1
|
Chang-Chien J, Huang JL, Tsai HJ, Wang SL, Kuo ML, Yao TC. Vitamin D ameliorates particulate matter induced mitochondrial damages and calcium dyshomeostasis in BEAS-2B human bronchial epithelial cells. Respir Res 2024; 25:321. [PMID: 39174953 PMCID: PMC11342659 DOI: 10.1186/s12931-024-02951-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Accepted: 08/12/2024] [Indexed: 08/24/2024] Open
Abstract
BACKGROUND Mitochondria is prone to oxidative damage by endogenous and exogenous sources of free radicals, including particulate matter (PM). Given the role of mitochondria in inflammatory disorders, such as asthma and chronic obstructive pulmonary disease, we hypothesized that supplementation of vitamin D may play a protective role in PM-induced mitochondrial oxidative damages of human bronchial epithelial BEAS-2B cells. METHODS BEAS-2B cells were pretreated with 1,25(OH)2D3, an active form of vitamin D, for 1 h prior to 24-hour exposure to PM (SRM-1648a). Oxidative stress was measured by flow cytometry. Mitochondrial functions including mitochondrial membrane potential, ATP levels, and mitochondrial DNA copy number were analyzed. Additionally, mitochondrial ultrastructure was examined using transmission electron microscopy. Intracellular and mitochondrial calcium concentration changes were assessed using flow cytometry based on the expression of Fluo-4 AM and Rhod-2 AM, respectively. Pro-inflammatory cytokines, including IL-6 and MCP-1, were quantified using ELISA. The expression levels of antioxidants, including SOD1, SOD2, CAT, GSH, and NADPH, were determined. RESULTS Our findings first showed that 24-hour exposure to PM led to the overproduction of reactive oxygen species (ROS) derived from mitochondria. PM-induced mitochondrial oxidation resulted in intracellular calcium accumulation, particularly within mitochondria, and alterations in mitochondrial morphology and functions. These changes included loss of mitochondrial membrane integrity, disarrayed cristae, mitochondrial membrane depolarization, reduced ATP production, and increased mitochondrial DNA copy number. Consequently, PM-induced mitochondrial damage triggered the release of certain inflammatory cytokines, such as IL-6 and MCP-1. Similar to the actions of mitochondrial ROS inhibitor MitoTEMPO, 1,25(OH)2D3 conferred protective effects on mtDNA alterations, mitochondrial damages, calcium dyshomeostasis, thereby decreasing the release of certain inflammatory cytokines. We found that greater cellular level of 1,25(OH)2D3 upregulated the expression of enzymatic (SOD1, SOD2, and CAT) and non-enzymatic (GSH and NADPH) antioxidants to modulate cellular redox homeostasis. CONCLUSION Our study provides new evidence that 1,25(OH)2D3 acts as an antioxidant, enhancing BEAS-2B antioxidant responses to regulate mitochondrial ROS homeostasis and mitochondrial function, thereby enhancing epithelial defense against air pollution exposure.
Collapse
Affiliation(s)
- Ju Chang-Chien
- Division of Allergy, Asthma, and Rheumatology, Department of Pediatrics, Chang Gung Memorial Hospital, 5 Fu-Hsin Street, Kweishan, Taoyuan, 33305, Taiwan
- School of Medicine, Chang Gung University College of Medicine, Taoyuan, Taiwan
| | - Jing-Long Huang
- School of Medicine, Chang Gung University College of Medicine, Taoyuan, Taiwan
- Department of Pediatrics, New Taipei Municipal TuCheng Hospital, New Taipei, Taiwan
- Community Medicine Research Center, Chang Gung Memorial Hospital at Keelung, Keelung, Taiwan
| | - Hui-Ju Tsai
- Institute of Population Health Sciences, National Health Research Institutes, Zhunan, Taiwan
- College of Life Science, National Tsing-Hua University, Hsinchu, Taiwan
| | - Shih-Ling Wang
- Division of Allergy, Asthma, and Rheumatology, Department of Pediatrics, Chang Gung Memorial Hospital, 5 Fu-Hsin Street, Kweishan, Taoyuan, 33305, Taiwan
| | - Ming-Ling Kuo
- Division of Allergy, Asthma, and Rheumatology, Department of Pediatrics, Chang Gung Memorial Hospital, 5 Fu-Hsin Street, Kweishan, Taoyuan, 33305, Taiwan.
- Department of Pediatrics, New Taipei Municipal TuCheng Hospital, New Taipei, Taiwan.
- Department of Microbiology and Immunology, Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, 259 Wenhua 1st Road, Kweishan, Taoyuan, 33302, Taiwan.
| | - Tsung-Chieh Yao
- Division of Allergy, Asthma, and Rheumatology, Department of Pediatrics, Chang Gung Memorial Hospital, 5 Fu-Hsin Street, Kweishan, Taoyuan, 33305, Taiwan.
- School of Medicine, Chang Gung University College of Medicine, Taoyuan, Taiwan.
- Community Medicine Research Center, Chang Gung Memorial Hospital at Keelung, Keelung, Taiwan.
| |
Collapse
|
2
|
Liu T, Hecker J, Liu S, Rui X, Boyer N, Wang J, Yu Y, Zhang Y, Mou H, Gomez-Escobar LG, Choi AM, Raby BA, Weiss ST, Zhou X. The Asthma Risk Gene, GSDMB, Promotes Mitochondrial DNA-induced ISGs Expression. JOURNAL OF RESPIRATORY BIOLOGY AND TRANSLATIONAL MEDICINE 2024; 1:10005. [PMID: 38737375 PMCID: PMC11086750 DOI: 10.35534/jrbtm.2024.10005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/14/2024]
Abstract
Released mitochondrial DNA (mtDNA) in cells activates cGAS-STING pathway, which induces expression of interferon-stimulated genes (ISGs) and thereby promotes inflammation, as frequently seen in asthmatic airways. However, whether the genetic determinant, Gasdermin B (GSDMB), the most replicated asthma risk gene, regulates this pathway remains unknown. We set out to determine whether and how GSDMB regulates mtDNA-activated cGAS-STING pathway and subsequent ISGs induction in human airway epithelial cells. Using qPCR, ELISA, native polyacrylamide gel electrophoresis, co-immunoprecipitation and immunofluorescence assays, we evaluated the regulation of GSDMB on cGAS-STING pathway in both BEAS-2B cells and primary normal human bronchial epithelial cells (nHBEs). mtDNA was extracted in plasma samples from human asthmatics and the correlation between mtDNA levels and eosinophil counts was analyzed. GSDMB is significantly associated with RANTES expression in asthmatic nasal epithelial brushing samples from the Genes-environments and Admixture in Latino Americans (GALA) II study. Over-expression of GSDMB promotes DNA-induced IFN and ISGs expression in bronchial epithelial BEAS-2B cells and nHBEs. Conversely, knockout of GSDMB led to weakened induction of interferon (IFNs) and ISGs in BEAS-2B cells. Mechanistically, GSDMB interacts with the C-terminus of STING, promoting the translocation of STING to Golgi, leading to the phosphorylation of IRF3 and induction of IFNs and ISGs. mtDNA copy number in serum from asthmatics was significantly correlated with blood eosinophil counts especially in male subjects. GSDMB promotes the activation of mtDNA and poly (dA:dT)-induced activation of cGAS-STING pathway in airway epithelial cells, leading to enhanced induction of ISGs.
Collapse
Affiliation(s)
- Tao Liu
- Channing Division of Network Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Julian Hecker
- Channing Division of Network Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Siqi Liu
- Channing Division of Network Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Xianliang Rui
- Channing Division of Network Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Nathan Boyer
- Channing Division of Network Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Jennifer Wang
- Channing Division of Network Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Yuzhen Yu
- Channing Division of Network Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Yihan Zhang
- The Mucosal Immunology and Biology Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Hongmei Mou
- The Mucosal Immunology and Biology Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02115, USA
| | | | - Augustine M.K. Choi
- Weil Cornell Medical School, Joan and Sanford I. Weill Department of Medicine, New York, NY 10065, USA
| | - Benjamin A. Raby
- Division of Pulmonary Medicine, Department of Pediatrics, Boston Children’s Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Scott T. Weiss
- Channing Division of Network Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Xiaobo Zhou
- Channing Division of Network Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
3
|
Miller RL, Rivera J, Lichtiger L, Govindarajulu US, Jung KH, Lovinsky-Desir S, Perera F, Balcer Whaley S, Newman M, Grant TL, McCormack M, Perzanowski M, Matsui EC. Associations between mitochondrial biomarkers, urban residential exposures and childhood asthma outcomes over 6 months. ENVIRONMENTAL RESEARCH 2023; 239:117342. [PMID: 37813137 PMCID: PMC10843300 DOI: 10.1016/j.envres.2023.117342] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 09/26/2023] [Accepted: 10/07/2023] [Indexed: 10/11/2023]
Abstract
Determining biomarkers of responses to environmental exposures and evaluating whether they predict respiratory outcomes may help optimize environmental and medical approaches to childhood asthma. Relative mitochondrial (mt) DNA abundance and other potential mitochondrial indicators of oxidative stress may provide a sensitive metric of the child's shifting molecular responses to its changing environment. We leveraged two urban childhood cohorts (Environmental Control as Add-on Therapy in Childhood Asthma (ECATCh); Columbia Center for Children's Environmental Health (CCCEH)) to ascertain whether biomarkers in buccal mtDNA associate with airway inflammation and altered lung function over 6 months of time and capture biologic responses to multiple external stressors such as indoor allergens and fine particulate matter (PM2.5). Relative mtDNA content was amplified by qPCR and methylation of transfer RNA phenylalanine/rRNA 12S (TF/RNR1), cytochrome c oxidase (CO1), and carboxypeptidase O (CPO) was measured by pyrosequencing. Data on residential exposures and respiratory outcomes were harmonized between the two cohorts. Repeated measures and multiple regression models were utilized to assess relationships between mitochondrial biomarkers, respiratory outcomes, and residential exposures (PM2.5, allergens), adjusted for potential confounders and time-varying asthma. We found across the 6 month visits, a 0.64 fold higher level of TF/RNR1 methylation was detected among those with asthma in comparison to those without asthma ((parameter estimate (PE) 0.64, standard error 0.28, p = 0.03). In prospective analyses, CPO methylation was associated with subsequent reduced forced vital capacity (FVC; PE -0.03, standard error 0.01, p = 0.02). Bedroom dust mouse allergen, but not indoor PM2.5, was associated with higher methylation of TF/RNR1 (PE 0.015, standard error 0.006, p = 0.01). Select mtDNA measures in buccal cells may indicate children's responses to toxic environmental exposures and associate selectively with asthma and lung function.
Collapse
Affiliation(s)
- Rachel L Miller
- Division of Clinical Immunology, Department of Medicine, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA; Columbia Center for Childrens Environmental Health, Columbia University Mailman School of Public Health, 722 West 168th Street, New York, NY, 10032, USA.
| | - Janelle Rivera
- Division of Clinical Immunology, Department of Medicine, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
| | - Lydia Lichtiger
- Division of Clinical Immunology, Department of Medicine, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
| | - Usha S Govindarajulu
- Center for Biostatistics, Department of Population Health Science and Policy, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
| | - Kyung Hwa Jung
- Division of Pediatric Pulmonary, Columbia University Irving Medical Center, 630 W. 168th St, New York, NY, 10032, USA
| | - Stephanie Lovinsky-Desir
- Division of Pediatric Pulmonary, Columbia University Irving Medical Center, 630 W. 168th St, New York, NY, 10032, USA
| | - Frederica Perera
- Columbia Center for Childrens Environmental Health, Columbia University Mailman School of Public Health, 722 West 168th Street, New York, NY, 10032, USA
| | - Susan Balcer Whaley
- Department of Population Health, Dell Medical School University of Texas at Austin, 1601 Trinity St., Bldg. B, Stop Z0500, Austin, TX, 78712, USA
| | - Michelle Newman
- Department of Epidemiology and Public Health, University of Maryland, 10 S. Pine St, MSTF 3-34, Baltimore, MD, 21201, USA
| | - Torie L Grant
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Meredith McCormack
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Matthew Perzanowski
- Columbia Center for Childrens Environmental Health, Columbia University Mailman School of Public Health, 722 West 168th Street, New York, NY, 10032, USA
| | - Elizabeth C Matsui
- Department of Population Health, Dell Medical School University of Texas at Austin, 1601 Trinity St., Bldg. B, Stop Z0500, Austin, TX, 78712, USA
| |
Collapse
|
4
|
Song MA, Kim JY, Gorr MW, Miller RA, Karpurapu M, Nguyen J, Patel D, Archer KJ, Pabla N, Shields PG, Wold LE, Christman JW, Chung S. Sex-specific lung inflammation and mitochondrial damage in a model of electronic cigarette exposure in asthma. Am J Physiol Lung Cell Mol Physiol 2023; 325:L568-L579. [PMID: 37697923 PMCID: PMC11068405 DOI: 10.1152/ajplung.00033.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 07/14/2023] [Accepted: 08/30/2023] [Indexed: 09/13/2023] Open
Abstract
The prevalence of electronic cigarette (EC) use among adult with asthma has continued to increase over time, in part due to the belief of being less harmful than smoking. However, the extent of their toxicity and the involved mechanisms contributing to the deleterious impact of EC exposure on patients with preexisting asthma have not been delineated. In the present project, we tested the hypothesis that EC use contributes to respiratory damage and worsening inflammation in the lungs of patients with asthma. To define the consequences of EC exposure in established asthma, we used a mouse model with/without preexisting asthma for short-term exposure to EC aerosols. C57/BL6J mice were sensitized and challenged with a DRA (dust mite, ragweed, Aspergillus fumigates, 200 µg/mL) mixture and exposed daily to EC with nicotine (2% nicotine in 30:70 propylene glycol: vegetable glycerin) or filtered air for 2 wk. The mice were evaluated at 24 h after the final EC exposure. After EC exposure in asthmatic mice, lung inflammatory cell infiltration and goblet cell hyperplasia were increased, whereas EC alone did not cause airway inflammation. Our data also show that mitochondrial DNA (mtDNA) content and a key mtDNA regulator, mitochondrial transcription factor A (TFAM), are reduced in asthmatic EC-exposed mice in a sex-dependent manner. Together, these results indicate that TFAM loss in lung epithelium following EC contributes to male-predominant sex pathological differences, including mitochondrial damage, inflammation, and remodeling in asthmatic airways.NEW & NOTEWORTHY Respiratory immunity is dysregulated in preexisting asthma, and further perturbations by EC use could exacerbate asthma severity. However, the extent of their toxicity and the involved mechanisms contributing to the deleterious impact of EC exposure on patients with preexisting asthma have not been delineated. We found that EC has unique biological impacts in lungs and potential sex differences with loss of TFAM, a key mtDNA regulator, in lung epithelial region from our animal EC study.
Collapse
Affiliation(s)
- Min-Ae Song
- Division of Environmental Health Sciences, College of Public Health, The Ohio State University, Columbus, Ohio, United States
| | - Ji Young Kim
- Division of Pharmaceutics and Pharmacology, College of Pharmacy and Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, United States
| | - Matthew W Gorr
- Division of Cardiac Surgery, Department of Surgery, College of Medicine, The Ohio State University, Columbus, Ohio, United States
| | - Roy A Miller
- Division of Cardiac Surgery, Department of Surgery, College of Medicine, The Ohio State University, Columbus, Ohio, United States
| | - Manjula Karpurapu
- Division of Pulmonary, Critical Care and Sleep Medicine, Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio, United States
| | - Jackie Nguyen
- Division of Pulmonary, Critical Care and Sleep Medicine, Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio, United States
| | - Devki Patel
- Division of Environmental Health Sciences, College of Public Health, The Ohio State University, Columbus, Ohio, United States
| | - Kellie J Archer
- Division of Biostatistics, College of Public Health, The Ohio State University, Columbus, Ohio, United States
| | - Navjot Pabla
- Division of Pharmaceutics and Pharmacology, College of Pharmacy and Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, United States
| | - Peter G Shields
- Comprehensive Cancer Center, The Ohio State University and James Cancer Hospital, Columbus, Ohio, United States
| | - Loren E Wold
- Division of Cardiac Surgery, Department of Surgery, College of Medicine, The Ohio State University, Columbus, Ohio, United States
| | - John W Christman
- Division of Pulmonary, Critical Care and Sleep Medicine, Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio, United States
| | - Sangwoon Chung
- Division of Pulmonary, Critical Care and Sleep Medicine, Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio, United States
| |
Collapse
|
5
|
He X, Sun Y, Lu J, Naz F, Ma S, Liu J. Cytoplasmic DNAs: Sources, sensing, and roles in the development of lung inflammatory diseases and cancer. Front Immunol 2023; 14:1117760. [PMID: 37122745 PMCID: PMC10130589 DOI: 10.3389/fimmu.2023.1117760] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 03/29/2023] [Indexed: 05/02/2023] Open
Abstract
Cytoplasmic DNA is emerging as a pivotal contributor to the pathogenesis of inflammatory diseases and cancer, such as COVID-19 and lung carcinoma. However, the complexity of various cytoplasmic DNA-related pathways and their crosstalk remains challenging to distinguish their specific roles in many distinct inflammatory diseases, especially for the underlying mechanisms. Here, we reviewed the latest findings on cytoplasmic DNA and its signaling pathways in inflammatory lung conditions and lung cancer progression. We found that sustained activation of cytoplasmic DNA sensing pathways contributes to the development of common lung diseases, which may result from external factors or mutations of key genes in the organism. We further discussed the interplays between cytoplasmic DNA and anti-inflammatory or anti-tumor effects for potential immunotherapy. In sum, this review aids in understanding the roles of cytoplasmic DNAs and exploring more therapeutic strategies.
Collapse
Affiliation(s)
- Xintong He
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
- Zhejiang University-University of Edinburgh Institute (ZJU-UoE Institute), Zhejiang University School of Medicine, Zhejiang University, Haining, China
- College of Medicine and Veterinary Medicine, The University of Edinburgh, Edinburgh, United Kingdom
| | - Ye Sun
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
- Zhejiang University-University of Edinburgh Institute (ZJU-UoE Institute), Zhejiang University School of Medicine, Zhejiang University, Haining, China
- College of Medicine and Veterinary Medicine, The University of Edinburgh, Edinburgh, United Kingdom
| | - Jianzhang Lu
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
- Zhejiang University-University of Edinburgh Institute (ZJU-UoE Institute), Zhejiang University School of Medicine, Zhejiang University, Haining, China
- College of Medicine and Veterinary Medicine, The University of Edinburgh, Edinburgh, United Kingdom
| | - Faiza Naz
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
- Zhejiang University-University of Edinburgh Institute (ZJU-UoE Institute), Zhejiang University School of Medicine, Zhejiang University, Haining, China
| | - Shenglin Ma
- Hangzhou Cancer Institution, Affiliated Hangzhou Cancer Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
- Cancer Center, Zhejiang University, Hangzhou, China
| | - Jian Liu
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
- Zhejiang University-University of Edinburgh Institute (ZJU-UoE Institute), Zhejiang University School of Medicine, Zhejiang University, Haining, China
- College of Medicine and Veterinary Medicine, The University of Edinburgh, Edinburgh, United Kingdom
- Cancer Center, Zhejiang University, Hangzhou, China
- Biomedical and Heath Translational Research Center of Zhejiang Province, Haining, China
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
6
|
Mori KM, McElroy JP, Weng DY, Chung S, Fadda P, Reisinger SA, Ying KL, Brasky TM, Wewers MD, Freudenheim JL, Shields PG, Song MA. Lung mitochondrial DNA copy number, inflammatory biomarkers, gene transcription and gene methylation in vapers and smokers. EBioMedicine 2022; 85:104301. [PMID: 36215783 PMCID: PMC9561685 DOI: 10.1016/j.ebiom.2022.104301] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 08/31/2022] [Accepted: 09/21/2022] [Indexed: 11/11/2022] Open
Abstract
BACKGROUND Mitochondrial DNA copy number (mtCN) maintains cellular function and homeostasis, and is linked to nuclear DNA methylation and gene expression. Increased mtCN in the blood is associated with smoking and respiratory disease, but has received little attention for target organ effects for smoking or electronic cigarette (EC) use. METHODS Bronchoscopy biospecimens from healthy EC users, smokers (SM), and never-smokers (NS) were assessed for associations of mtCN with mtDNA point mutations, immune responses, nuclear DNA methylation and gene expression using linear regression. Ingenuity pathway analysis was used for enriched pathways. GEO and TCGA respiratory disease datasets were used to explore the involvement of mtCN-associated signatures. FINDINGS mtCN was higher in SM than NS, but EC was not statistically different from either. Overall there was a negative association of mtCN with a point mutation in the D-loop but no difference within groups. Positive associations of mtCN with IL-2 and IL-4 were found in EC only. mtCN was significantly associated with 71,487 CpGs and 321 transcripts. 263 CpGs were correlated with nearby transcripts for genes enriched in the immune system. EC-specific mtCN-associated-CpGs and genes were differentially expressed in respiratory diseases compared to controls, including genes involved in cellular movement, inflammation, metabolism, and airway hyperresponsiveness. INTERPRETATION Smoking may elicit a lung toxic effect through mtCN. While the impact of EC is less clear, EC-specific associations of mtCN with nuclear biomarkers suggest exposure may not be harmless. Further research is needed to understand the role of smoking and EC-related mtCN on lung disease risks. FUNDING The National Cancer Institute, the National Heart, Lung, and Blood Institute, the Food and Drug Administration Center for Tobacco Products, the National Center For Advancing Translational Sciences, and Pelotonia Intramural Research Funds.
Collapse
Affiliation(s)
- Kellie M Mori
- Division of Environmental Health Sciences, College of Public Health, The Ohio State University, Columbus, OH, United States
| | - Joseph P McElroy
- Comprehensive Cancer Center, The Ohio State University and James Cancer Hospital, Columbus, OH, United States
| | - Daniel Y Weng
- Comprehensive Cancer Center, The Ohio State University and James Cancer Hospital, Columbus, OH, United States
| | - Sangwoon Chung
- Pulmonary and Critical Care Medicine, Department of Internal Medicine, The Ohio State University, Columbus, OH, United States
| | - Paolo Fadda
- Genomics Shared Resource, The Ohio State University and James Cancer Hospital, Columbus, OH, United States
| | - Sarah A Reisinger
- Comprehensive Cancer Center, The Ohio State University and James Cancer Hospital, Columbus, OH, United States
| | - Kevin L Ying
- Comprehensive Cancer Center, The Ohio State University and James Cancer Hospital, Columbus, OH, United States
| | - Theodore M Brasky
- Comprehensive Cancer Center, The Ohio State University and James Cancer Hospital, Columbus, OH, United States
| | - Mark D Wewers
- Pulmonary and Critical Care Medicine, Department of Internal Medicine, The Ohio State University, Columbus, OH, United States
| | - Jo L Freudenheim
- Department of Epidemiology and Environmental Health, School of Public Health and Health Professions, University at Buffalo, Buffalo, NY, United States
| | - Peter G Shields
- Comprehensive Cancer Center, The Ohio State University and James Cancer Hospital, Columbus, OH, United States.
| | - Min-Ae Song
- Division of Environmental Health Sciences, College of Public Health, The Ohio State University, Columbus, OH, United States.
| |
Collapse
|
7
|
Lee SY, Lee HS, Park HW. Transcriptome analysis of sputum cells reveals two distinct molecular phenotypes of “asthma and chronic obstructive pulmonary disease overlap” in the elderly. Eur J Med Res 2022; 27:215. [DOI: 10.1186/s40001-022-00861-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Accepted: 10/18/2022] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Little is known about the pathogenesis of asthma and chronic obstructive pulmonary disease (COPD) overlap (ACO). This study examined the molecular phenotypes of ACO in the elderly.
Methods
A genome-wide investigation of gene expression in sputum cells from the elderly with asthma, ACO, or COPD was performed using gene set variation analysis (GSVA) with predefined asthma- or COPD-specific gene signatures. We then performed a subsequent cluster analysis using enrichment scores (ESs) to identify molecular clusters in the elderly with ACO. Finally, a second GSVA was conducted with curated gene signatures to gain insight into the pathogenesis of ACO associated with the identified molecular clusters.
Results
Seventy elderly individuals were enrolled (17 with asthma, 41 with ACO, and 12 with COPD). Two distinct molecular clusters of ACO were identified. Clinically, ACO cluster 1 (N = 23) was characterized by male and smoker dominance, more obstructive lung function, and higher proportions of both neutrophil and eosinophil in induced sputum compared to ACO cluster 2 (N = 18). ACO cluster 1 had molecular features similar to both asthma and COPD, with mitochondria and peroxisome dysfunction as important mechanisms in the pathogenesis of these diseases. The molecular features of ACO cluster 2 differed from those of asthma and COPD, with enhanced innate immune reactions to microorganisms identified as being important in the pathogenesis of this form of ACO.
Conclusion
Recognition of the unique biological pathways associated with the two distinct molecular phenotypes of ACO will deepen our understanding of ACO in the elderly.
Collapse
|
8
|
Emma R, Caruso M, Campagna D, Pulvirenti R, Li Volti G. The Impact of Tobacco Cigarettes, Vaping Products and Tobacco Heating Products on Oxidative Stress. Antioxidants (Basel) 2022; 11:1829. [PMID: 36139904 PMCID: PMC9495690 DOI: 10.3390/antiox11091829] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 09/06/2022] [Accepted: 09/08/2022] [Indexed: 12/02/2022] Open
Abstract
Cells constantly produce oxidizing species because of their metabolic activity, which is counteracted by the continuous production of antioxidant species to maintain the homeostasis of the redox balance. A deviation from the metabolic steady state leads to a condition of oxidative stress. The source of oxidative species can be endogenous or exogenous. A major exogenous source of these species is tobacco smoking. Oxidative damage can be induced in cells by chemical species contained in smoke through the generation of pro-inflammatory compounds and the modulation of intracellular pro-inflammatory pathways, resulting in a pathological condition. Cessation of smoking reduces the morbidity and mortality associated with cigarette use. Next-generation products (NGPs), as alternatives to combustible cigarettes, such as electronic cigarettes (e-cig) and tobacco heating products (THPs), have been proposed as a harm reduction strategy to reduce the deleterious impacts of cigarette smoking. In this review, we examine the impact of tobacco smoke and MRPs on oxidative stress in different pathologies, including respiratory and cardiovascular diseases and tumors. The impact of tobacco cigarette smoke on oxidative stress signaling in human health is well established, whereas the safety profile of MRPs seems to be higher than tobacco cigarettes, but further, well-conceived, studies are needed to better understand the oxidative effects of these products with long-term exposure.
Collapse
Affiliation(s)
- Rosalia Emma
- Department of Biomedical and Biotechnological Sciences, University of Catania, Via S. Sofia, 97, 95123 Catania, Italy
| | - Massimo Caruso
- Department of Biomedical and Biotechnological Sciences, University of Catania, Via S. Sofia, 97, 95123 Catania, Italy
- Center of Excellence for the Acceleration of Harm Reduction (CoEHAR), University of Catania, Via S. Sofia, 89, 95123 Catania, Italy
| | - Davide Campagna
- Center of Excellence for the Acceleration of Harm Reduction (CoEHAR), University of Catania, Via S. Sofia, 89, 95123 Catania, Italy
- Department of Clinical and Experimental Medicine, University of Catania, Via S. Sofia, 97, 95123 Catania, Italy
| | - Roberta Pulvirenti
- Department of Biomedical and Biotechnological Sciences, University of Catania, Via S. Sofia, 97, 95123 Catania, Italy
| | - Giovanni Li Volti
- Department of Biomedical and Biotechnological Sciences, University of Catania, Via S. Sofia, 97, 95123 Catania, Italy
- Center of Excellence for the Acceleration of Harm Reduction (CoEHAR), University of Catania, Via S. Sofia, 89, 95123 Catania, Italy
| |
Collapse
|
9
|
Aghali A, Koloko Ngassie ML, Pabelick CM, Prakash YS. Cellular Senescence in Aging Lungs and Diseases. Cells 2022; 11:cells11111781. [PMID: 35681476 PMCID: PMC9179897 DOI: 10.3390/cells11111781] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Revised: 05/23/2022] [Accepted: 05/26/2022] [Indexed: 12/10/2022] Open
Abstract
Cellular senescence represents a state of irreversible cell cycle arrest occurring naturally or in response to exogenous stressors. Following the initial arrest, progressive phenotypic changes define conditions of cellular senescence. Understanding molecular mechanisms that drive senescence can help to recognize the importance of such pathways in lung health and disease. There is increasing interest in the role of cellular senescence in conditions such as chronic obstructive pulmonary disease (COPD) and idiopathic pulmonary fibrosis (IPF) in the context of understanding pathophysiology and identification of novel therapies. Herein, we discuss the current knowledge of molecular mechanisms and mitochondrial dysfunction regulating different aspects of cellular senescence-related to chronic lung diseases to develop rational strategies for modulating the senescent cell phenotype in the lung for therapeutic benefit.
Collapse
Affiliation(s)
- Arbi Aghali
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA; (A.A.); (C.M.P.)
| | - Maunick Lefin Koloko Ngassie
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands;
- Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, University of Groningen, 9700 RB Groningen, The Netherlands
| | - Christina M. Pabelick
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA; (A.A.); (C.M.P.)
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Y. S. Prakash
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA; (A.A.); (C.M.P.)
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, MN 55905, USA
- Correspondence:
| |
Collapse
|
10
|
Lin CR, Bahmed K, Kosmider B. Dysregulated Cell Signaling in Pulmonary Emphysema. Front Med (Lausanne) 2022; 8:762878. [PMID: 35047522 PMCID: PMC8762198 DOI: 10.3389/fmed.2021.762878] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Accepted: 12/06/2021] [Indexed: 01/19/2023] Open
Abstract
Pulmonary emphysema is characterized by the destruction of alveolar septa and irreversible airflow limitation. Cigarette smoking is the primary cause of this disease development. It induces oxidative stress and disturbs lung physiology and tissue homeostasis. Alveolar type II (ATII) cells have stem cell potential and can repair the denuded epithelium after injury; however, their dysfunction is evident in emphysema. There is no effective treatment available for this disease. Challenges in this field involve the large complexity of lung pathophysiological processes and gaps in our knowledge on the mechanisms of emphysema progression. It implicates dysregulation of various signaling pathways, including aberrant inflammatory and oxidative responses, defective antioxidant defense system, surfactant dysfunction, altered proteostasis, disrupted circadian rhythms, mitochondrial damage, increased cell senescence, apoptosis, and abnormal proliferation and differentiation. Also, genetic predispositions are involved in this disease development. Here, we comprehensively review studies regarding dysregulated cell signaling, especially in ATII cells, and their contribution to alveolar wall destruction in emphysema. Relevant preclinical and clinical interventions are also described.
Collapse
Affiliation(s)
- Chih-Ru Lin
- Department of Microbiology, Immunology, and Inflammation, Temple University, Philadelphia, PA, United States.,Center for Inflammation and Lung Research, Temple University, Philadelphia, PA, United States
| | - Karim Bahmed
- Center for Inflammation and Lung Research, Temple University, Philadelphia, PA, United States.,Department of Thoracic Medicine and Surgery, Temple University, Philadelphia, PA, United States
| | - Beata Kosmider
- Department of Microbiology, Immunology, and Inflammation, Temple University, Philadelphia, PA, United States.,Center for Inflammation and Lung Research, Temple University, Philadelphia, PA, United States
| |
Collapse
|
11
|
Schuliga M, Read J, Knight DA. Ageing mechanisms that contribute to tissue remodeling in lung disease. Ageing Res Rev 2021; 70:101405. [PMID: 34242806 DOI: 10.1016/j.arr.2021.101405] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 06/13/2021] [Accepted: 07/02/2021] [Indexed: 12/12/2022]
Abstract
Age is a major risk factor for chronic respiratory diseases such as idiopathic pulmonary fibrosis (IPF), chronic obstructive pulmonary disease (COPD) and certain phenotypes of asthma. The recent COVID-19 pandemic also highlights the increased susceptibility of the elderly to acute respiratory distress syndrome (ARDS), a diffuse inflammatory lung injury with often long-term effects (ie parenchymal fibrosis). Collectively, these lung conditions are characterized by a pathogenic reparative process that, rather than restoring organ function, contributes to structural and functional tissue decline. In the ageing lung, the homeostatic control of wound healing following challenge or injury has an increased likelihood of being perturbed, increasing susceptibility to disease. This loss of fidelity is a consequence of a diverse range of underlying ageing mechanisms including senescence, mitochondrial dysfunction, proteostatic stress and diminished autophagy that occur within the lung, as well as in other tissues, organs and systems of the body. These ageing pathways are highly interconnected, involving localized and systemic increases in inflammatory mediators and damage associated molecular patterns (DAMPs); along with corresponding changes in immune cell function, metabolism and composition of the pulmonary and gut microbiomes. Here we comprehensively review the roles of ageing mechanisms in the tissue remodeling of lung disease.
Collapse
Affiliation(s)
- Michael Schuliga
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, NSW, Australia; Hunter Medical Research Institute, New Lambton Heights, NSW, Australia.
| | - Jane Read
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, NSW, Australia; Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Darryl A Knight
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, NSW, Australia; Hunter Medical Research Institute, New Lambton Heights, NSW, Australia; Providence Health Care Research Institute, Vancouver, British Columbia, Canada
| |
Collapse
|
12
|
Mitochondrial Dysfunction in Chronic Respiratory Diseases: Implications for the Pathogenesis and Potential Therapeutics. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:5188306. [PMID: 34354793 PMCID: PMC8331273 DOI: 10.1155/2021/5188306] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 06/30/2021] [Accepted: 07/16/2021] [Indexed: 02/07/2023]
Abstract
Mitochondria are indispensable for energy metabolism and cell signaling. Mitochondrial homeostasis is sustained with stabilization of mitochondrial membrane potential, balance of mitochondrial calcium, integrity of mitochondrial DNA, and timely clearance of damaged mitochondria via mitophagy. Mitochondrial dysfunction is featured by increased generation of mitochondrial reactive oxygen species, reduced mitochondrial membrane potential, mitochondrial calcium imbalance, mitochondrial DNA damage, and abnormal mitophagy. Accumulating evidence indicates that mitochondrial dysregulation causes oxidative stress, inflammasome activation, apoptosis, senescence, and metabolic reprogramming. All these cellular processes participate in the pathogenesis and progression of chronic respiratory diseases, including chronic obstructive pulmonary disease, pulmonary fibrosis, and asthma. In this review, we provide a comprehensive and updated overview of the impact of mitochondrial dysfunction on cellular processes involved in the development of these respiratory diseases. This not only implicates mechanisms of mitochondrial dysfunction for the pathogenesis of chronic lung diseases but also provides potential therapeutic approaches for these diseases by targeting dysfunctional mitochondria.
Collapse
|
13
|
Fujino N, Sugiura H. ACO (Asthma-COPD Overlap) Is Independent from COPD, a Case in Favor: A Systematic Review. Diagnostics (Basel) 2021; 11:859. [PMID: 34064650 PMCID: PMC8150952 DOI: 10.3390/diagnostics11050859] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Accepted: 05/10/2021] [Indexed: 01/03/2023] Open
Abstract
Asthma and chronic obstructive pulmonary disease (COPD) are now recognized to be able to co-exist as asthma-COPD overlap (ACO). It is clinically relevant to evaluate whether patients with COPD concurrently have components of asthma in primary care. This is because: (i) ACO is a relatively common condition among asthma (over 40 years of age) or COPD irrespective of its diagnosis criteria; (ii) patients with ACO can have higher frequency of exacerbation and more rapid decline in lung function than those with asthma or COPD; and (iii) asthmatic features such as eosinophilic airway inflammation are promising indicators for prediction of inhaled corticosteroid-responsiveness in COPD. The aim of this review to evaluate diagnostic markers for ACO. We searched PubMed for articles related to ACO published until 2020. Articles associated with diagnostic biomarkers were included. We identified a total of 25 studies, some of which have revealed that a combination of biomarkers such as fractional exhaled nitric oxide and serum immunoglobulin E is useful to discern type 2 inflammation in the airways of COPD. Here, we review the current understanding of the clinical characteristics, biomarkers and molecular pathophysiology of ACO in the context of how ACO can be differentiated from COPD.
Collapse
Affiliation(s)
- Naoya Fujino
- Department of Respiratory Medicine, Tohoku University Graduate School of Medicine, Sendai 980-8574, Japan;
| | | |
Collapse
|
14
|
Snyder RJ, Kleeberger SR. Role of Mitochondrial DNA in Inflammatory Airway Diseases. Compr Physiol 2021; 11:1485-1499. [PMID: 33577124 DOI: 10.1002/cphy.c200010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
The mitochondrial genome is a small, circular, and highly conserved piece of DNA which encodes only 13 protein subunits yet is vital for electron transport in the mitochondrion and, therefore, vital for the existence of multicellular life on Earth. Despite this importance, mitochondrial DNA (mtDNA) is located in one of the least-protected areas of the cell, exposing it to high concentrations of intracellular reactive oxygen species (ROS) and threat from exogenous substances and pathogens. Until recently, the quality control mechanisms which ensured the stability of the nuclear genome were thought to be minimal or nonexistent in the mitochondria, and the thousands of redundant copies of mtDNA in each cell were believed to be the primary mechanism of protecting these genes. However, a vast network of mechanisms has been discovered that repair mtDNA lesions, replace and recycle mitochondrial chromosomes, and conduct alternate RNA processing for previously undescribed mitochondrial proteins. New mtDNA/RNA-dependent signaling pathways reveal a mostly undiscovered biochemical landscape in which the mitochondria interface with their host cells/organisms. As the myriad ways in which the function of the mitochondrial genome can affect human health have become increasingly apparent, the use of mitogenomic biomarkers (such as copy number and heteroplasmy) as toxicological endpoints has become more widely accepted. In this article, we examine several pathologies of human airway epithelium, including particle exposures, inflammatory diseases, and hyperoxia, and discuss the role of mitochondrial genotoxicity in the pathogenesis and/or exacerbation of these conditions. © 2021 American Physiological Society. Compr Physiol 11:1485-1499, 2021.
Collapse
Affiliation(s)
- Ryan J Snyder
- National Institute of Environmental Health Sciences, NIH, Durham, North Carolina, USA
| | - Steven R Kleeberger
- National Institute of Environmental Health Sciences, NIH, Durham, North Carolina, USA
| |
Collapse
|
15
|
Cocco MP, White E, Xiao S, Hu D, Mak A, Sleiman P, Yang M, Bobbitt KR, Gui H, Levin AM, Hochstadt S, Whitehouse K, Rynkowski D, Barczak AJ, Abecasis G, Blackwell TW, Kang HM, Nickerson DA, Germer S, Ding J, Lanfear DE, Gilliland F, Gauderman WJ, Kumar R, Erle DJ, Martinez F, Hakonarson H, Burchard EG, Williams LK. Asthma and its relationship to mitochondrial copy number: Results from the Asthma Translational Genomics Collaborative (ATGC) of the Trans-Omics for Precision Medicine (TOPMed) program. PLoS One 2020; 15:e0242364. [PMID: 33237978 PMCID: PMC7688161 DOI: 10.1371/journal.pone.0242364] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Accepted: 10/30/2020] [Indexed: 02/07/2023] Open
Abstract
Background Mitochondria support critical cellular functions, such as energy production through oxidative phosphorylation, regulation of reactive oxygen species, apoptosis, and calcium homeostasis. Objective Given the heightened level of cellular activity in patients with asthma, we sought to determine whether mitochondrial DNA (mtDNA) copy number measured in peripheral blood differed between individuals with and without asthma. Methods Whole genome sequence data was generated as part of the Trans-Omics for Precision Medicine (TOPMed) Program on participants from the Study of Asthma Phenotypes and Pharmacogenomic Interactions by Race-ethnicity (SAPPHIRE) and the Study of African Americans, Asthma, Genes, & Environment II (SAGE II). We restricted our analysis to individuals who self-identified as African American (3,651 asthma cases and 1,344 controls). Mitochondrial copy number was estimated using the sequencing read depth ratio for the mitochondrial and nuclear genomes. Respiratory complex expression was assessed using RNA-sequencing. Results Average mitochondrial copy number was significantly higher among individuals with asthma when compared with controls (SAPPHIRE: 218.60 vs. 200.47, P<0.001; SAGE II: 235.99 vs. 223.07, P<0.001). Asthma status was significantly associated with mitochondrial copy number after accounting for potential explanatory variables, such as participant age, sex, leukocyte counts, and mitochondrial haplogroup. Despite the consistent relationship between asthma status and mitochondrial copy number, the latter was not associated with time-to-exacerbation or patient-reported asthma control. Mitochondrial respiratory complex gene expression was disproportionately lower in individuals with asthma when compared with individuals without asthma and other protein-encoding genes. Conclusions We observed a robust association between asthma and higher mitochondrial copy number. Asthma having an effect on mitochondria function was also supported by lower respiratory complex gene expression in this group.
Collapse
Affiliation(s)
- Maxwell P. Cocco
- Center for Individualized and Genomic Medicine Research (CIGMA), Department of Internal Medicine, Henry Ford Health System, Detroit, Michigan, United States of America
| | - Evan White
- Center for Individualized and Genomic Medicine Research (CIGMA), Department of Internal Medicine, Henry Ford Health System, Detroit, Michigan, United States of America
| | - Shujie Xiao
- Center for Individualized and Genomic Medicine Research (CIGMA), Department of Internal Medicine, Henry Ford Health System, Detroit, Michigan, United States of America
| | - Donglei Hu
- Department of Medicine, University of California San Francisco, San Francisco, California, United States of America
| | - Angel Mak
- Department of Medicine, University of California San Francisco, San Francisco, California, United States of America
| | - Patrick Sleiman
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, United States of America
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Mao Yang
- Center for Individualized and Genomic Medicine Research (CIGMA), Department of Internal Medicine, Henry Ford Health System, Detroit, Michigan, United States of America
| | - Kevin R. Bobbitt
- Department of Public Health Sciences, Henry Ford Health System, Detroit, Michigan, United States of America
| | - Hongsheng Gui
- Center for Individualized and Genomic Medicine Research (CIGMA), Department of Internal Medicine, Henry Ford Health System, Detroit, Michigan, United States of America
| | - Albert M. Levin
- Department of Public Health Sciences, Henry Ford Health System, Detroit, Michigan, United States of America
| | - Samantha Hochstadt
- Center for Individualized and Genomic Medicine Research (CIGMA), Department of Internal Medicine, Henry Ford Health System, Detroit, Michigan, United States of America
| | - Kyle Whitehouse
- Center for Individualized and Genomic Medicine Research (CIGMA), Department of Internal Medicine, Henry Ford Health System, Detroit, Michigan, United States of America
| | - Dean Rynkowski
- Center for Individualized and Genomic Medicine Research (CIGMA), Department of Internal Medicine, Henry Ford Health System, Detroit, Michigan, United States of America
| | - Andrea J. Barczak
- Lung Biology Center and UCSF CoLabs, University of California San Francisco, San Francisco, California, United States of America
| | - Gonçalo Abecasis
- Center for Statistical Genetics, University of Michigan, Ann Arbor, Michigan, United States of America
- Regeneron Pharmaceuticals, Inc., Tarrytown, New York, United States of America
| | - Thomas W. Blackwell
- Center for Statistical Genetics, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Hyun Min Kang
- Center for Statistical Genetics, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Deborah A. Nickerson
- Department of Genome Sciences, University of Washington, Seattle, Washington, United States of America
- Northwest Genomics Center, Seattle, Washington, United States of America
- Brotman Baty Institute, Seattle, Washington, United States of America
| | - Soren Germer
- New York Genome Center, New York, New York, United States of America
| | - Jun Ding
- Human Statistical Genetics Unit, Laboratory of Genetics and Genomics, National Institute on Aging, National Institutes of Health, Bethesda, Maryland, United States of America
| | - David E. Lanfear
- Center for Individualized and Genomic Medicine Research (CIGMA), Department of Internal Medicine, Henry Ford Health System, Detroit, Michigan, United States of America
| | - Frank Gilliland
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
| | - W. James Gauderman
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
| | - Rajesh Kumar
- Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, Illinois, United States of America
| | - David J. Erle
- Department of Medicine, University of California San Francisco, San Francisco, California, United States of America
- Lung Biology Center and UCSF CoLabs, University of California San Francisco, San Francisco, California, United States of America
| | - Fernando Martinez
- Arizona Respiratory Center and Department of Pediatrics, University of Arizona, Tucson, Arizona, United States of America
| | - Hakon Hakonarson
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, United States of America
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Esteban G. Burchard
- Department of Medicine, University of California San Francisco, San Francisco, California, United States of America
- Department of Bioengineering & Therapeutic Sciences, University of California San Francisco, San Francisco, California, United States of America
| | - L. Keoki Williams
- Center for Individualized and Genomic Medicine Research (CIGMA), Department of Internal Medicine, Henry Ford Health System, Detroit, Michigan, United States of America
- * E-mail:
| |
Collapse
|
16
|
Manevski M, Muthumalage T, Devadoss D, Sundar IK, Wang Q, Singh KP, Unwalla HJ, Chand HS, Rahman I. Cellular stress responses and dysfunctional Mitochondrial-cellular senescence, and therapeutics in chronic respiratory diseases. Redox Biol 2020; 33:101443. [PMID: 32037306 PMCID: PMC7251248 DOI: 10.1016/j.redox.2020.101443] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2019] [Revised: 01/14/2020] [Accepted: 01/22/2020] [Indexed: 02/06/2023] Open
Abstract
The abnormal inflammatory responses due to the lung tissue damage and ineffective repair/resolution in response to the inhaled toxicants result in the pathological changes associated with chronic respiratory diseases. Investigation of such pathophysiological mechanisms provides the opportunity to develop the molecular phenotype-specific diagnostic assays and could help in designing the personalized medicine-based therapeutic approaches against these prevalent diseases. As the central hubs of cell metabolism and energetics, mitochondria integrate cellular responses and interorganellar signaling pathways to maintain cellular and extracellular redox status and the cellular senescence that dictate the lung tissue responses. Specifically, as observed in chronic obstructive pulmonary disease (COPD) and pulmonary fibrosis, the mitochondria-endoplasmic reticulum (ER) crosstalk is disrupted by the inhaled toxicants such as the combustible and emerging electronic nicotine-delivery system (ENDS) tobacco products. Thus, the recent research efforts have focused on understanding how the mitochondria-ER dysfunctions and oxidative stress responses can be targeted to improve inflammatory and cellular dysfunctions associated with these pathologic illnesses that are exacerbated by viral infections. The present review assesses the importance of these redox signaling and cellular senescence pathways that describe the role of mitochondria and ER on the development and function of lung epithelial responses, highlighting the cause and effect associations that reflect the disease pathogenesis and possible intervention strategies.
Collapse
Affiliation(s)
- Marko Manevski
- Department of Immunology and NanoMedicine, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, USA
| | - Thivanka Muthumalage
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Dinesh Devadoss
- Department of Immunology and NanoMedicine, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, USA
| | - Isaac K Sundar
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Qixin Wang
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Kameshwar P Singh
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Hoshang J Unwalla
- Department of Immunology and NanoMedicine, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, USA
| | - Hitendra S Chand
- Department of Immunology and NanoMedicine, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, USA
| | - Irfan Rahman
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY, USA.
| |
Collapse
|
17
|
Gong S, Pu Y, Xie L, Yang X, Mao H. Fraction of Exhaled Nitric Oxide Is Elevated in Patients With Stable Chronic Obstructive Pulmonary Disease: A Meta-analysis. Am J Med Sci 2020; 360:166-175. [PMID: 32536416 DOI: 10.1016/j.amjms.2020.04.038] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Revised: 04/10/2020] [Accepted: 04/29/2020] [Indexed: 02/08/2023]
Abstract
BACKGROUND Fraction of exhaled nitric oxide (FeNO) is a noninvasive indicator of eosinophilic airway inflammation and has been used for the diagnosis and treatment of asthma. The levels of FeNO are controversial in patients with stable chronic obstructive pulmonary disease (COPD). Accordingly, this study aimed to assess FeNO levels in patients with stable COPD. MATERIALS AND METHODS A search of the Medline, Embase, Web of Science, ClinicalTrials.gov and The Cochrane Library databases was performed in August 2019. The literature search was restricted to articles published in English. Studies were included if they reported data addressing FeNO levels in patients with stable COPD and healthy controls. Review Manager version 5.3 (The Nordic Cochrane Centre, The Cochrane Collaboration, Copenhagen, Denmark) was used for meta-analysis. RESULTS A total of 19 studies were included. Analysis revealed that FeNO levels in patients with stable COPD were higher than those in the healthy control group (mean difference [MD] 2.49 [95% confidence interval {CI} 0.99-4.00]; P < 0.05), those in nonsmoking patients with stable COPD were higher than those in the healthy control group (MD 5.04 [95% CI 2.19-7.89]; P < 0.05) and those in smoking patients with stable COPD were not higher than those in the healthy control group (MD 0.30 [95% CI -2.81 to 3.41]; P = 0.85). FeNO measured using a chemiluminescence analyzer in nonsmoking patients with stable COPD was higher than those in the healthy control group (MD 4.84 [95% CI 1.83-7.86]; P < 0.05). CONCLUSIONS Findings suggested that FeNO levels in patients with stable COPD were elevated, and that smokers exhibited decreased levels.
Collapse
Affiliation(s)
- Shenglan Gong
- Department of Respiratory and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yin Pu
- Department of Respiratory and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Lingli Xie
- Department of Respiratory and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xiaoya Yang
- Department of Respiratory and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Hui Mao
- Department of Respiratory and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| |
Collapse
|
18
|
New Insights into the Implication of Mitochondrial Dysfunction in Tissue, Peripheral Blood Mononuclear Cells, and Platelets during Lung Diseases. J Clin Med 2020; 9:jcm9051253. [PMID: 32357474 PMCID: PMC7287602 DOI: 10.3390/jcm9051253] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Revised: 04/16/2020] [Accepted: 04/20/2020] [Indexed: 12/13/2022] Open
Abstract
Lung diseases such as chronic obstructive pulmonary disease, asthma, pulmonary arterial hypertension, or idiopathic pulmonary fibrosis are major causes of morbidity and mortality. Complex, their physiopathology is multifactorial and includes lung mitochondrial dysfunction and enhanced reactive oxygen species (ROS) release, which deserves increased attention. Further, and importantly, circulating blood cells (peripheral blood mononuclear cells-(PBMCs) and platelets) likely participate in these systemic diseases. This review presents the data published so far and shows that circulating blood cells mitochondrial oxidative capacity are likely to be reduced in chronic obstructive pulmonary disease (COPD), but enhanced in asthma and pulmonary arterial hypertension in a context of increased oxidative stress. Besides such PBMCs or platelets bioenergetics modifications, mitochondrial DNA (mtDNA) changes have also been observed in patients. These new insights open exciting challenges to determine their role as biomarkers or potential guide to a new therapeutic approach in lung diseases.
Collapse
|
19
|
Zhang WZ, Rice MC, Hoffman KL, Oromendia C, Barjaktarevic IZ, Wells JM, Hastie AT, Labaki WW, Cooper CB, Comellas AP, Criner GJ, Krishnan JA, Paine R, Hansel NN, Bowler RP, Barr RG, Peters SP, Woodruff PG, Curtis JL, Han MK, Ballman KV, Martinez FJ, Choi AM, Nakahira K, Cloonan SM, Choi ME. Association of urine mitochondrial DNA with clinical measures of COPD in the SPIROMICS cohort. JCI Insight 2020; 5:133984. [PMID: 31895696 DOI: 10.1172/jci.insight.133984] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Accepted: 12/26/2019] [Indexed: 12/16/2022] Open
Abstract
BACKGROUNDMitochondrial dysfunction, a proposed mechanism of chronic obstructive pulmonary disease (COPD) pathogenesis, is associated with the leakage of mitochondrial DNA (mtDNA), which may be detected extracellularly in various bodily fluids. Despite evidence for the increased prevalence of chronic kidney disease in COPD subjects and for mitochondrial dysfunction in the kidneys of murine COPD models, whether urine mtDNA (u-mtDNA) associates with measures of disease severity in COPD is unknown.METHODSCell-free u-mtDNA, defined as copy number of mitochondrially encoded NADH dehydrogenase-1 (MTND1) gene, was measured by quantitative PCR and normalized to urine creatinine in cell-free urine samples from participants in the Subpopulations and Intermediate Outcome Measures in COPD Study (SPIROMICS) cohort. Urine albumin/creatinine ratios (UACR) were measured in the same samples. Associations between u-mtDNA, UACR, and clinical disease parameters - including FEV1 % predicted, clinical measures of exercise tolerance, respiratory symptom burden, and chest CT measures of lung structure - were examined.RESULTSU-mtDNA and UACR levels were measured in never smokers (n = 64), smokers without airflow obstruction (n = 109), participants with mild/moderate COPD (n = 142), and participants with severe COPD (n = 168). U-mtDNA was associated with increased respiratory symptom burden, especially among smokers without COPD. Significant sex differences in u-mtDNA levels were observed, with females having higher u-mtDNA levels across all study subgroups. U-mtDNA associated with worse spirometry and CT emphysema in males only and with worse respiratory symptoms in females only. Similar associations were not found with UACR.CONCLUSIONU-mtDNA levels may help to identify distinct clinical phenotypes and underlying pathobiological differences in males versus females with COPD.TRIAL REGISTRATIONThis study has been registered at ClinicalTrials.gov ( NCT01969344).FUNDINGUS NIH, National Heart, Lung and Blood Institute, supplemented by contributions made through the Foundation for the NIH and the COPD Foundation from AstraZeneca/MedImmune, Bayer, Bellerophon Therapeutics, Boehringer-Ingelheim Pharmaceuticals Inc., Chiesi Farmaceutici S.p.A., Forest Research Institute Inc., GlaxoSmithKline, Grifols Therapeutics Inc., Ikaria Inc., Novartis Pharmaceuticals Corporation, Nycomed GmbH, ProterixBio, Regeneron Pharmaceuticals Inc., Sanofi, Sunovion, Takeda Pharmaceutical Company, and Theravance Biopharma and Mylan.
Collapse
Affiliation(s)
- William Z Zhang
- Division of Pulmonary and Critical Care Medicine, Joan and Sanford I. Weill Department of Medicine, New York, New York, USA.,New York-Presbyterian Hospital, Weill Cornell Medicine, New York, New York, USA
| | - Michelle C Rice
- Division of Nephrology and Hypertension, Joan and Sanford I. Weill Department of Medicine, and
| | - Katherine L Hoffman
- Department of Healthcare Policy and Research, Division of Biostatistics and Epidemiology, Weill Cornell Medicine, New York, New York, USA
| | - Clara Oromendia
- Department of Healthcare Policy and Research, Division of Biostatistics and Epidemiology, Weill Cornell Medicine, New York, New York, USA
| | - Igor Z Barjaktarevic
- Division of Pulmonary and Critical Care Medicine, UCLA Medical Center, Los Angeles, California, USA
| | - J Michael Wells
- University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Annette T Hastie
- Pulmonary, Critical Care, Allergy, and Immunologic Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Wassim W Labaki
- Division of Pulmonary and Critical Care Medicine, University of Michigan Health System, Ann Arbor, Michigan, USA
| | - Christopher B Cooper
- Division of Pulmonary and Critical Care Medicine, UCLA Medical Center, Los Angeles, California, USA
| | - Alejandro P Comellas
- Division of Pulmonary and Critical Care, University of Iowa, Iowa City, Iowa, USA
| | - Gerard J Criner
- Department of Thoracic Medicine and Surgery, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania, USA
| | - Jerry A Krishnan
- Division of Pulmonary, Critical Care, Sleep and Allergy, University of Illinois College of Medicine, Chicago, Illinois, USA
| | - Robert Paine
- Division of Respiratory, Critical Care, and Occupational Pulmonary Medicine, University of Utah, Salt Lake City, Utah, USA
| | - Nadia N Hansel
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Russell P Bowler
- Division of Pulmonary, Critical Care Medicine, National Jewish Health, Denver, Colorado, USA
| | - R Graham Barr
- Columbia University Medical Center, New York, New York, USA
| | - Stephen P Peters
- Pulmonary, Critical Care, Allergy, and Immunologic Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Prescott G Woodruff
- Division of Pulmonary and Critical Care Medicine, UCSF, School of Medicine, San Francisco, California, USA
| | - Jeffrey L Curtis
- Division of Pulmonary and Critical Care Medicine, University of Michigan Health System, Ann Arbor, Michigan, USA.,Veterans Affairs Ann Arbor Healthcare System, Ann Arbor, Michigan, USA
| | - Meilan K Han
- Division of Pulmonary and Critical Care Medicine, University of Michigan Health System, Ann Arbor, Michigan, USA
| | - Karla V Ballman
- Department of Healthcare Policy and Research, Division of Biostatistics and Epidemiology, Weill Cornell Medicine, New York, New York, USA
| | - Fernando J Martinez
- Division of Pulmonary and Critical Care Medicine, Joan and Sanford I. Weill Department of Medicine, New York, New York, USA.,New York-Presbyterian Hospital, Weill Cornell Medicine, New York, New York, USA
| | - Augustine Mk Choi
- Division of Pulmonary and Critical Care Medicine, Joan and Sanford I. Weill Department of Medicine, New York, New York, USA.,New York-Presbyterian Hospital, Weill Cornell Medicine, New York, New York, USA
| | - Kiichi Nakahira
- Division of Pulmonary and Critical Care Medicine, Joan and Sanford I. Weill Department of Medicine, New York, New York, USA
| | - Suzanne M Cloonan
- Division of Pulmonary and Critical Care Medicine, Joan and Sanford I. Weill Department of Medicine, New York, New York, USA
| | - Mary E Choi
- New York-Presbyterian Hospital, Weill Cornell Medicine, New York, New York, USA.,Division of Nephrology and Hypertension, Joan and Sanford I. Weill Department of Medicine, and
| | | |
Collapse
|
20
|
Benmerzoug S, Ryffel B, Togbe D, Quesniaux VF. Self-DNA Sensing in Lung Inflammatory Diseases. Trends Immunol 2019; 40:719-734. [DOI: 10.1016/j.it.2019.06.001] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Revised: 05/31/2019] [Accepted: 06/04/2019] [Indexed: 02/07/2023]
|
21
|
Lal C, Strange C. A review of current and developing fixed-dose LABA/LAMA combinations for treating COPD. Expert Opin Pharmacother 2017; 18:1833-1843. [PMID: 29115881 DOI: 10.1080/14656566.2017.1403583] [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] [Indexed: 12/14/2022]
Abstract
INTRODUCTION The current GOLD (Global Initiative for Chronic Obstructive Lung Disease) recommendations suggest using long acting β2 agonists (LABA) and long acting muscarinic antagonists (LAMA) in combination for group B COPD patients with persistent symptoms, group C COPD patients with further exacerbations on LAMA therapy alone and for group D COPD patients with or without combination with inhaled corticosteroids (ICS). Thus, there is a lot of interest in developing LABA/LAMA combinations for maintenance therapy of chronic stable COPD. Areas covered: Many LABA/LAMA combinations have successfully been approved through carefully designed pivotal clinical trials. The current clinical use of LABA/LAMA combinations in COPD will continue to evolve as new trials with and without inhaled corticosteroids are completed. Expert opinion: Combining different classes of bronchodilators in a single inhaler is an attractive concept that can potentially improve patient adherence to therapy. Because LABA/LAMA combinations are the preferred treatment option for preventing COPD exacerbations in the updated GOLD guidelines for COPD, they will be clinically used. Future treatment of COPD should revolve around a personalized approach based on characterization of the COPD phenotype.
Collapse
Affiliation(s)
- Chitra Lal
- a Pulmonary, Critical Care, Allergy and Sleep Medicine , Medical University of South Carolina , Charleston , SC , USA
| | - Charlie Strange
- a Pulmonary, Critical Care, Allergy and Sleep Medicine , Medical University of South Carolina , Charleston , SC , USA
| |
Collapse
|
22
|
Hines KL, Peebles RS. Management of the Asthma-COPD Overlap Syndrome (ACOS): a Review of the Evidence. Curr Allergy Asthma Rep 2017; 17:15. [PMID: 28283854 DOI: 10.1007/s11882-017-0683-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
PURPOSE OF REVIEW Examine the definition of the asthma-COPD overlap syndrome (ACOS) and current treatment strategies. RECENT FINDINGS Patients with the ACOS have a lower quality of life and suffer from more complications than those affected by either disease alone. Diagnosis of ACOS is difficult because of the clinical similarities between the two diseases and the various phenotypes that comprise the syndrome. Defining treatment strategies for ACOS has been challenging because many clinical trials for asthma therapy have purposefully excluded patients with features of COPD, and COPD clinical trials have not included patients who might have an asthmatic component to their disease. Therefore, there are few randomized treatment trials which have included patients who have ACOS. Smoking cessation and appropriate vaccinations are cornerstone therapies, and pharmacologic therapy has focused on bronchodilators and inhaled corticosteroids. The role of biologics, such as omalizumab and IL-5 antagonists, in ACOS treatment is still being defined. As of now, with the paucity of randomized control trials guiding treatment strategies, the Global Initiative for Asthma (GINA) and Global Initiative for Chronic Obstructive Lung Disease (GOLD) guidelines recommend treating ACOS according to the dominant phenotype.
Collapse
Affiliation(s)
- Kristin L Hines
- Department of Medicine, Vanderbilt University Medical Center, T-1218 MCN, 1161 21st Avenue South, Nashville, TN, 37232-2650, USA
| | - R Stokes Peebles
- Department of Medicine, Vanderbilt University Medical Center, T-1218 MCN, 1161 21st Avenue South, Nashville, TN, 37232-2650, USA.
| |
Collapse
|
23
|
Corlateanu A, Covantev S, Mathioudakis AG, Botnaru V, Siafakas N. Ashtma-Chronic obstructive pulmonary disease overlap syndrome (ACOS): current evidence and future research directions. ACTA ACUST UNITED AC 2017. [DOI: 10.1186/s40749-017-0025-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
24
|
Lerner CA, Sundar IK, Rahman I. Mitochondrial redox system, dynamics, and dysfunction in lung inflammaging and COPD. Int J Biochem Cell Biol 2016; 81:294-306. [PMID: 27474491 DOI: 10.1016/j.biocel.2016.07.026] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Revised: 07/21/2016] [Accepted: 07/23/2016] [Indexed: 01/01/2023]
Abstract
Myriad forms of endogenous and environmental stress disrupt mitochondrial function by impacting critical processes in mitochondrial homeostasis, such as mitochondrial redox system, oxidative phosphorylation, biogenesis, and mitophagy. External stressors that interfere with the steady state activity of mitochondrial functions are generally associated with an increase in reactive oxygen species, inflammatory response, and induction of cellular senescence (inflammaging) potentially via mitochondrial damage associated molecular patterns (DAMPS). Many of these are the key events in the pathogenesis of chronic obstructive pulmonary disease (COPD) and its exacerbations. In this review, we highlight the primary mitochondrial quality control mechanisms that are influenced by oxidative stress/redox system, including role of mitochondria during inflammation and cellular senescence, and how mitochondrial dysfunction contributes to the pathogenesis of COPD and its exacerbations via pathogenic stimuli.
Collapse
Affiliation(s)
- Chad A Lerner
- Department of Environmental Medicine, Lung Biology and Disease Program, University of Rochester Medical Center, Rochester, NY, USA
| | - Isaac K Sundar
- Department of Environmental Medicine, Lung Biology and Disease Program, University of Rochester Medical Center, Rochester, NY, USA
| | - Irfan Rahman
- Department of Environmental Medicine, Lung Biology and Disease Program, University of Rochester Medical Center, Rochester, NY, USA.
| |
Collapse
|