic pulmonary fibrosis (IPF) often complicates the systematic study of disease, management of patients and clinical investigations. Objective To describe combined pulmonary fibrosis emphysema (CPFE) phenotype in a rural Appalachian IPF cohort with the highest smoking rates in the United States. Methods CPFE patients (n = 60) in a developed IPF cohort (n = 153) were characterized. Groups (CPFE vs IPF without emphysema) were categorized based on the predominant HRCT patterns of UIP (n = 109). Demographics, clinical variables, and treatment details were recorded. Kaplan–Meier survival and multivariate logistic regression analysis were performed. Results The prevalence of CPFE in our IPF cohort was 45% (n = 49). The CPFE group was younger (73.9 vs 78.2), had a more extensive smoking history (93.9% vs 53.3%) with greater mean smoking pack years (49.09 vs 15.39) and had lower percentage predicted DLCO on presentation (38.35 vs 51.09) compared to IPF without emphysema group. Both groups shared equivalent higher burden of comorbidities, including pulmonary hypertension (PH) (46.9% vs 33.3%). One-fifth of patients were prescribed antifibrotics and only a subset (5%) of patients underwent lung transplantation. There was a non-significant trend towards reduced survival in CPFE (p = 0.076). Smoking status and DLCO predicted CPFE in our cohort. Body mass index (BMI), PH, and pirfenidone use were significant predictors of mortality. Conclusion CPFE was highly prevalent in our rural IPF cohort. In contrast to previous studies, CPFE group was older and had higher female (approx. 30%) occurrence. A greater exposure to cigarette smoke and reduced DLCO at diagnosis predicted CPFE. Lower BMI and PH predicted higher mortality whereas use of pirfenidone improved survival in our cohort. This study highlights a complex interaction of cigarette smoking, advanced fibrosis of UIP, PH and potential utility of antifibrotic agents in CPFE phenotype. Substantial burden of comorbidities, older age, and the limited utilization of advanced therapeutics in the cohort emphasize the challenges faced by rural Appalachian patients.
Collapse
Affiliation(s)
- Rahul Sangani
- Section of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, West Virginia University, Morgantown, WV, USA
| | - Andrew Ghio
- Human Studies Facility, US Environmental Protection Agency, Chapel Hill, NC, USA
| | - Stacey Culp
- Department of Biostatistics, West Virginia University, Morgantown, WV, USA
| | - Zalak Patel
- Department of Radiology, West Virginia University, Morgantown, WV, USA
| | - Sunil Sharma
- Section of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, West Virginia University, Morgantown, WV, USA
| |
Collapse
|
6
|
Taha MM, Saad-Hussein A, Mahdy-Abdallah H. Association of microsomal epoxide hydrolase gene (fast genotype) with lung functions impairment in wood workers. JOURNAL OF COMPLEMENTARY & INTEGRATIVE MEDICINE 2021; 18:609-615. [PMID: 33794079 DOI: 10.1515/jcim-2020-0085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/15/2020] [Accepted: 07/09/2020] [Indexed: 06/12/2023]
Abstract
OBJECTIVES Exposure to wood dust may lead to impairment of the lung functions. Microsomal epoxide hydrolase enzyme (EPHX1) was shown to take part in protection against oxidative stress. An alteration in enzyme activity might be associated with its gene polymorphisms. In vitro polymorphisms in exons 3 (His113Tyr) and 4 (Arg139His) lead to reduced activity (slow allele) and increased activity (fast allele). Macrophage inflammatory protein 2 (MIP-2) is produced in rat lung epithelial cells after exposure to fine particles. We aimed to investigate the associations between mEPHX1 polymorphisms (in exon 3 and 4) and lung function in furniture workers and assessment of MIP-2 effect. METHODS Our study was performed on 70 wood dust exposed male workers and 70 matched normal controls subjects. Ventilatory function tests were measured by spirometer, MIP-2 was performed by ELISA methods and EPHX gene was done by polymerase chain reaction-restriction fragment length polymorphism (PCR-RFLP) methods for each participant. RESULTS Significant reduction in forced vital capacity (FVC%) and forced expiratory volume in the first second (FEV1) levels in Tyr-Tyr and Tyr-Hist genotypes of EPHX (exon 3) was observed. Reduced peak expiratory flow (PEF) levels and significant rise in MIP-2 levels were detected in Tyr-Tyr genotype. While high significant reduction in FVC% and FEV1 levels were shown in different genotypes in exon 4. Significant rise was observed in MIP-2 levels in Hist-Hist genotype of exon 4. An increase in duration of exposure showed positive correlation with fall in ventilatory functions. CONCLUSIONS It was concluded that in Hist139Arg of EPHX gene, fast genotype (Arg-Arg) was associated with impaired ventilatory functions.
Collapse
Affiliation(s)
- Mona M Taha
- Department of Environmental and Occupational Medicine, Environmental Research Division, National Research Centre, Giza, Egypt
| | - Amal Saad-Hussein
- Department of Environmental and Occupational Medicine, Environmental Research Division, National Research Centre, Giza, Egypt
| | - Heba Mahdy-Abdallah
- Department of Environmental and Occupational Medicine, Environmental Research Division, National Research Centre, Giza, Egypt
| |
Collapse
|
7
|
Lungarella G, Lucattelli M, Cavarra E, De Cunto G. Comments on “Air Space Distension Precedes Spontaneous Fibrotic Remodeling and Impaired Cholesterol Metabolism in the Absence of Surfactant Protein C”. Am J Respir Cell Mol Biol 2020; 63:398-399. [DOI: 10.1165/rcmb.2020-0125le] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
|
8
|
De Cunto G, Cavarra E, Bartalesi B, Lungarella G, Lucattelli M. Alveolar Macrophage Phenotype and Compartmentalization Drive Different Pulmonary Changes in Mouse Strains Exposed to Cigarette Smoke. COPD 2020; 17:429-443. [PMID: 32597232 DOI: 10.1080/15412555.2020.1783648] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
COPD can manifest itself with different clinical phenotypes characterized by different disease progression and response to therapy. Although a remarkable number of studies have been carried out, little is known about the mechanisms underlying phenotypes that could guide the development of viable future therapies. Several murine strains mirror some human phenotypes after smoke exposure. It was of interest to investigate in these strains whether different pattern of activation of macrophages, and their distribution in lungs, is associated to changes characterizing different phenotypes. We chose C57Bl/6, and Lck deficient mice, which show significant emphysema, DBA/2 mice that develop changes similar to those of "pulmonary fibrosis/emphysema syndrome", p66Shc ko mice that develop bronchiolitis with fibrosis but not emphysema, and finally ICR mice that do not develop changes at 7 months after smoke exposure. Unlike other strains, ICR mice show very few activated macrophages (Mac-3 positive) mostly negative to M1 or M2 markers. On the other hand, a large population of M1 macrophages predominates in the lung periphery of DBA/2, C57Bl/6 and in Lck deficient mice, where emphysema is more evident. M2 macrophages are mainly observed in subpleural and intraparenchymal areas of DBA/2 mice and around bronchioles of p66Shc ko mice where fibrotic changes are present. We observed slight but significant differences in mRNA expression of iNOS, ECF-L, arginase 1, IL-4, IL-13 and TGF-β between air- and smoke-exposed mice. These differences together with the different compartmentalization of macrophages may offer an explanation for the diversity of lesions and their distribution that we observed among the strains.
Collapse
Affiliation(s)
- Giovanna De Cunto
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - Eleonora Cavarra
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - Barbara Bartalesi
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - Giuseppe Lungarella
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - Monica Lucattelli
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| |
Collapse
|
9
|
Dawod YT, Cook NE, Graham WB, Madhani-Lovely F, Thao C. Smoking-associated interstitial lung disease: update and review. Expert Rev Respir Med 2020; 14:825-834. [PMID: 32379511 DOI: 10.1080/17476348.2020.1766971] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
INTRODUCTION Smoking-associated interstitial lung disease manifests as several heterogeneous disorders involving the airways, pleura, and lung parenchyma with various radiological patterns. The clinical history, radiologic, and pathologic findings are important to distinguish these more uncommon diseases. A multidisciplinary approach is recommended for diagnosis and to manage these conditions appropriately. AREAS COVERED This review provides an overview of the epidemiology, risk factors, pathogenesis, clinical features, diagnosis, and treatment of acute eosinophilic pneumonia, e-cigarettes, or vaping associated lung injury, respiratory bronchiolitis interstitial lung disease, desquamative interstitial pneumonitis, pulmonary Langerhans cell histiocytosis, idiopathic pulmonary fibrosis, and combined pulmonary fibrosis emphysema. EXPERT OPINION Cigarette smoking is associated with a variety of pathologic conditions that affect the airways and lungs. E-cigarette use and vaping present new challenges to the clinician. Consensus between the clinical, radiographic, and pathologic findings is important in identifying and differentiating between the various entities to properly diagnose smoking-related interstitial lung diseases discussed in this review.
Collapse
Affiliation(s)
- Yaser T Dawod
- Section of Pulmonary and Critical Care Medicine, Medstar Washington Hospital Center , Washington, USA
| | - Noah E Cook
- Department of Pulmonary and Critical Care Medicine, University of Nevada Las Vegas School of Medicine , Las Vegas, USA
| | - William B Graham
- Department of Pulmonary and Critical Care Medicine, Renown Health , Reno, USA
| | | | - Choua Thao
- Department of Pulmonary and Critical Care Medicine, Renown Health , Reno, USA
| |
Collapse
|
10
|
De Cunto G, Cavarra E, Bartalesi B, Lucattelli M, Lungarella G. Innate Immunity and Cell Surface Receptors in the Pathogenesis of COPD: Insights from Mouse Smoking Models. Int J Chron Obstruct Pulmon Dis 2020; 15:1143-1154. [PMID: 32547002 PMCID: PMC7246326 DOI: 10.2147/copd.s246219] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Accepted: 05/03/2020] [Indexed: 12/23/2022] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is mainly associated with smoking habit. Inflammation is the major initiating process whereby neutrophils and monocytes are attracted into the lung microenvironment by external stimuli present in tobacco leaves and in cigarette smoke, which promote chemotaxis, adhesion, phagocytosis, release of superoxide anions and enzyme granule contents. A minority of smokers develops COPD and different molecular factors, which contribute to the onset of the disease, have been put forward. After many years of research, the pathogenesis of COPD is still an object of debate. In vivo models of cigarette smoke-induced COPD may help to unravel cellular and molecular mechanisms underlying the pathogenesis of COPD. The mouse represents the most favored animal choice with regard to the study of immune mechanisms due to its genetic and physiological similarities to humans, the availability of a large variability of inbred strains, the presence in the species of several genetic disorders analogous to those in man, and finally on the possibility to create models “made-to-measure” by genetic manipulation. The review outlines the different response of mouse strains to cigarette smoke used in COPD studies while retaining a strong focus on their relatability to human patients. These studies reveal the importance of innate immunity and cell surface receptors in the pathogenesis of pulmonary injury induced by cigarette smoking. They further advance the way in which we use wild type or genetically manipulated strains to improve our overall understanding of a multifaceted disease such as COPD. The structural and functional features, which have been found in the different strains of mice after chronic exposure to cigarette smoke, can be used in preclinical studies to develop effective new therapeutic agents for the different phenotypes in human COPD.
Collapse
Affiliation(s)
- Giovanna De Cunto
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - Eleonora Cavarra
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - Barbara Bartalesi
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - Monica Lucattelli
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - Giuseppe Lungarella
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| |
Collapse
|
11
|
De Cunto G, Brancaleone V, Riemma MA, Cerqua I, Vellecco V, Spaziano G, Cavarra E, Bartalesi B, D'Agostino B, Lungarella G, Cirino G, Lucattelli M, Roviezzo F. Functional contribution of sphingosine-1-phosphate to airway pathology in cigarette smoke-exposed mice. Br J Pharmacol 2019; 177:267-281. [PMID: 31499592 DOI: 10.1111/bph.14861] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Revised: 07/16/2019] [Accepted: 08/13/2019] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND AND PURPOSE A critical role for sphingosine kinase/sphingosine-1-phosphate (S1P) pathway in the control of airway function has been demonstrated in respiratory diseases. Here, we address S1P contribution in a mouse model of mild chronic obstructive pulmonary disease (COPD). EXPERIMENTAL APPROACH C57BL/6J mice have been exposed to room air or cigarette smoke up to 11 months and killed at different time points. Functional and molecular studies have been performed. KEY RESULTS Cigarette smoke caused emphysematous changes throughout the lung parenchyma coupled to a progressive collagen deposition in both peribronchiolar and peribronchial areas. The high and low airways showed an increased reactivity to cholinergic stimulation and α-smooth muscle actin overexpression. Similarly, an increase in airway reactivity and lung resistances following S1P challenge occurred in smoking mice. A high expression of S1P, Sph-K2 , and S1P receptors (S1P2 and S1P3 ) has been detected in the lung of smoking mice. Sphingosine kinases inhibition reversed the increased cholinergic response in airways of smoking mice. CONCLUSIONS AND IMPLICATIONS S1P signalling up-regulation follows the disease progression in smoking mice and is involved in the development of airway hyperresponsiveness. Our study defines a therapeutic potential for S1P inhibitors in management of airways hyperresponsiveness associated to emphysema in smokers with both asthma and COPD.
Collapse
Affiliation(s)
- Giovanna De Cunto
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | | | | | - Ida Cerqua
- Department of Pharmacy, University of Naples Federico II, Naples, Italy
| | | | - Giuseppe Spaziano
- Department of Experimental Medicine L. Donatelli, Section of Pharmacology, School of Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Eleonora Cavarra
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - Barbara Bartalesi
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - Bruno D'Agostino
- Department of Experimental Medicine L. Donatelli, Section of Pharmacology, School of Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Giuseppe Lungarella
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - Giuseppe Cirino
- Department of Pharmacy, University of Naples Federico II, Naples, Italy
| | - Monica Lucattelli
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | | |
Collapse
|
12
|
Flores-Suárez LF, Sacoto G. Interstitial Lung Disease and ANCA-Associated Vasculitis. CURRENT TREATMENT OPTIONS IN RHEUMATOLOGY 2019. [DOI: 10.1007/s40674-019-00127-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
|
13
|
Cornwell WD, Kim C, Lastra AC, Dass C, Bolla S, Wang H, Zhao H, Ramsey FV, Marchetti N, Rogers TJ, Criner GJ. Inflammatory signature in lung tissues in patients with combined pulmonary fibrosis and emphysema. Biomarkers 2018; 24:232-239. [PMID: 30411980 DOI: 10.1080/1354750x.2018.1542458] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Background: The aetiology and inflammatory profile of combined pulmonary fibrosis and emphysema (CPFE) remain uncertain currently. Objective: We aimed to examine the levels of inflammatory proteins in lung tissue in a cohort of patients with emphysema, interstitial pulmonary fibrosis (IPF), and CPFE. Materials and methods: Explanted lungs were obtained from subjects with emphysema, IPF, CPFE, (or normal subjects), and tissue extracts were prepared. Thirty-four inflammatory proteins were measured in each tissue section. Results: The levels of all 34 proteins were virtually indistinguishable in IPF compared with CPFE tissues, and collectively, the inflammatory profile in the emphysematous tissues were distinct from IPF and CPFE. Moreover, inflammatory protein levels were independent of the severity of the level of diseased tissue. Conclusions: We find that emphysematous lung tissues have a distinct inflammatory profile compared with either IPF or CPFE. However, the inflammatory profile in CPFE lungs is essentially identical to lungs from patients with IPF. These data suggest that distinct inflammatory processes collectively contribute to the disease processes in patients with emphysema, when compared to IPF and CPFE.
Collapse
Affiliation(s)
- William D Cornwell
- a Center for Inflammation, Translational and Clinical Lung Research, Lewis Katz School of Medicine , Temple University , Philadelphia , PA , USA.,b Department of Thoracic Medicine and Surgery, Lewis Katz School of Medicine , Temple University , Philadelphia , PA , USA
| | - Cynthia Kim
- b Department of Thoracic Medicine and Surgery, Lewis Katz School of Medicine , Temple University , Philadelphia , PA , USA
| | - Alejandra C Lastra
- b Department of Thoracic Medicine and Surgery, Lewis Katz School of Medicine , Temple University , Philadelphia , PA , USA
| | - Chandra Dass
- c Department of Radiology, Lewis Katz School of Medicine , Temple University , Philadelphia , PA , USA
| | - Sudhir Bolla
- b Department of Thoracic Medicine and Surgery, Lewis Katz School of Medicine , Temple University , Philadelphia , PA , USA
| | - He Wang
- d Department of Pathology and Laboratory Medicine, Lewis Katz School of Medicine , Temple University , Philadelphia , PA , USA
| | - Huaqing Zhao
- e Department of Clinical Sciences, Lewis Katz School of Medicine , Temple University , Philadelphia , PA , USA
| | - Frederick V Ramsey
- e Department of Clinical Sciences, Lewis Katz School of Medicine , Temple University , Philadelphia , PA , USA
| | - Nathaniel Marchetti
- a Center for Inflammation, Translational and Clinical Lung Research, Lewis Katz School of Medicine , Temple University , Philadelphia , PA , USA.,b Department of Thoracic Medicine and Surgery, Lewis Katz School of Medicine , Temple University , Philadelphia , PA , USA
| | - Thomas J Rogers
- a Center for Inflammation, Translational and Clinical Lung Research, Lewis Katz School of Medicine , Temple University , Philadelphia , PA , USA.,b Department of Thoracic Medicine and Surgery, Lewis Katz School of Medicine , Temple University , Philadelphia , PA , USA
| | - Gerard J Criner
- a Center for Inflammation, Translational and Clinical Lung Research, Lewis Katz School of Medicine , Temple University , Philadelphia , PA , USA.,b Department of Thoracic Medicine and Surgery, Lewis Katz School of Medicine , Temple University , Philadelphia , PA , USA
| |
Collapse
|
14
|
De Cunto G, Bartalesi B, Cavarra E, Balzano E, Lungarella G, Lucattelli M. Ongoing Lung Inflammation and Disease Progression in Mice after Smoking Cessation: Beneficial Effects of Formyl-Peptide Receptor Blockade. THE AMERICAN JOURNAL OF PATHOLOGY 2018; 188:2195-2206. [PMID: 30031729 DOI: 10.1016/j.ajpath.2018.06.010] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Revised: 06/04/2018] [Accepted: 06/18/2018] [Indexed: 01/24/2023]
Abstract
The most important risk factor for chronic obstructive pulmonary disease (COPD) is cigarette smoking. Until now, smoking cessation (SC) is the only treatment effective in slowing down the progression of the disease. However, in many cases SC may only relieve the airflow obstruction and inflammatory response. Consequently, a persistent lung inflammation in ex-smokers is associated with progressive deterioration of respiratory functions. This is an increasingly important clinical problem whose mechanistic basis remains poorly understood. Available therapies do not adequately suppress inflammation and are not able to stop the vicious cycle that is at the basis of persistent inflammation. In addition, in mice after SC an ongoing inflammation and progressive lung deterioration is observed. After 4 months of smoke exposure mice show mild emphysematous changes. Lung inflammation is still present after SC, and emphysema progresses during the next 6-month period of observation. Destruction of alveolar walls is associated with airways remodeling (goblet cell metaplasia and peribronchiolar fibrosis). Modulation of formyl-peptide receptor signaling with antagonists mitigates inflammation and prevents deterioration of lung structures. This study suggests an important role for N-formylated peptides in the progression and exacerbation of COPD. Modulating formyl-peptide receptor signal should be explored as a potential new therapy for COPD.
Collapse
Affiliation(s)
- Giovanna De Cunto
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - Barbara Bartalesi
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - Eleonora Cavarra
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - Emilia Balzano
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - Giuseppe Lungarella
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - Monica Lucattelli
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy.
| |
Collapse
|
15
|
Alba MA, Flores-Suárez LF, Henderson AG, Xiao H, Hu P, Nachman PH, Falk RJ, Charles Jennette J. Interstital lung disease in ANCA vasculitis. Autoimmun Rev 2017; 16:722-729. [PMID: 28479484 DOI: 10.1016/j.autrev.2017.05.008] [Citation(s) in RCA: 85] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2017] [Accepted: 04/07/2017] [Indexed: 02/03/2023]
Abstract
Anti-neutrophil cytoplasmic antibodies (ANCA) vasculitides are immune-mediated disorders that primarily affect small blood vessels of the airway and kidneys. Lung involvement, one of the hallmarks of microscopic polyangiitis and granulomatosis with polyangiitis, is associated with increased mortality and morbidity. In recent years, several retrospective series and case reports have described the association of interstitial lung disease (ILD) and ANCA vasculitis, particularly those positive for ANCA specific for myeloperoxidase. In the majority of these patients pulmonary fibrosis occurs concurrently or predates the diagnosis of ANCA vasculitis. More importantly, these studies have shown that ILD has an adverse impact on the long-term prognosis of ANCA vasculitis. This review focuses on the main clinical and radiologic features of pulmonary fibrosis associated with anti-neutrophil cytoplasmic antibodies. Major histopathology features, prognosis and therapeutic options are summarized.
Collapse
Affiliation(s)
- Marco A Alba
- Department of Pathology and Laboratory Medicine, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Luis Felipe Flores-Suárez
- Primary Systemic Vasculitis Clinic, Instituto Nacional de Enfermedades Respiratorias (INER), Mexico City, Mexico
| | - Ashley G Henderson
- Division of Pulmonary & Critical Care Medicine, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Hong Xiao
- Department of Pathology and Laboratory Medicine, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Peiqi Hu
- Department of Pathology and Laboratory Medicine, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Patrick H Nachman
- University of North Carolina Kidney Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Ronald J Falk
- University of North Carolina Kidney Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - J Charles Jennette
- Department of Pathology and Laboratory Medicine, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| |
Collapse
|
16
|
Yamagata M, Ikeda K, Tsushima K, Iesato K, Abe M, Ito T, Kashiwakuma D, Kagami SI, Iwamoto I, Nakagomi D, Sugiyama T, Maruyama Y, Furuta S, Jayne D, Uno T, Tatsumi K, Nakajima H. Prevalence and Responsiveness to Treatment of Lung Abnormalities on Chest Computed Tomography in Patients With Microscopic Polyangiitis: A Multicenter, Longitudinal, Retrospective Study of One Hundred Fifty Consecutive Hospital-Based Japanese Patients. Arthritis Rheumatol 2016; 68:713-23. [PMID: 26474081 DOI: 10.1002/art.39475] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2014] [Accepted: 10/13/2015] [Indexed: 12/27/2022]
Abstract
OBJECTIVE To determine the prevalence of lung abnormalities on chest computed tomography (CT) in patients with microscopic polyangiitis (MPA), to assess the responsiveness of such abnormalities to initial treatment, and to assess associations between these abnormalities and patient and disease characteristics and mortality. METHODS We retrospectively identified 167 consecutive hospital-based patients with MPA in 3 hospitals in Japan. We longitudinally collected clinical information for 150 of these patients, for whom CT images obtained before treatment were available. We then determined the presence of 22 imaging components of lung abnormalities in these patients. RESULTS The vast majority of patients (97%) had at least 1 lung abnormality on chest CT images, including interstitial lung lesions (66%), airway lesions (66%), pleural lesions (53%), and emphysematous lesions (37%). In multivariate analyses, ground-glass opacity was associated with the Birmingham Vasculitis Activity Score, whereas 3 of 4 airway lesions were associated with myeloperoxidase-antineutrophil cytoplasmic antibodies. Latent class analysis identified a distinct group of patients with airway-predominant lung lesions. Airway lesions such as bronchiolitis and bronchovascular bundle thickening were among the components that showed improvement within 3 months of the initial treatment. An idiopathic pulmonary fibrosis pattern was the only chest CT variable that was independently associated with shorter survival. CONCLUSION Abnormalities in a wide range of anatomic areas, including the whole airway, can be identified in the lungs of patients with MPA before treatment. The prevalence, clustering patterns, and responsiveness to treatment of individual lung abnormalities provide groundwork for informing future studies to understand the pathophysiology of MPA.
Collapse
Affiliation(s)
| | | | | | | | | | - Takashi Ito
- Chiba University, Chica City and Asahi General Hospital, Asahi, Chiba, Japan
| | | | | | | | - Daiki Nakagomi
- National Hospital Organization Shimoshizu Hospital, Yotsukaido, Chiba, Japan
| | - Takao Sugiyama
- National Hospital Organization Shimoshizu Hospital, Yotsukaido, Chiba, Japan
| | | | - Shunsuke Furuta
- Chiba University, Chiba, Japan and Addenbrooke's Hospital, Cambridge University Hospitals, Cambridge, UK
| | - David Jayne
- Addenbrooke's Hospital, Cambridge University Hospitals, Cambridge, UK
| | | | | | | |
Collapse
|
17
|
Papaioannou AI, Kostikas K, Manali ED, Papadaki G, Roussou A, Kolilekas L, Borie R, Bouros D, Papiris SA. Combined pulmonary fibrosis and emphysema: The many aspects of a cohabitation contract. Respir Med 2016; 117:14-26. [PMID: 27492509 DOI: 10.1016/j.rmed.2016.05.005] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2015] [Revised: 03/20/2016] [Accepted: 05/05/2016] [Indexed: 12/12/2022]
Abstract
Combined pulmonary fibrosis and emphysema (CPFE) is a clinical entity characterized by the coexistence of upper lobe emphysema and lower lobe fibrosis. Patients with this condition experience severe dyspnea and impaired gas exchange with preserved lung volumes. The diagnosis of the CPFE syndrome is based on HRCT imaging, showing the coexistence of emphysema and pulmonary fibrosis both in varying extent and locations within the lung parenchyma. Individual genetic background seem to predispose to the development of the disease. The risk of the development of pulmonary hypertension in patients with CPFE is high and related to poor prognosis. CPFE patients also present a high risk of lung cancer. Mortality is significant in patients with CPFE and median survival is reported between 2.1 and 8.5 years. Currently, no specific recommendations are available regarding the management of patients with CPFE. In this review we provide information on the existing knowledge on CPFE regarding the pathophysiology, clinical manifestations, imaging, complications, possible therapeutic interventions and prognosis of the disease.
Collapse
Affiliation(s)
- Andriana I Papaioannou
- 2nd Respiratory Medicine Department, "Attikon" University Hospital, Athens Medical School, National and Kapodistrian University of Athens, Greece.
| | - Konstantinos Kostikas
- 2nd Respiratory Medicine Department, "Attikon" University Hospital, Athens Medical School, National and Kapodistrian University of Athens, Greece.
| | - Effrosyni D Manali
- 2nd Respiratory Medicine Department, "Attikon" University Hospital, Athens Medical School, National and Kapodistrian University of Athens, Greece.
| | - Georgia Papadaki
- 2nd Respiratory Medicine Department, "Attikon" University Hospital, Athens Medical School, National and Kapodistrian University of Athens, Greece.
| | - Aneza Roussou
- 2nd Respiratory Medicine Department, "Attikon" University Hospital, Athens Medical School, National and Kapodistrian University of Athens, Greece.
| | - Likurgos Kolilekas
- 7th Department of Pneumonology, "Sotiria" Chest Diseases Hospital, Athens, Greece.
| | - Raphaël Borie
- APHP, Hôpital Bichat, DHU FIRE Service de Pneumologie A, Centre de compétence des maladies pulmonaires rares, INSERM, Unité 1152, Université Paris Diderot, Paris, France.
| | - Demosthenis Bouros
- 1st Respiratory Medicine Department, "Sotiria" Chest Diseases Hospital, Athens, Medical School, National and Kapodistrian University of Athens, Greece.
| | - Spyridon A Papiris
- 2nd Respiratory Medicine Department, "Attikon" University Hospital, Athens Medical School, National and Kapodistrian University of Athens, Greece.
| |
Collapse
|
18
|
De Cunto G, Lunghi B, Bartalesi B, Cavarra E, Fineschi S, Ulivieri C, Lungarella G, Lucattelli M. Severe Reduction in Number and Function of Peripheral T Cells Does Not Afford Protection toward Emphysema and Bronchial Remodeling Induced in Mice by Cigarette Smoke. THE AMERICAN JOURNAL OF PATHOLOGY 2016; 186:1814-1824. [PMID: 27157991 DOI: 10.1016/j.ajpath.2016.03.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Revised: 02/24/2016] [Accepted: 03/04/2016] [Indexed: 11/28/2022]
Abstract
The protein Lck (p56(Lck)) is a Src family tyrosine kinase expressed at all stages of thymocyte development and is required for maturation of T cells. The targeted disruption of Lck gene in mice results in severe block in thymocyte maturation with substantial reduction in the development of CD4(+)CD8(+) thymocytes, severe reduction of peripheral T cells, and disruption of T-cell receptor signaling with defective function of T-cell responses. To investigate the role of T lymphocyte in the development of cigarette smoke-induced pulmonary changes, Lck(-/-) mice and corresponding congenic wild-type mice were chronically exposed to cigarette smoke, and their lungs were analyzed by biochemical, immunologic, and morphometric methods. Smoking mice from both genotypes showed disseminated foci of emphysema and large areas of goblet cell metaplasia in bronchial and bronchiolar epithelium. Morphometric evaluation of lung changes and lung elastin determination confirmed that mice from both genotypes showed the same degree of emphysematous lesions. Thus, cigarette smoke exposure in the presence of severe reduction in number and function of peripheral T cells does not influence the development of pulmonary changes induced by cigarette smoke. The data obtained suggest that innate immunity is a leading actor in the early development of pulmonary changes in smoking mice and that the adaptive immune response may play a role at later stages.
Collapse
Affiliation(s)
- Giovanna De Cunto
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - Benedetta Lunghi
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - Barbara Bartalesi
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - Eleonora Cavarra
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - Silvia Fineschi
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | | | - Giuseppe Lungarella
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy.
| | - Monica Lucattelli
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy.
| |
Collapse
|
19
|
Trajano LASN, Trajano ETL, Lanzetti M, Mendonça MSA, Guilherme RF, Figueiredo RT, Benjamim CF, Valenca SS, Costa AMA, Porto LC. Elastase modifies bleomycin-induced pulmonary fibrosis in mice. Acta Histochem 2016; 118:203-12. [PMID: 26852294 DOI: 10.1016/j.acthis.2015.12.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2015] [Revised: 11/17/2015] [Accepted: 12/23/2015] [Indexed: 01/24/2023]
Abstract
Pulmonary fibrosis (PF) is characterized by excessive accumulation of collagen in the lungs. Emphysema is characterized by loss of the extracellular matrix (ECM) and alveolar enlargement. We studied the co-participation of elastase-induced mild emphysema in bleomycin-induced PF in mice by analyzing oxidative stress, inflammation and lung histology. C57BL/6 mice were divided into four groups: control; bleomycin (0.1U/mouse); elastase (using porcine pancreatic elastase (PPE)+bleomycin (3U/mouse 14 days before 0.1U/mouse of bleomycin; PPE+B); elastase (3U/mouse). Mice were humanely sacrificed 7, 14 and 21 days after treatment with bleomycin or vehicle. PF was observed 14 days and 21 days after bleomycin treatment but was observed after 14 days only in the PPE+B group. In the PPE+B group at 21 days, we observed many alveoli and alveolar septa with few PF areas. We also observed marked and progressive increases of collagens 7, 14 and 21 days after bleomycin treatment whereas, in the PPE+B group, collagen deposition was observed only at 14 days. There was a reduction in activities of the antioxidant enzymes superoxide dismutase (p<0.05), catalase (p<0.01) and glutathione peroxidase (p<0.01) parallel with an increase in nitrite (p<0.01) 21 days after bleomycin treatment compared with the control group. These endpoints were also reduced (p<0.05, p<0.05 and p<0.01, respectively) and increased (p<0.01) in the PPE+B group at 21 days compared with the control group. Interleukin (IL)-1β expression was upregulated (p<0.01) whereas IL-6 was downregulated (p<0.05) in the PPE+B group at 21 days compared with the control group. PF and emphysema did not coexist in our model of lung disease and despite increased levels of oxidative stress and inflammatory markers after combined stimulus (elastase and bleomycin) overall histology was improved to that of the nearest control group.
Collapse
|
20
|
Checa M, Hagood JS, Velazquez-Cruz R, Ruiz V, García-De-Alba C, Rangel-Escareño C, Urrea F, Becerril C, Montaño M, García-Trejo S, Cisneros Lira J, Aquino-Gálvez A, Pardo A, Selman M. Cigarette Smoke Enhances the Expression of Profibrotic Molecules in Alveolar Epithelial Cells. PLoS One 2016; 11:e0150383. [PMID: 26934369 PMCID: PMC4775036 DOI: 10.1371/journal.pone.0150383] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Accepted: 02/12/2016] [Indexed: 12/11/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a progressive and lethal disease of unknown etiology. A growing body of evidence indicates that it may result from an aberrant activation of alveolar epithelium, which induces the expansion of the fibroblast population, their differentiation to myofibroblasts and the excessive accumulation of extracellular matrix. The mechanisms that activate the alveolar epithelium are unknown, but several studies indicate that smoking is the main environmental risk factor for the development of IPF. In this study we explored the effect of cigarette smoke on the gene expression profile and signaling pathways in alveolar epithelial cells. Lung epithelial cell line from human (A549), was exposed to cigarette smoke extract (CSE) for 1, 3, and 5 weeks at 1, 5 and 10% and gene expression was evaluated by complete transcriptome microarrays. Signaling networks were analyzed with the Ingenuity Pathway Analysis software. At 5 weeks of exposure, alveolar epithelial cells acquired a fibroblast-like phenotype. At this time, gene expression profile revealed a significant increase of more than 1000 genes and deregulation of canonical signaling pathways such as TGF-β and Wnt. Several profibrotic genes involved in EMT were over-expressed, and incomplete EMT was observed in these cells, and corroborated in mouse (MLE-12) and rat (RLE-6TN) epithelial cells. The secretion of activated TGF-β1 increased in cells exposed to cigarette smoke, which decreased when the integrin alpha v gene was silenced. These findings suggest that the exposure of alveolar epithelial cells to CSE induces the expression and release of a variety of profibrotic genes, and the activation of TGF-β1, which may explain at least partially, the increased risk of developing IPF in smokers.
Collapse
Affiliation(s)
- Marco Checa
- Instituto Nacional de Enfermedades Respiratorias, Ismael Cosío Villegas, Ciudad de México, México
| | - James S. Hagood
- Department of Pediatrics, Division of Respiratory Medicine, University of California San Diego, and Rady Children's Hospital of San Diego, San Diego, California, United States of America
| | | | - Victor Ruiz
- Instituto Nacional de Enfermedades Respiratorias, Ismael Cosío Villegas, Ciudad de México, México
| | - Carolina García-De-Alba
- Instituto Nacional de Enfermedades Respiratorias, Ismael Cosío Villegas, Ciudad de México, México
| | | | - Francisco Urrea
- Instituto Nacional de Enfermedades Respiratorias, Ismael Cosío Villegas, Ciudad de México, México
| | - Carina Becerril
- Instituto Nacional de Enfermedades Respiratorias, Ismael Cosío Villegas, Ciudad de México, México
| | - Martha Montaño
- Instituto Nacional de Enfermedades Respiratorias, Ismael Cosío Villegas, Ciudad de México, México
| | - Semiramis García-Trejo
- Instituto Nacional de Enfermedades Respiratorias, Ismael Cosío Villegas, Ciudad de México, México
| | - José Cisneros Lira
- Instituto Nacional de Enfermedades Respiratorias, Ismael Cosío Villegas, Ciudad de México, México
| | - Arnoldo Aquino-Gálvez
- Instituto Nacional de Enfermedades Respiratorias, Ismael Cosío Villegas, Ciudad de México, México
| | - Annie Pardo
- Facultad de Ciencias, Universidad Nacional Autónoma de México, Ciudad de México, México
| | - Moisés Selman
- Instituto Nacional de Enfermedades Respiratorias, Ismael Cosío Villegas, Ciudad de México, México
| |
Collapse
|
21
|
Lin H, Jiang S. Combined pulmonary fibrosis and emphysema (CPFE): an entity different from emphysema or pulmonary fibrosis alone. J Thorac Dis 2015; 7:767-79. [PMID: 25973246 DOI: 10.3978/j.issn.2072-1439.2015.04.17] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2014] [Accepted: 02/04/2015] [Indexed: 11/14/2022]
Abstract
Chronic obstructive pulmonary disease (COPD) and idiopathic interstitial pneumonias (IIP), with different radiological, pathological, functional and prognostic characteristics, have been regarded as separate entities for a long time. However, there is an increasing recognition of the coexistence of emphysema and pulmonary fibrosis in individuals. The association was first described as a syndrome by Cottin in 2005, named "combined pulmonary fibrosis and emphysema (CPFE)", which is characterized by exertional dyspnea, upper-lobe emphysema and lower-lobe fibrosis, preserved lung volume and severely diminished capacity of gas exchange. CPFE is frequently complicated by pulmonary hypertension, acute lung injury and lung cancer and prognosis of it is poor. Treatments for CPFE patients with severe pulmonary hypertension are less effective other than lung transplantation. However, CPFE has not yet attracted wide attention of clinicians and there is no research systematically contrasting the differences among CPFE, emphysema/COPD and IIP at the same time. The authors will review the existing knowledge of CPFE and compare them to either entity alone for the first time, with the purpose of improving the awareness of this syndrome and exploring novel effective therapeutic strategies in clinical practice.
Collapse
Affiliation(s)
- Huijin Lin
- 1 Department of Respiratory Medicine, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China ; 2 Sun Yat-Sen University Institute of Respiratory Disease, Guangzhou 510275, China
| | - Shanping Jiang
- 1 Department of Respiratory Medicine, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China ; 2 Sun Yat-Sen University Institute of Respiratory Disease, Guangzhou 510275, China
| |
Collapse
|
22
|
Zosky GR, Boylen CE, Wong RS, Smirk MN, Gutiérrez L, Woodward RC, Siah WS, Devine B, Maley F, Cook A. Variability and consistency in lung inflammatory responses to particles with a geogenic origin. Respirology 2014; 19:58-66. [PMID: 23796236 DOI: 10.1111/resp.12150] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2012] [Revised: 03/06/2013] [Accepted: 05/06/2013] [Indexed: 11/29/2022]
Abstract
BACKGROUND AND OBJECTIVE Particulate matter <10 μm (PM10 ) is well recognized as being an important driver of respiratory health; however, the impact of PM10 of geogenic origin on inflammatory responses in the lung is poorly understood. This study aimed to assess the lung inflammatory response to community sampled geogenic PM10 . METHODS This was achieved by collecting earth material from two regional communities in Western Australia (Kalgoorlie-Boulder and Newman), extracting the PM10 fraction and exposing mice by intranasal instillation to these particles. The physicochemical characteristics of the particles were assessed and lung inflammatory responses were compared to control particles. The primary outcomes were cellular influx and cytokine production in the lungs of the exposed mice. RESULTS The physical and chemical characteristics of the PM10 from Kalgoorlie and Newman differed with the latter having a higher concentration of Fe and a larger median diameter. Control particles (2.5 μm polystyrene) caused a significant influx of inflammatory cells (neutrophils) with little production of proinflammatory cytokines. In contrast, the geogenic particles induced the production of MIP-2, IL-6 and a significant influx of neutrophils. Qualitatively, the response following exposure to particles from Kalgoorlie and Newman were consistent; however, the magnitude of the response was substantially higher in the mice exposed to particles from Newman. CONCLUSIONS The unique physicochemical characteristics of geogenic particles induced a proinflammatory response in the lung. These data suggest that particle composition should be considered when setting community standards for PM exposure, particularly in areas exposed to high geogenic particulate loads.
Collapse
Affiliation(s)
- Graeme R Zosky
- Telethon Institute for Child Health Research, The University of Western Australia, Perth, Western Australia; Centre for Child Health Research, The University of Western Australia, Perth, Western Australia; Cooperative Research Centre for Asthma and Airways, Sydney, New South Wales, Australia
| | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
|
24
|
Papiris SA, Triantafillidou C, Manali ED, Kolilekas L, Baou K, Kagouridis K, Bouros D. Combined pulmonary fibrosis and emphysema. Expert Rev Respir Med 2013; 7:19-31; quiz 32. [PMID: 23362797 DOI: 10.1586/ers.12.80] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The advent of computed tomography permitted recognition of the coexistence of pulmonary fibrosis and emphysema (CPFE). Emphysema is usually encountered in the upper lobes preceding fibrosis of the lower lobes, and patients are smokers, predominantly male, with distinct physiologic profile characterized by preserved lung volumes and markedly reduced diffusion capacity. Actually, the term CPFE is reserved for the coexistence of any type and grade of radiological pulmonary emphysema and the idiopathic usual interstitial pneumonia computed tomography pattern as well as any pathologically confirmed case. CPFE is complicated by pulmonary hypertension, lung cancer and acute lung injury and may present different outcome than that of its components.
Collapse
Affiliation(s)
- Spyros A Papiris
- Second Pulmonary Medicine Department, Attikon University Hospital, Athens Medical School, National and Kapodistrian University of Athens, Greece.
| | | | | | | | | | | | | |
Collapse
|
25
|
Abstract
Proteases are enzymes that have the capacity to hydrolyze peptide bonds and degrade other proteins. Proteases can promote inflammation by regulating expression and activity of different pro-inflammatory cytokines, chemokines and other immune components in the lung compartment. They are categorized in three major subcategories: serine proteases, metalloproteases and cysteine proteases especially in case of lung diseases. Neutrophil-derived serine proteases (NSPs), metalloproteases and some mast cell-derived proteases are mainly focused here. Their modes of actions are different in different diseases for e.g. NE induces the release of IL-8 from lung epithelial cells through a MyD88/IRAK/TRAF-6-dependent pathway and also through EGFR MAPK pathway. NSPs contribute to immune regulation during inflammation through the cleavage and activation of specific cellular receptors. MMPs can also influence the progression of various inflammatory processes and there are many non-matrix substrates for MMPs, such as chemokines, growth factors and receptors. During lung inflammation interplay between NE and MMP is an important significant phenomenon. They have been evaluated as therapeutic targets in several inflammatory lung diseases. Here we review the role of proteases in various lung inflammatory diseases with emphasis on their mode of action and contribution to immune regulation during inflammation.
Collapse
Affiliation(s)
- Sajal Chakraborti
- Department of Biochemistry and Biophysics, University of Kalyani, Kalyani, West Bengal India
| | - Naranjan S. Dhalla
- Institute of Cardiovascular Sciences, University of Manitoba, St. Boniface Hospital Research Centre, Winnipeg, Manitoba Canada
| |
Collapse
|
26
|
Abstract
PURPOSE OF REVIEW This review discusses combined pulmonary fibrosis and emphysema (CPFE) in the setting of connective tissue disease. RECENT FINDINGS CPFE is a recently identified syndrome in smokers or ex-smokers characterized by dyspnea often severe, preserved lung volumes, severely impaired gas exchanges, and an increased risk of pulmonary hypertension associated with a dismal prognosis, and possibly lung cancer. It may be encountered in the setting of connective tissue diseases, especially rheumatoid arthritis and systemic sclerosis, with generally similar features as 'idiopathic' (tobacco-related) CPFE. The diagnosis is based on the presence of both emphysema predominating in the upper lobes and frequently paraseptal, and interstitial abnormalities suggesting pulmonary fibrosis in the lower lung zones with velcro crackles at auscultation. Pathologic radiological correlations are difficult owing to various pathology and difficulties in identifying honeycombing at chest high-resolution computed tomography in the setting of coexistent emphysema. Tobacco smoking is associated with an increased risk of developing most of the individual components of the syndrome (i.e. emphysema, pulmonary fibrosis, pulmonary hypertension, rheumatoid arthritis, and pulmonary fibrosis among patients with rheumatoid arthritis). CPFE impacts modalities of follow-up for pulmonary function and detection of pulmonary hypertension especially in systemic sclerosis. SUMMARY The syndrome of CPFE is a distinct pulmonary manifestation in the spectrum of lung diseases associated with connective tissue diseases, especially in smokers or ex-smokers.
Collapse
|
27
|
Fukushi M, Yamashita M, Miyoshi-Akiyama T, Kubo S, Yamamoto K, Kudo K. Laninamivir octanoate and artificial surfactant combination therapy significantly increases survival of mice infected with lethal influenza H1N1 Virus. PLoS One 2012; 7:e42419. [PMID: 22879974 PMCID: PMC3409853 DOI: 10.1371/journal.pone.0042419] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2012] [Accepted: 07/03/2012] [Indexed: 01/11/2023] Open
Abstract
Background Patients with influenza virus infection can develop severe pneumonia and acute respiratory distress syndrome (ARDS) which have a high mortality. Influenza virus infection is treated worldwide mainly by neuraminidase inhibitors (NAIs). However, monotherapy with NAIs is insufficient for severe pneumonia secondary to influenza virus infection. We previously demonstrated that mice infected with a lethal dose of influenza virus develop diffuse alveolar damage (DAD) with alveolar collapse similar to that seen in ARDS in humans. Additionally, pulmonary surfactant proteins were gradually increased in mouse serum, suggesting a decrease in pulmonary surfactant in the lung. Therefore, the present study examined whether combination therapy of NAI with exogenous artificial surfactant affects mortality of influenza virus-infected mice. Methodology/Principal Findings BALB/c mice were inoculated with several viral doses of influenza A/Puerto Rico/8/34 (PR8) virus (H1N1). The mice were additionally administered exogenous artificial surfactant in the presence or absence of a new NAI, laninamivir octanoate. Mouse survival, body weight and general condition were observed for up to 20 days after inoculation. Viral titer and cytokine/chemokine levels in the lungs, lung weight, pathological analysis, and blood O2 and CO2 pressures were evaluated. Infected mice treated with combination therapy of laninamivir octanoate with artificial surfactant showed a significantly higher survival rate compared with those that received laninamivir octanoate monotherapy (p = 0.003). However, virus titer, lung weight and cytokine/chemokine responses were not different between the groups. Histopathological examination, a hydrostatic lung test and blood gas analysis showed positive results in the combination therapy group. Conclusions/Significance Combination therapy of laninamivir octanoate with artificial surfactant reduces lethality in mice infected with influenza virus, and eventually suppresses DAD formation and preserves lung function. This combination could be effective for prevention of severe pneumonia secondary to influenza virus infection in humans, which is not improved by NAI monotherapy.
Collapse
Affiliation(s)
- Masaya Fukushi
- Disease Control and Prevention Center, National Center for Global Health and Medicine, Tokyo, Japan
- Deputy Director-General's Laboratory, Research Institute, National Center for Global Health and Medicine, Tokyo, Japan
- Department of Virology, Institute of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
- * E-mail:
| | - Makoto Yamashita
- Biological Research Laboratories, Daiichi Sankyo Co., Ltd., Tokyo, Japan
| | - Tohru Miyoshi-Akiyama
- Department of Infectious Diseases, Research Institute, National Center for Global Health and Medicine, Tokyo, Japan
| | - Shuku Kubo
- Biological Research Laboratories, Daiichi Sankyo Co., Ltd., Tokyo, Japan
| | - Kenji Yamamoto
- Deputy Director-General's Laboratory, Research Institute, National Center for Global Health and Medicine, Tokyo, Japan
| | - Koichiro Kudo
- Disease Control and Prevention Center, National Center for Global Health and Medicine, Tokyo, Japan
| |
Collapse
|
28
|
Jankowich MD, Rounds SIS. Combined pulmonary fibrosis and emphysema syndrome: a review. Chest 2012; 141:222-231. [PMID: 22215830 DOI: 10.1378/chest.11-1062] [Citation(s) in RCA: 204] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
There is increasing clinical, radiologic, and pathologic recognition of the coexistence of emphysema and pulmonary fibrosis in the same patient, resulting in a clinical syndrome known as combined pulmonary fibrosis and emphysema (CPFE) that is characterized by dyspnea, upper-lobe emphysema, lower-lobe fibrosis, and abnormalities of gas exchange. This syndrome frequently is complicated by pulmonary hypertension, acute lung injury, and lung cancer. The CPFE syndrome typically occurs in male smokers, and the mortality associated with this condition, especially if pulmonary hypertension is present, is significant. In this review, we explore the current state of the literature and discuss etiologic factors and clinical characteristics of the CPFE syndrome.
Collapse
Affiliation(s)
- Matthew D Jankowich
- Vascular Research Laboratory, Providence VA Medical Center, and Division of Pulmonary, Critical Care, and Sleep Medicine, Alpert Medical School of Brown University, Providence, RI.
| | - Sharon I S Rounds
- Vascular Research Laboratory, Providence VA Medical Center, and Division of Pulmonary, Critical Care, and Sleep Medicine, Alpert Medical School of Brown University, Providence, RI
| |
Collapse
|
29
|
Cardini S, Dalli J, Fineschi S, Perretti M, Lungarella G, Lucattelli M. Genetic ablation of the fpr1 gene confers protection from smoking-induced lung emphysema in mice. Am J Respir Cell Mol Biol 2012; 47:332-9. [PMID: 22461430 DOI: 10.1165/rcmb.2012-0036oc] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Cigarette smoke (CS) is the main causative factor of chronic obstructive pulmonary disease (COPD). Current research supports the concept that airway inflammation is central to the development and progression of the disease. Studies have demonstrated that neutrophils are increased in COPD lungs and that neutrophil-associated products correlate with the development and severity of COPD. The peptide FMLP is an active component of CS. FMLP interacts on the neutrophil and macrophage membranes with a high-affinity receptor subtype (FPR1) and with a low-affinity subtype FPRL1, promoting a chemotactic response, superoxide anion production, and degranulation. Bacterial colonization of the lower respiratory tract and lung cell damage may represent further sources of formyl peptides in patients with COPD. We investigated the role of FPR in a mouse model on lung inflammation and emphysema induced by CS. Here, we report the novel observation that genetic ablation of the FPR1 gene (Fpr1) confers protection from smoking-induced lung emphysema in mice. Compared with wild-type mice, Fpr1 knockout mice displayed marked decreases in the lung migration of neutrophils and macrophages after CS exposure. Upon transgenic approach, the changes in cell numbers were accompanied by marked modulation of the expression of genes implicated in the inflammatory response. Administration of the FPR1 antagonist cyclosporine H to wild-type mice attenuated the acute inflammatory response evoked by CS. These findings may have clinical significance because current smokers and subjects with emphysema showed increased FPR expression in bronchoalveolar fluids and on peripheral neutrophils. Modulating the FPR1 signal should be explored as a potential new therapy.
Collapse
Affiliation(s)
- Silvia Cardini
- Dept. Physiopathology & Experimental Medicine, University of Siena, via Aldo Moro 6, I-53100 Siena, Italy
| | | | | | | | | | | |
Collapse
|
30
|
Fukushi M, Ito T, Oka T, Kitazawa T, Miyoshi-Akiyama T, Kirikae T, Yamashita M, Kudo K. Serial histopathological examination of the lungs of mice infected with influenza A virus PR8 strain. PLoS One 2011; 6:e21207. [PMID: 21701593 PMCID: PMC3118813 DOI: 10.1371/journal.pone.0021207] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2011] [Accepted: 05/22/2011] [Indexed: 01/11/2023] Open
Abstract
Avian influenza H5N1 and pandemic (H1N1) 2009 viruses are known to induce viral pneumonia and subsequent acute respiratory distress syndrome (ARDS) with diffuse alveolar damage (DAD). The mortality rate of ARDS/DAD is extremely high, at approximately 60%, and no effective treatment for ARDS/DAD has been established. We examined serial pathological changes in the lungs of mice infected with influenza virus to determine the progress from viral pneumonia to ARDS/DAD. Mice were intranasally infected with influenza A/Puerto Rico/8/34 (PR8) virus, and their lungs were examined both macro- and micro-pathologically every 2 days. We also evaluated general condition, survival rate, body weight, viral loads in lung, and surfactant proteins in serum. As a result, all infected mice died within 9 days postinfection. At 2 days postinfection, inflammation in alveolar septa, i.e., interstitial pneumonia, was observed around bronchioles. From 4 to 6 days postinfection, interstitial pneumonia with alveolar collapse expanded throughout the lungs. From 6 to 9 days postinfection, DAD with severe alveolar collapse was observed in the lungs of all of dying and dead mice. In contrast, DAD was not observed in the live infected-mice from 2 to 6 days postinfection, despite their poor general condition. In addition, histopathological analysis was performed in mice infected with a dose of PR8 virus which was 50% of the lethal dose for mice in the 20-day observation period. DAD with alveolar collapse was observed in all dead mice. However, in the surviving mice, instead of DAD, glandular metaplasia was broadly observed in their lungs. The present study indicates that DAD with severe alveolar collapse is associated with death in this mouse infection model of influenza virus. Inhibition of the development of DAD with alveolar collapse may decrease the mortality rate in severe viral pneumonia caused by influenza virus infection.
Collapse
Affiliation(s)
- Masaya Fukushi
- Disease Control and Prevention Center, National Center for Global Health and Medicine, Tokyo, Japan.
| | | | | | | | | | | | | | | |
Collapse
|
31
|
Al-Dissi AN, Weber LP. Resveratrol preserves cardiac function, but does not prevent endothelial dysfunction or pulmonary inflammation after environmental tobacco smoke exposure. Food Chem Toxicol 2011; 49:1584-91. [PMID: 21501646 DOI: 10.1016/j.fct.2011.04.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2011] [Revised: 04/01/2011] [Accepted: 04/05/2011] [Indexed: 12/30/2022]
Abstract
The mechanisms by which environmental tobacco smoke (ETS) causes adverse cardiovascular effects remain unclear. Resveratrol is a natural polyphenol from red wine which may be beneficial to the cardiovascular system. Therefore, the ability of daily oral resveratrol (5mg/kg) to prevent adverse effects of a 14-day ETS exposure (1 h/day) on endothelial function (flow-mediated dilation), left ventricular function (echocardiography) and blood pressure (oscillometry) was assessed in juvenile male pigs (n=4 pigs/group). After a 14-day exposure to ETS, flow-mediated dilation was impaired while plasma nitrotyrosine was increased compared to sham-exposed pigs indicating impaired endothelial function. In ETS-exposed pigs, plasma C-reactive protein levels, lung cytochrome P4501A1 activity, bronchoalveolar lavage fluid total white blood cell count and leukocyte elastase activity were all significantly increased compared to sham-exposed pigs. Resveratrol treatment failed to prevent most ETS-mediated effects examined, but did increase left ventricular end-diastolic volume and ejection fraction in the presence of ETS exposure. In summary, ETS exposure impaired endothelial function and increased oxidative stress which was associated with pulmonary and systemic inflammation, but resveratrol failed to protect against these changes. More importantly, resveratrol exerted a positive effect on left ventricular function which may help explain the French paradox.
Collapse
Affiliation(s)
- Ahmad N Al-Dissi
- Toxicology Graduate Program, University of Saskatchewan, Saskatoon, Canada SK S7N 5B3
| | | |
Collapse
|
32
|
Todd NW, Jeudy J, Lavania S, Franks TJ, Galvin JR, Deepak J, Britt EJ, Atamas SP. Centrilobular emphysema combined with pulmonary fibrosis results in improved survival. FIBROGENESIS & TISSUE REPAIR 2011; 4:6. [PMID: 21324139 PMCID: PMC3055815 DOI: 10.1186/1755-1536-4-6] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/13/2010] [Accepted: 02/15/2011] [Indexed: 12/21/2022]
Abstract
Background We hypothesized that, in patients with pulmonary fibrosis combined with emphysema, clinical characteristics and outcomes may differ from patients with pulmonary fibrosis without emphysema. We identified 102 patients who met established criteria for pulmonary fibrosis. The amount of emphysema (numerical score) and type of emphysema (centrilobular, paraseptal, or mixed) were characterized in each patient. Clinical characteristics, pulmonary function tests and patient survival were analysed. Results Based on the numerical emphysema score, patients were classified into those having no emphysema (n = 48), trivial emphysema (n = 26) or advanced emphysema (n = 28). Patients with advanced emphysema had a significantly higher amount of smoking in pack/years than patients with no emphysema or trivial emphysema (P < 0.0001). Median survival [1st, 3rd quartiles] of patients with advanced emphysema was 63 [36, 82] months compared to 29 [18, 49] months in patients without emphysema and 32 [19, 48] months in patients with trivial emphysema (P < 0.001). Median forced vital capacity (FVC) and total lung capacity (TLC) were higher in the advanced emphysema group compared to patients with no emphysema (P < 0.01 and P < 0.001, respectively), whereas median DLCO did not differ among groups and was overall low. Within the advanced emphysema group (n = 28), further characterization of the type of emphysema was performed and, within these subgroups of patients, survival was 75 [58, 85] months for patients with centrilobular emphysema, 75 [48, 85] months for patients with mixed centrilobular/paraseptal emphysema, and 24 [22, 35] months for patients with paraseptal emphysema (P < 0.01). Patients with advanced paraseptal emphysema had similar survival times to patients without emphysema. Conclusions Patients with pulmonary fibrosis combined with advanced centrilobular or mixed emphysema have an improved survival compared with patients with pulmonary fibrosis without emphysema, with trivial emphysema or with advanced paraseptal emphysema.
Collapse
Affiliation(s)
- Nevins W Todd
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA.
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Cottin V, Nunes H, Mouthon L, Gamondes D, Lazor R, Hachulla E, Revel D, Valeyre D, Cordier JF. Combined pulmonary fibrosis and emphysema syndrome in connective tissue disease. ACTA ACUST UNITED AC 2011; 63:295-304. [PMID: 20936629 DOI: 10.1002/art.30077] [Citation(s) in RCA: 119] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
OBJECTIVE Connective tissue diseases (CTDs) are associated with several interstitial lung diseases. The aim of this study was to describe the recently individualized syndrome of combined pulmonary fibrosis and emphysema (CPFE) in a population of patients with CTD. METHODS In this multicenter study, we retrospectively investigated data from patients with CTD who also have CPFE. The demographic characteristics of the patients, the results of pulmonary function testing, high-resolution computed tomography, lung biopsy, and treatment, and the outcomes of the patients were analyzed. RESULTS Data from 34 patients with CTD who were followed up for a mean±SD duration of 8.3±7.0 years were analyzed. Eighteen of the patients had rheumatoid arthritis (RA), 10 had systemic sclerosis (SSc), 4 had mixed or overlap CTD, and 2 had other CTDs. The mean±SD age of the patients was 57±11 years, 23 were men, and 30 were current or former smokers. High-resolution computed tomography revealed emphysema of the upper lung zones and pulmonary fibrosis of the lower zones in all patients, and all patients exhibited dyspnea during exercise. Moderately impaired pulmonary function test results and markedly reduced carbon monoxide transfer capacity were observed. Five patients with SSc exhibited pulmonary hypertension. Four patients died during followup. Patients with CTD and CPFE were significantly younger than an historical control group of patients with idiopathic CPFE and more frequently were female. In addition, patients with CTD and CPFE had higher lung volumes, lower diffusion capacity, higher pulmonary pressures, and more frequently were male than those with CTD and lung fibrosis without emphysema. CONCLUSION CPFE warrants inclusion as a novel, distinct pulmonary manifestation within the spectrum of CTD-associated lung diseases in smokers or former smokers, especially in patients with RA or SSc.
Collapse
Affiliation(s)
- Vincent Cottin
- Hospices Civils de Lyon, Hôpital Louis Pradel, Université de Lyon, UMR754, and IFR128, Lyon, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Bergin DA, Reeves EP, Meleady P, Henry M, McElvaney OJ, Carroll TP, Condron C, Chotirmall SH, Clynes M, O'Neill SJ, McElvaney NG. α-1 Antitrypsin regulates human neutrophil chemotaxis induced by soluble immune complexes and IL-8. J Clin Invest 2010; 120:4236-50. [PMID: 21060150 DOI: 10.1172/jci41196] [Citation(s) in RCA: 216] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2009] [Accepted: 09/15/2010] [Indexed: 12/19/2022] Open
Abstract
Hereditary deficiency of the protein α-1 antitrypsin (AAT) causes a chronic lung disease in humans that is characterized by excessive mobilization of neutrophils into the lung. However, the reason for the increased neutrophil burden has not been fully elucidated. In this study we have demonstrated using human neutrophils that serum AAT coordinates both CXCR1- and soluble immune complex (sIC) receptor-mediated chemotaxis by divergent pathways. We demonstrated that glycosylated AAT can bind to IL-8 (a ligand for CXCR1) and that AAT-IL-8 complex formation prevented IL-8 interaction with CXCR1. Second, AAT modulated neutrophil chemotaxis in response to sIC by controlling membrane expression of the glycosylphosphatidylinositol-anchored (GPI-anchored) Fc receptor FcγRIIIb. This process was mediated through inhibition of ADAM-17 enzymatic activity. Neutrophils isolated from clinically stable AAT-deficient patients were characterized by low membrane expression of FcγRIIIb and increased chemotaxis in response to IL-8 and sIC. Treatment of AAT-deficient individuals with AAT augmentation therapy resulted in increased AAT binding to IL-8, increased AAT binding to the neutrophil membrane, decreased FcγRIIIb release from the neutrophil membrane, and normalization of chemotaxis. These results provide new insight into the mechanism underlying the effect of AAT augmentation therapy in the pulmonary disease associated with AAT deficiency.
Collapse
Affiliation(s)
- David A Bergin
- 1Respiratory Research Division, Department of Medicine, Royal College of Surgeons in Ireland, Education and Research Centre, Beaumont Hospital, Dublin, Ireland
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Ngan DA, Vickerman SV, Granville DJ, Man SFP, Sin DD. The possible role of granzyme B in the pathogenesis of chronic obstructive pulmonary disease. Ther Adv Respir Dis 2009; 3:113-29. [PMID: 19638369 DOI: 10.1177/1753465809341965] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a highly prevalent inflammatory lung condition characterized by airways disease and emphysema, and the precise mechanism of pathogenesis is poorly understood. The consistent features of COPD include protease-antiprotease imbalance, inflammation and accelerated aging caused by apoptosis or senescence. One family of molecules involved in all of these processes is the granzymes, serine proteases with the best-known member being granzyme B (GzmB). The majority of GzmB is released unidirectionally towards target cells, but GzmB can also be released nonspecifically and escape into the extracellular environment. GzmB is capable of cleaving extracellular matrix (ECM) proteins in vitro, and the accumulation of GzmB in the extracellular milieu during chronic inflammation in COPD could contribute to ECM degradation and remodelling and, consequently, the emphysematous phenotype in the lung. Preliminary studies suggest that increased GzmB expression is associated with increased COPD severity, and this may represent a promising new target for drug and biomarker discovery in COPD. In this paper, we review the potential pathogenic contributions of GzmB to the pathogenesis of COPD.
Collapse
Affiliation(s)
- David A Ngan
- James Hogg Research Laboratories, Providence Heart + Lung Institute at St. Paul's Hospital and Department of Medicine, University of British Columbia, Vancouver, BC, Canada
| | | | | | | | | |
Collapse
|
36
|
Vanoirbeek JAJ, Rinaldi M, De Vooght V, Haenen S, Bobic S, Gayan-Ramirez G, Hoet PHM, Verbeken E, Decramer M, Nemery B, Janssens W. Noninvasive and invasive pulmonary function in mouse models of obstructive and restrictive respiratory diseases. Am J Respir Cell Mol Biol 2009; 42:96-104. [PMID: 19346316 DOI: 10.1165/rcmb.2008-0487oc] [Citation(s) in RCA: 250] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Pulmonary function analysis is an important tool in the evaluation of mouse respiratory disease models, but much controversy still exists on the validity of some tests. Most commonly used pulmonary function variables of humans are not routinely applied in mice, and the question of which pulmonary function is optimal for the monitoring of a particular disease model remains largely unanswered. Our study aimed to delineate the potential and restrictions of existing pulmonary function techniques in different respiratory disease models, and to determine some common variables between humans and mice. A noninvasive (unrestrained plethysmography) and two invasive pulmonary function devices (forced maneuvers system from Buxco Research Systems [Wilmington, NC] and forced oscillation technique from SCIREQ [Montreal, PQ, Canada]) were evaluated in well-established models of asthma (protein and chemical induced): a model of elastase-induced pulmonary emphysema, and a model of bleomycin-induced pulmonary fibrosis. In contrast to noninvasive tests, both invasive techniques were efficacious for the quantification of parenchymal disease via changes in functional residual capacity, total lung capacity, vital capacity, and compliance of the respiratory system. Airflow obstruction and airflow limitation at baseline were only present in emphysema, but could be significantly induced after methacholine challenge in mice with asthma, which correlated best with an increase of respiratory resistance. Invasive pulmonary functions allow distinction between respiratory diseases in mice by clinically relevant variables, and should become standard in the functional evaluation of pathological disease models.
Collapse
Affiliation(s)
- Jeroen A J Vanoirbeek
- Lung Toxicology Research Unit, Katholieke Universiteit Leuven, Herestraat 49 bus 706, Leuven, Belgium.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Boutin M, Berthelette C, Gervais FG, Scholand MB, Hoidal J, Leppert MF, Bateman KP, Thibault P. High-sensitivity nanoLC-MS/MS analysis of urinary desmosine and isodesmosine. Anal Chem 2009; 81:1881-7. [PMID: 19178285 PMCID: PMC2787797 DOI: 10.1021/ac801745d] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Chronic obstructive pulmonary disease (COPD) is characterized by the degradation of elastin, the major insoluble protein of lung tissues. The degradation of elastin gives rise to desmosine (DES) and isodesmosine (IDES), two major urinary products typified by a hydrophilic pyridinium-based cross-linker structure. A high sensitivity method based on nanoflow liquid chromatography tandem mass spectrometry with multiple reaction monitoring was developed for the analysis of urinary DES and IDES. The analytes were derivatized with propionic anhydride and deuterated DES (D(4)-DES) was used as an internal standard. This method enables the quantification of DES and IDES in as little as 50 microL of urine and provides a detection limit of 0.10 ng/mL (0.95 fmol on-column). We report the analysis of DES and IDES in a cohort of 40 urine specimens from four groups of individuals: (a) COPD rapid decliners (11.8 +/- 3.7 ng/mg creatine (crea)), (b) COPD slow decliners (16.0 +/- 3.1 ng/mg crea), (c) healthy smokers (13.2 +/- 1.9 ng/mg crea), and (d) healthy nonsmokers (14.9 +/- 2.9 ng/mg crea). Our analysis reveals a statistically significant decrease in the level of urinary DES and IDES in COPD rapid decliner patients compared to healthy nonsmoker controls and COPD slow decliner patients. This methodology may be useful for monitoring DES and IDES levels in well controlled animal models for COPD or for longitudinal studies in COPD patients.
Collapse
Affiliation(s)
- Michel Boutin
- Institute for Research in Immunology and Cancer and Department of Chemistry, Université de Montréal, P.O. Box 6128, Station Centre-Ville Montréal QC H3C 3J7, Canada
| | | | | | - Mary-Beth Scholand
- Department of Internal Medicine, University of Utah, Salt Lake City, Utah 84132
| | - John Hoidal
- Department of Internal Medicine, University of Utah, Salt Lake City, Utah 84132
| | - Mark F. Leppert
- Department of Human Genetics, University of Utah, Salt Lake City, Utah 84112
| | | | - Pierre Thibault
- Institute for Research in Immunology and Cancer and Department of Chemistry, Université de Montréal, P.O. Box 6128, Station Centre-Ville Montréal QC H3C 3J7, Canada
- Université de Montréal, P.O. Box 6128, Station Centre-Ville Montréal QC H3C 3J7, Canada
| |
Collapse
|
38
|
HRCT and histopathological evaluation of fibrosis and tissue destruction in IPF associated with pulmonary emphysema. Respir Med 2008; 102:1753-61. [PMID: 18723334 DOI: 10.1016/j.rmed.2008.07.010] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2008] [Accepted: 07/02/2008] [Indexed: 02/06/2023]
Abstract
Idiopathic pulmonary fibrosis has been associated with emphysema in cigarette smokers as a new clinical entity: combined pulmonary fibrosis and emphysema (CPFE). In order to compare histomorphometrical, roentgenological and immunohistochemical aspects of usual interstitial pneumonia (UIP) with and without associated pulmonary emphysema, 17 patients with biopsy-proven UIP were evaluated. Morphometrical evaluation of lung parenchyma destruction was used to divide patients in two subgroups: emphysema/UIP (n=9) and UIP alone (n=8); four patients with biopsy-proven emphysema without fibrosis were also evaluated. At HRTC scan, emphysematous lesions were prevalent in the upper fields of both emphysema/UIP and emphysema groups and the distribution of fibrotic lesions was similar in emphysema/UIP compared to UIP alone. The semiquantitative histopathological fibrotic score was also similar in emphysema/UIP and UIP alone. In addition, the expression of tumor necrosis factor (TNF)-alpha, matrix metalloproteinase (MMP)-2, MMP-9, MMP-7 and membrane type 1-metalloproteinase (MT1-MMP) by fibroblasts of myofibroblastic foci was similar in emphysema/UIP and UIP alone patients. In contrast, fibroblasts in areas of parenchymal destruction of emphysema/UIP expressed MMP-2, MMP-9, MMP-7 and MT1-MMP at variable but significantly higher levels when compared to emphysema subjects, in the presence of similar levels of TIMP-1, TIMP-2 and TNF-alpha. Fibrotic and emphysematous lesions in emphysema/UIP patients appear to follow the roentgenological and histopathological patterns expected for either UIP or emphysema. Interstitial fibroblast activation is more pronounced in the areas of lung destruction in emphysema/UIP compared to those with emphysema alone, as for exaggerated tissue remodeling.
Collapse
|
39
|
de Ryk J, Thiesse J, Namati E, McLennan G. Stress distribution in a three dimensional, geometric alveolar sac under normal and emphysematous conditions. Int J Chron Obstruct Pulmon Dis 2008; 2:81-91. [PMID: 18044070 PMCID: PMC2692121 DOI: 10.2147/copd.2007.2.1.81] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2022] Open
Abstract
Pulmonary emphysema is usually the result of chronic exposure to cigarette smoke in at risk individuals. To investigate the hypothesis that lung damage in emphysema results from coincident weakening in the structural properties of the tissue and increased mechanical forces—as one explanation of the continued development of pulmonary emphysema after smoking cessation—we developed a three dimensional, geometric dodecahedron-based acinar model. Using the model numerical analysis of the stress distribution in normal conditions could be compared with those resulting in emphysematous conditions. Finite element analysis was used to evaluate the model at a number of lung inflation levels, using quasi-static loading of the alveolar pressure. When internal alveolar pressure was increased along with the adjustment of the material properties to represent a weakening of one wall in the acinus, increased stress resulted at the perimeters of the weakened area. In particular this increased stress was localized at the junction points of the internal alveolar septa. It was also found that under the proposed simulated emphysematous conditions, a significant disruption in the stress distribution within the acinus model occurred at low, rather than high, lung volumes. This is supportive of the physiological observation that destruction of the diseased tissue can occur under less stress than those existing in the normal state.
Collapse
Affiliation(s)
- Jessica de Ryk
- Department of Internal Medicine, University of Iowa Hospitals and Clinics, Iowa City, Iowa, USA
| | | | | | | |
Collapse
|
40
|
Yang S, Nugent MA, Panchenko MP. EGF antagonizes TGF-beta-induced tropoelastin expression in lung fibroblasts via stabilization of Smad corepressor TGIF. Am J Physiol Lung Cell Mol Physiol 2008; 295:L143-51. [PMID: 18441095 DOI: 10.1152/ajplung.00289.2007] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
We previously reported that neutrophil elastase (NE) downregulates transforming growth factor-beta (TGF-beta)-maintained tropoelastin mRNA levels in lung fibroblasts through transactivation of the epidermal growth factor (EGF) receptor (EGFR)/Mek/Erk pathway, which is dependent on the NE-initiated release of soluble EGFR ligands. In the present study, we investigated the mechanism by which EGF downregulates tropoelastin expression. We found that EGF downregulates tropoelastin expression through inhibition of TGF-beta signaling. We show that EGF does not prevent the TGF-beta-induced nuclear accumulation of Smad2/3; rather, EGF stabilizes the short-lived Smad transcriptional corepressor TG-interacting factor (TGIF) via EGFR/Mek/Erk-mediated phosphorylation of TGIF. Elevation of TGIF levels, either by TGIF overexpression or prevention of TGIF degradation, is sufficient to inhibit TGF-beta-induced tropoelastin expression. Moreover, TGIF is essential for EGF-mediated downregulation of tropoelastin expression, inasmuch as small interfering RNA knockdown of TGIF blocked EGF-induced downregulation of tropoelastin. Finally, we demonstrated that NE treatment, which releases EGF-like growth factors, causes stabilization of TGIF through the EGFR/Mek/Erk pathway. These results suggest that EGFR/Mek/Erk signaling specifically antagonizes the proelastogenic action of TGF-beta in lung fibroblasts by stabilizing the Smad transcriptional corepressor TGIF.
Collapse
Affiliation(s)
- Shenghong Yang
- Department of Biochemistry, Boston University School of Medicine, 715 Albany St, Boston, Massachusetts 02118, USA
| | | | | |
Collapse
|
41
|
Bizeto L, Mazzolini AB, Ribeiro M, Stelmach R, Cukier A, Nunes MPT. Interrelationship between serum and sputum inflammatory mediators in chronic obstructive pulmonary disease. ACTA ACUST UNITED AC 2008; 41:193-8. [PMID: 18327434 DOI: 10.1590/s0100-879x2008005000010] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2007] [Accepted: 11/19/2007] [Indexed: 12/23/2022]
Abstract
Little is known about airway inflammatory markers in chronic obstructive pulmonary disease (COPD). The objective of the present study was to identify and try to correlate pulmonary and peripheral blood inflammatory markers in COPD. In a cross-sectional study on patients with stable COPD, induced sputum and blood samples were collected for the determination of C-reactive protein, eosinophilic cationic protein, serum amyloid A protein, alpha-1 antitrypsin (alpha-1AT), and neutrophil elastase. Twenty-two patients were divided into two groups according to post-bronchodilator forced expiratory volume in the first second (%FEV1): group 1 (N = 12, FEV1 <40%) and group 2 (N = 10, FEV1 > or =40%). An increase in serum elastase, eosinophilic cationic protein and alpha-1AT was observed in serum markers in both groups. Cytology revealed the same total number of cells in groups 1 and 2. There was a significantly higher number of neutrophils in group 1 compared to group 2 (P < 0.05). No difference in eosinophils or macrophages was observed between groups. Serum elastase was positively correlated with serum alpha-1AT (group 1, r = 0.81, P < 0.002 and group 2, r = 0.83, P < 0.17) and negatively correlated with FEV1 (r = -0.85, P < 0.03 and -0.14, P < 0.85, respectively). The results indicate the presence of chronic and persistent pulmonary inflammation in stable patients with COPD. Induced sputum permitted the demonstration of the existence of a subpopulation of cells in which neutrophils predominated. The serum concentration of all inflammatory markers did not correlate with the pulmonary functional impairment.
Collapse
Affiliation(s)
- L Bizeto
- Departamento de Clínica Médica, Faculdade de Medicina, Universidade de São Paulo, Brasil
| | | | | | | | | | | |
Collapse
|
42
|
Pottier N, Chupin C, Defamie V, Cardinaud B, Sutherland R, Rios G, Gauthier F, Wolters PJ, Berthiaume Y, Barbry P, Mari B. Relationships between early inflammatory response to bleomycin and sensitivity to lung fibrosis: a role for dipeptidyl-peptidase I and tissue inhibitor of metalloproteinase-3? Am J Respir Crit Care Med 2007; 176:1098-107. [PMID: 17673693 DOI: 10.1164/rccm.200607-1051oc] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
RATIONALE Different sensitivities to profibrotic compounds such as bleomycin are observed among mouse strains. OBJECTIVES To identify genetic factors contributing to the outcome of lung injury. METHODS Physiological comparison of C57BL/6 (sensitive) and BALB/c (resistant) mice challenged by intratracheal bleomycin instillation revealed several early differences: global gene expression profiles were thus established from lungs derived from the two strains, in the absence of any bleomycin administration. MEASUREMENTS AND MAIN RESULTS Expression of 25 genes differed between the two strains. Among them, two molecules, not previously associated with pulmonary fibrosis, were identified. The first corresponded to dipeptidyl-peptidase I (DPPI), a cysteine peptidase (also known as cathepsin C) essential for the activation of serine proteinases produced by immune/inflammatory cells. The second corresponded to tissue inhibitor of matrix metalloproteinase-3, which also inhibits members of the ADAM (a disintegrin and metalloproteinase) family, such as the tumor necrosis factor-converting enzyme. In functional studies performed in the bleomycin-induced lung fibrosis model, the level of expression of these two genes was closely correlated with specific early events associated with lung fibrosis, namely activation of polymorphonuclear neutrophil-derived serine proteases and tumor necrosis factor-alpha-dependent inflammatory syndrome. Surprisingly, genetic deletion of DPPI in the context of a C57BL/6 genetic background did not protect against bleomycin-mediated fibrosis, suggesting additional function(s) for this key enzyme. CONCLUSIONS This study highlights the importance of the early inflammatory events that follow bleomycin instillation in the development of lung fibrosis, and describes for the first time the roles that DPPI and tissue inhibitor of matrix metalloproteinase-3 may play in this process.
Collapse
Affiliation(s)
- Nicolas Pottier
- IPMC, CNRS UMR6097, 660, route des Lucioles, F06560 Sophia Antipolis, France
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Khan R. Examining potential therapies targeting myocardial fibrosis through the inhibition of transforming growth factor-beta 1. Cardiology 2007; 108:368-80. [PMID: 17308385 DOI: 10.1159/000099111] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2005] [Accepted: 10/20/2006] [Indexed: 01/25/2023]
Abstract
After injury, the heart undergoes a remodeling process consisting primarily of myocyte hypertrophy, apoptosis and interstitial fibrosis. Although initially beneficial, excess fibrosis gradually results in alteration of left ventricular properties and cardiac dysfunction. Transforming growth factor-beta 1 (TGF-beta(1)) is thought to be a primary mediator of fibrosis within the heart after injury. Currently, angiotensin II blockade is used to inhibit the actions of TGF-beta(1). However, recent studies indicate that angiotensin II blockade alone may not be sufficient to prevent TGF-beta(1)-induced fibrosis. Thus far, both in vivo and in vitro models have shown that direct TGF-beta(1) inhibition, NAPDH oxidase inhibitors, growth factors and hormonal treatment regimens targeting TGF-beta(1) may significantly reduce cardiac fibrosis after injury. This study attempts to underline these alternatives to angiotensin II blockade in combating TGF-beta(1)-induced cardiac dysfunction.
Collapse
Affiliation(s)
- Razi Khan
- McGill University, Faculty of Medicine, Montreal, Canada.
| |
Collapse
|
44
|
Gauldie J, Kolb M, Ask K, Martin G, Bonniaud P, Warburton D. Smad3 signaling involved in pulmonary fibrosis and emphysema. Ann Am Thorac Soc 2007; 3:696-702. [PMID: 17065376 PMCID: PMC2647655 DOI: 10.1513/pats.200605-125sf] [Citation(s) in RCA: 101] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The incidence of finding evidence of both emphysema and pulmonary fibrosis in the same patient has received increased attention. Several investigators have found on biopsy the presence of emphysema of the upper zones and diffuse parenchymal disease with fibrosis of the lower zones of the lung, especially associated with current or previous heavy smokers. Believed previously to be two different disease mechanisms, there are now data to implicate some common pathways of cell and molecular activation leading to the different morphologic and physiologic outcomes. According to a current view, emphysema may originate from a protease/antiprotease imbalance, whereas a role for antiproteases has been proposed in the modulation of fibrosis. Overexpression of transforming growth factor beta (TGF-beta) in experimental rodent models leads to progressive pulmonary fibrosis, accompanied with marked up-regulation of protease inhibitors, such as tissue inhibitor of metalloproteinases (TIMP) and plasminogen activator inhibitor-1 (PAI-1) genes, along with excessive matrix accumulation. It may be that a "matrix degrading" pulmonary microenvironment, one in which metalloproteinase activities prevail, favors the development of emphysema, whereas a "matrix nondegrading" microenvironment, with enhanced presence of TIMPs, would lead to matrix accumulation and fibrosis. Surprisingly, although Smad3 null mice, deficient in TGF-beta signal transmission, are resistant to bleomycin- and TGF-beta-mediated fibrosis, they develop spontaneous age-related airspace enlargement, consistent with emphysema, with a lack of ability to repair tissue damage appropriately. A common element is tissue damage and repair, with TGF-beta and the Smad signaling pathway playing prominent molecular roles. Both changes can be followed in experimental models with noninvasive imaging and physiologic measurements.
Collapse
Affiliation(s)
- Jack Gauldie
- Department of Pathology and Molecular Medicine, McMaster University, 1200 Main Street, W-MDCL-4016, Hamilton, ON, Canada L8N 3Z5.
| | | | | | | | | | | |
Collapse
|
45
|
|
46
|
DiCamillo SJ, Yang S, Panchenko MV, Toselli PA, Naggar EF, Rich CB, Stone PJ, Nugent MA, Panchenko MP. Neutrophil elastase-initiated EGFR/MEK/ERK signaling counteracts stabilizing effect of autocrine TGF-beta on tropoelastin mRNA in lung fibroblasts. Am J Physiol Lung Cell Mol Physiol 2006; 291:L232-43. [PMID: 16473861 DOI: 10.1152/ajplung.00530.2005] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Neutrophil elastase (NE) plays an important role in emphysema, a pulmonary disease associated with excessive elastolysis and ineffective repair of interstitial elastin. Besides its direct elastolytic activity, NE releases soluble epidermal growth factor receptor (EGFR) ligands and initiates EGFR/MEK/ERK signaling to downregulate tropoelastin mRNA in neonatal rat lung fibroblasts (DiCamillo SJ, Carreras I, Panchenko MV, Stone PJ, Nugent MA, Foster JA, and Panchenko MP. J Biol Chem 277: 18938-18946, 2002). We now report that NE downregulates tropoelastin mRNA in the rat fetal lung fibroblast line RFL-6. The tropoelastin mRNA downregulation is preceded by release of EGF-like and TGF-alpha-like polypeptides and requires EGFR/MEK/ERK signaling, because it is prevented by the EGFR inhibitor AG1478 and the MEK/ERK uncoupler U0126. Tropoelastin expression in RFL-6 fibroblasts is governed by autocrine TGF-beta signaling, because TGF-beta type I receptor kinase inhibitor or TGF-beta neutralizing antibody dramatically decreases tropoelastin mRNA and protein levels. Half-life of tropoelastin mRNA in RFL-6 cells is >24 h, but it is decreased to approximately 8 h by addition of TGF-beta neutralizing antibody, EGF, TGF-alpha, or NE. Tropoelastin mRNA destabilization by NE, EGF, or TGF-alpha is abolished by AG1478 or U0126. EGF-dependent tropoelastin mRNA downregulation is reversed upon ligand withdrawal, whereas chronic EGF treatment leads to persistent downregulation of tropoelastin mRNA and protein levels and decreases insoluble elastin deposition. We conclude that NE-initiated EGFR/MEK/ERK signaling cascade overrides the autocrine TGF-beta signaling on tropoelastin mRNA stability and, therefore, decreases the elastogenic response in RFL-6 fibroblasts. We hypothesize that persistent EGFR/MEK/ERK signaling could impede the TGF-beta-induced elastogenesis/elastin repair in the chronically inflamed, elastase/anti-elastase imbalanced lung in emphysema.
Collapse
Affiliation(s)
- Sandra J DiCamillo
- Department of Biochemistry, Boston University School of Medicine, MA 02118, USA
| | | | | | | | | | | | | | | | | |
Collapse
|