1
|
Miklós Z, Horváth I. The Role of Oxidative Stress and Antioxidants in Cardiovascular Comorbidities in COPD. Antioxidants (Basel) 2023; 12:1196. [PMID: 37371927 DOI: 10.3390/antiox12061196] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Revised: 05/28/2023] [Accepted: 05/30/2023] [Indexed: 06/29/2023] Open
Abstract
Oxidative stress driven by several environmental and local airway factors associated with chronic obstructive bronchiolitis, a hallmark feature of COPD, plays a crucial role in disease pathomechanisms. Unbalance between oxidants and antioxidant defense mechanisms amplifies the local inflammatory processes, worsens cardiovascular health, and contributes to COPD-related cardiovascular dysfunctions and mortality. The current review summarizes recent developments in our understanding of different mechanisms contributing to oxidative stress and its countermeasures, with special attention to those that link local and systemic processes. Major regulatory mechanisms orchestrating these pathways are also introduced, with some suggestions for further research in the field.
Collapse
Affiliation(s)
- Zsuzsanna Miklós
- National Korányi Institute for Pulmonology, Korányi F. Street 1, H-1121 Budapest, Hungary
| | - Ildikó Horváth
- National Korányi Institute for Pulmonology, Korányi F. Street 1, H-1121 Budapest, Hungary
- Department of Pulmonology, University of Debrecen, Nagyerdei krt 98, H-4032 Debrecen, Hungary
| |
Collapse
|
2
|
Dailah HG. Therapeutic Potential of Small Molecules Targeting Oxidative Stress in the Treatment of Chronic Obstructive Pulmonary Disease (COPD): A Comprehensive Review. Molecules 2022; 27:molecules27175542. [PMID: 36080309 PMCID: PMC9458015 DOI: 10.3390/molecules27175542] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 08/21/2022] [Accepted: 08/25/2022] [Indexed: 12/02/2022] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is an increasing and major global health problem. COPD is also the third leading cause of death worldwide. Oxidative stress (OS) takes place when various reactive species and free radicals swamp the availability of antioxidants. Reactive nitrogen species, reactive oxygen species (ROS), and their counterpart antioxidants are important for host defense and physiological signaling pathways, and the development and progression of inflammation. During the disturbance of their normal steady states, imbalances between antioxidants and oxidants might induce pathological mechanisms that can further result in many non-respiratory and respiratory diseases including COPD. ROS might be either endogenously produced in response to various infectious pathogens including fungi, viruses, or bacteria, or exogenously generated from several inhaled particulate or gaseous agents including some occupational dust, cigarette smoke (CS), and air pollutants. Therefore, targeting systemic and local OS with therapeutic agents such as small molecules that can increase endogenous antioxidants or regulate the redox/antioxidants system can be an effective approach in treating COPD. Various thiol-based antioxidants including fudosteine, erdosteine, carbocysteine, and N-acetyl-L-cysteine have the capacity to increase thiol content in the lungs. Many synthetic molecules including inhibitors/blockers of protein carbonylation and lipid peroxidation, catalytic antioxidants including superoxide dismutase mimetics, and spin trapping agents can effectively modulate CS-induced OS and its resulting cellular alterations. Several clinical and pre-clinical studies have demonstrated that these antioxidants have the capacity to decrease OS and affect the expressions of several pro-inflammatory genes and genes that are involved with redox and glutathione biosynthesis. In this article, we have summarized the role of OS in COPD pathogenesis. Furthermore, we have particularly focused on the therapeutic potential of numerous chemicals, particularly antioxidants in the treatment of COPD.
Collapse
Affiliation(s)
- Hamad Ghaleb Dailah
- Research and Scientific Studies Unit, College of Nursing, Jazan University, Jazan 45142, Saudi Arabia
| |
Collapse
|
3
|
Li H, Hong W, Zeng Z, Gong S, Wu F, Wang Z, Tian H, Cheng J, Sun R, Gao M, Liang C, Cao W, Hu G, Li Y, Wei L, Zhou Y, Ran P. Association Between Extracellular Superoxide Dismutase Activity and 1-Year All-Cause Mortality in Patients With Acute Exacerbations of Chronic Obstructive Pulmonary Disease: A Prospective Cohort Study. Front Med (Lausanne) 2022; 9:811975. [PMID: 35360751 PMCID: PMC8963916 DOI: 10.3389/fmed.2022.811975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 02/03/2022] [Indexed: 11/13/2022] Open
Abstract
Background and ObjectivesAccumulating evidence suggests that oxidative stress is involved in the development of chronic obstructive pulmonary disease (COPD) and its progression. Activity of extracellular superoxide dismutase (ecSOD), the only extracellular enzyme eliminating superoxide radicals, has been reported to decline in acute exacerbations of COPD (AECOPD). However, the association between serum ecSOD activity and 1-year all-cause mortality in AECOPD patients remains unclear. The objective of our study was to explore the usefulness of ecSOD activity on admission in AECOPD as an objective predictor for 1-year all-cause mortality.MethodsWe measured serum ecSOD activity in AECOPD patients on admission in a prospective cohort study. We also recorded their laboratory and clinical data. Multivariate Cox regression was used to analyze the association between ecSOD activity and the risk of 1-year all-cause mortality. Restricted cubic spline curves were used to visualize the relationship between ecSOD activity and the hazard ratio of 1-year all-cause mortality.ResultsA total of 367 patients were followed up for 1 year, and 29 patients died during a 1-year follow-up period. Compared with survivors, the non-survivors were older (79.52 ± 8.39 vs. 74.38 ± 9.34 years old, p = 0.004) and had increased levels of tobacco consumption (47.07 ± 41.67 vs. 33.83 ± 31.79 pack-years, p = 0.037). Having an ecSOD activity ≤ 98.8 U/ml was an independent risk factor of 1-year all-cause mortality after adjustment for baseline differences, clinical variables and comorbidities [hazard ratio = 5.51, 95% confidence interval (CI): 2.35–12.95, p < 0.001].ConclusionLower serum ecSOD activity was a strong and independent predictor of 1-year all-cause mortality in AECOPD patients.
Collapse
Affiliation(s)
- Haiqing Li
- Department of Respiratory Medicine, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- State Key Laboratory of Respiratory Disease, National Center for Respiratory Medicine, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Wei Hong
- GMU-GIBH Joint School of Life Sciences, Guangzhou Medical University, Guangzhou, China
| | - Zixiong Zeng
- Department of Respiratory Medicine, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Shan Gong
- Department of Respiratory Medicine, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Fan Wu
- State Key Laboratory of Respiratory Disease, National Center for Respiratory Medicine, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Zihui Wang
- State Key Laboratory of Respiratory Disease, National Center for Respiratory Medicine, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Heshen Tian
- State Key Laboratory of Respiratory Disease, National Center for Respiratory Medicine, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Juan Cheng
- Department of Respiratory Medicine, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Ruiting Sun
- State Key Laboratory of Respiratory Disease, National Center for Respiratory Medicine, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Mi Gao
- GMU-GIBH Joint School of Life Sciences, Guangzhou Medical University, Guangzhou, China
| | - Chunxiao Liang
- State Key Laboratory of Respiratory Disease, National Center for Respiratory Medicine, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Weitao Cao
- State Key Laboratory of Respiratory Disease, National Center for Respiratory Medicine, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Guoping Hu
- Department of Respiratory Medicine, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- *Correspondence: Guoping Hu
| | - Yuqun Li
- Department of Respiratory Medicine, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Liping Wei
- Department of Respiratory Medicine, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yumin Zhou
- State Key Laboratory of Respiratory Disease, National Center for Respiratory Medicine, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Pixin Ran
- State Key Laboratory of Respiratory Disease, National Center for Respiratory Medicine, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Pixin Ran
| |
Collapse
|
4
|
Okeleji LO, Ajayi AF, Adebayo-Gege G, Aremu VO, Adebayo OI, Adebayo ET. Epidemiologic evidence linking oxidative stress and pulmonary function in healthy populations. Chronic Dis Transl Med 2021; 7:88-99. [PMID: 34136768 PMCID: PMC8180443 DOI: 10.1016/j.cdtm.2020.11.004] [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: 10/12/2020] [Indexed: 12/11/2022] Open
Abstract
Respiratory health in the general population declines regardless of the presence of pulmonary diseases. Oxidative stress has been implicated as one of the mechanisms involved in respiratory dysfunction. This review was to evaluate studies that relate oxidative stress factors with pulmonary function among the general population without prior respiratory illnesses. The search yielded 54 citations. Twenty-one studies qualified for incorporation in this review. Owing to the heterogeneity of the review, studies were discussed based on identified oxidative stress factors responsible for pulmonary dysfunction. Oxidative stress biomarkers, including gene polymorphisms of nuclear factor erythroid 2-related factor 2, heme oxygenase 1, glutathione S transferase, superoxide dismutase, and lipid peroxidation products were involved in lung function decline. In addition, the antioxidant status of individuals in reference to dietary antioxidant intake and exposure to environmental pollutants affected oxidative stress and pulmonary function, as indicated by forced expired volume in one second, forced vital capacity, and forced expiratory flow at 25%–75%. This review indicated that oxidative stress is implicated in the gradual decline of lung function among the general population, and gene polymorphism along the antioxidant defense line and/or their interaction with air pollutants reduce lung function. Different polymorphic forms among individuals explain why the rate of lung function decline differs among people. Dietary antioxidants have respiratory health benefits in antioxidant gene polymorphic forms. Therefore, the genetic composition of an individual may be considered for monitoring and identifying people at risk of respiratory illnesses.
Collapse
Affiliation(s)
- Lateef Olabisi Okeleji
- Cardio-thoracic Unit, Obafemi Awolowo University Teaching Hospital, Ile-Ife, Osun state, Nigeria
| | - Ayodeji Folorunsho Ajayi
- Department of Physiology, Ladoke Akintola University of Technology, Ogbomoso, Oyo State, Nigeria
| | - Grace Adebayo-Gege
- Department of Physiology, Baze University, Kuchigoro, Jabi, Abuja, Nigeria
| | - Victoria Oyetayo Aremu
- Department of Physiology, Ladoke Akintola University of Technology, Ogbomoso, Oyo State, Nigeria
| | | | - Emmanuel Tayo Adebayo
- Department of Physiology, Ladoke Akintola University of Technology, Ogbomoso, Oyo State, Nigeria
| |
Collapse
|
5
|
Löfgren M, Larsson P, Lindberg R, Hörnaeus K, Tydén E. Expression of xenobiotic metabolising enzymes in lungs of horses with or without histological evidence of lower airway inflammation. Vet Med Sci 2020; 7:16-24. [PMID: 32791560 PMCID: PMC7840205 DOI: 10.1002/vms3.331] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Revised: 04/03/2020] [Accepted: 07/13/2020] [Indexed: 01/11/2023] Open
Abstract
Mild, moderate and severe equine asthma is a problem for equine welfare. The aetiology of the disease is not known in detail but is likely multi‐factorial. One important factor may be inhaled dust which carries harmful substances which may be bioactivated and thus can lead to local inflammation in the airways. The aim of this study was to investigate gene expression and protein localisation of cytochrome P450 (CYP) enzymes, superoxide dismutase and glutathione‐S‐transferases (GST) involved in bioactivation and detoxification of harmful substances in lungs of horses with or without histological evidence of lower airway inflammation. Significantly lower gene expression of CYP2A13 and GSTM1 was observed in lungs from horses with histological evidence of lower airway inflammation compared with horses without. A higher expression, although not significant, was found for CYP1A1 in horses with histological evidence of lower airway inflammation. There were no differences in gene expression of GSTP1 and SOD3. The proteins were localised in the respiratory epithelium which is of relevance as a defence to local exposure of inhaled harmful substances. In conclusion, our study reports differential gene expression of enzymes involved in bioactivation and detoxification of foreign substances in the lungs of horses with histological evidence of lower airway inflammation compared with horses without.
Collapse
Affiliation(s)
- Maria Löfgren
- Department of Biomedical Sciences and Veterinary Public Health, Swedish University of Agricultural Sciences, Uppsala, Sweden
| | - Pia Larsson
- Department of Biomedical Sciences and Veterinary Public Health, Swedish University of Agricultural Sciences, Uppsala, Sweden
| | - Ronny Lindberg
- Department of Biomedical Sciences and Veterinary Public Health, Swedish University of Agricultural Sciences, Uppsala, Sweden
| | - Katarina Hörnaeus
- Department of Biomedical Sciences and Veterinary Public Health, Swedish University of Agricultural Sciences, Uppsala, Sweden
| | - Eva Tydén
- Department of Biomedical Sciences and Veterinary Public Health, Swedish University of Agricultural Sciences, Uppsala, Sweden
| |
Collapse
|
6
|
Yan Z, Spaulding HR. Extracellular superoxide dismutase, a molecular transducer of health benefits of exercise. Redox Biol 2020; 32:101508. [PMID: 32220789 PMCID: PMC7109453 DOI: 10.1016/j.redox.2020.101508] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 03/14/2020] [Accepted: 03/16/2020] [Indexed: 02/06/2023] Open
Abstract
Extracellular superoxide dismutase (EcSOD) is the only extracellular scavenger of superoxide anion (O2.-) with unique binding capacity to cell surface and extracellular matrix through its heparin-binding domain. Enhanced EcSOD activity prevents oxidative stress and damage, which are fundamental in a variety of disease pathologies. In this review we will discuss the findings in humans and animal studies supporting the benefits of EcSOD induced by exercise training in reducing oxidative stress in various tissues. In particularly, we will highlight the importance of skeletal muscle EcSOD, which is induced by endurance exercise and redistributed through the circulation to the peripheral tissues, as a molecular transducer of exercise training to confer protection against oxidative stress and damage in various disease conditions.
Collapse
Affiliation(s)
- Zhen Yan
- Center for Skeletal Muscle Research at Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA, 22908, USA; Department of Medicine, University of Virginia School of Medicine, Charlottesville, VA, 22908, USA; Department of Pharmacology, University of Virginia School of Medicine, Charlottesville, VA, 22908, USA; Department of Molecular Physiology and Biological Physics, University of Virginia School of Medicine, Charlottesville, VA, 22908, USA.
| | - Hannah R Spaulding
- Center for Skeletal Muscle Research at Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA, 22908, USA
| |
Collapse
|
7
|
Ma X, Wu Y, Zhang L, Yuan W, Yan L, Fan S, Lian Y, Zhu X, Gao J, Zhao J, Zhang P, Tang H, Jia W. Comparison and development of machine learning tools for the prediction of chronic obstructive pulmonary disease in the Chinese population. J Transl Med 2020; 18:146. [PMID: 32234053 PMCID: PMC7110698 DOI: 10.1186/s12967-020-02312-0] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Accepted: 03/17/2020] [Indexed: 02/08/2023] Open
Abstract
Background Chronic obstructive pulmonary disease (COPD) is a major public health problem and cause of mortality worldwide. However, COPD in the early stage is usually not recognized and diagnosed. It is necessary to establish a risk model to predict COPD development. Methods A total of 441 COPD patients and 192 control subjects were recruited, and 101 single-nucleotide polymorphisms (SNPs) were determined using the MassArray assay. With 5 clinical features as well as SNPs, 6 predictive models were established and evaluated in the training set and test set by the confusion matrix AU-ROC, AU-PRC, sensitivity (recall), specificity, accuracy, F1 score, MCC, PPV (precision) and NPV. The selected features were ranked. Results Nine SNPs were significantly associated with COPD. Among them, 6 SNPs (rs1007052, OR = 1.671, P = 0.010; rs2910164, OR = 1.416, P < 0.037; rs473892, OR = 1.473, P < 0.044; rs161976, OR = 1.594, P < 0.044; rs159497, OR = 1.445, P < 0.045; and rs9296092, OR = 1.832, P < 0.045) were risk factors for COPD, while 3 SNPs (rs8192288, OR = 0.593, P < 0.015; rs20541, OR = 0.669, P < 0.018; and rs12922394, OR = 0.651, P < 0.022) were protective factors for COPD development. In the training set, KNN, LR, SVM, DT and XGboost obtained AU-ROC values above 0.82 and AU-PRC values above 0.92. Among these models, XGboost obtained the highest AU-ROC (0.94), AU-PRC (0.97), accuracy (0.91), precision (0.95), F1 score (0.94), MCC (0.77) and specificity (0.85), while MLP obtained the highest sensitivity (recall) (0.99) and NPV (0.87). In the validation set, KNN, LR and XGboost obtained AU-ROC and AU-PRC values above 0.80 and 0.85, respectively. KNN had the highest precision (0.82), both KNN and LR obtained the same highest accuracy (0.81), and KNN and LR had the same highest F1 score (0.86). Both DT and MLP obtained sensitivity (recall) and NPV values above 0.94 and 0.84, respectively. In the feature importance analyses, we identified that AQCI, age, and BMI had the greatest impact on the predictive abilities of the models, while SNPs, sex and smoking were less important. Conclusions The KNN, LR and XGboost models showed excellent overall predictive power, and the use of machine learning tools combining both clinical and SNP features was suitable for predicting the risk of COPD development.
Collapse
Affiliation(s)
- Xia Ma
- Department of Pulmonary and Critical Care Medicine, General Hospital of Datong Coal Mine Group Co., Ltd., Datong, 037000, China.,Department of Pulmonary and Critical Care Medicine, The First Hospital of Shanxi Medical University, Taiyuan, 030001, China
| | - Yanping Wu
- Department of Respiratory, General Hospital of Tisco (Sixth Hospital of Shanxi Medical University), 2 Yingxin Street, Jiancaoping District, Taiyuan, 030008, Shanxi Province, China
| | - Ling Zhang
- Department of Respiratory, Linfen People's Hospital, Linfen, 041000, China
| | - Weilan Yuan
- Shanghai Biotecan Pharmaceuticals Co., Ltd., 180 Zhangheng Road, Shanghai, 201204, China.,Shanghai Zhangjiang Institute of Medical Innovation, Shanghai, 201204, China
| | - Li Yan
- Department of Respiratory Medicine, Hebei General Hospital, Shijiazhuang, 050000, China
| | - Sha Fan
- Department of Respiratory Medicine, Heji Hospital Affiliated with Changzhi Medical College, Changzhi, 046011, China
| | - Yunzhi Lian
- Department of Clinical Laboratory, JinCheng People's Hospital, Jincheng, 048000, China
| | - Xia Zhu
- Department of Respiratory, General Hospital of Tisco (Sixth Hospital of Shanxi Medical University), 2 Yingxin Street, Jiancaoping District, Taiyuan, 030008, Shanxi Province, China
| | - Junhui Gao
- Shanghai Biotecan Pharmaceuticals Co., Ltd., 180 Zhangheng Road, Shanghai, 201204, China.,Shanghai Zhangjiang Institute of Medical Innovation, Shanghai, 201204, China
| | - Jiangman Zhao
- Shanghai Biotecan Pharmaceuticals Co., Ltd., 180 Zhangheng Road, Shanghai, 201204, China.,Shanghai Zhangjiang Institute of Medical Innovation, Shanghai, 201204, China
| | - Ping Zhang
- Department of Clinical Laboratory, Linfen People's Hospital, West of Rainbow Bridge, West Binhe Road, Yaodu District, Linfen, 041000, Shanxi Province, China.
| | - Hui Tang
- Shanghai Biotecan Pharmaceuticals Co., Ltd., 180 Zhangheng Road, Shanghai, 201204, China. .,Shanghai Zhangjiang Institute of Medical Innovation, Shanghai, 201204, China.
| | - Weihua Jia
- Department of Respiratory, General Hospital of Tisco (Sixth Hospital of Shanxi Medical University), 2 Yingxin Street, Jiancaoping District, Taiyuan, 030008, Shanxi Province, China.
| |
Collapse
|
8
|
Allawzi A, McDermott I, Delaney C, Nguyen K, Banimostafa L, Trumpie A, Hernandez-Lagunas L, Riemondy K, Gillen A, Hesselberth J, El Kasmi K, Sucharov CC, Janssen WJ, Stenmark K, Bowler R, Nozik-Grayck E. Redistribution of EC-SOD resolves bleomycin-induced inflammation via increased apoptosis of recruited alveolar macrophages. FASEB J 2019; 33:13465-13475. [PMID: 31560857 PMCID: PMC6894081 DOI: 10.1096/fj.201901038rr] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Accepted: 08/26/2019] [Indexed: 01/16/2023]
Abstract
A human single nucleotide polymorphism (SNP) in the matrix-binding domain of extracellular superoxide dismutase (EC-SOD), with arginine to glycine substitution at position 213 (R213G), redistributes EC-SOD from the matrix into extracellular fluids. We reported that, following bleomycin (bleo), knockin mice harboring the human R213G SNP (R213G mice) exhibit enhanced resolution of inflammation and protection against fibrosis, compared with wild-type (WT) littermates. In this study, we tested the hypothesis that the EC-SOD R213G SNP promotes resolution via accelerated apoptosis of recruited alveolar macrophage (AM). RNA sequencing and Ingenuity Pathway Analysis 7 d postbleo in recruited AM implicated increased apoptosis and blunted inflammatory responses in the R213G strain exhibiting accelerated resolution. We validated that the percentage of apoptosis was significantly elevated in R213G recruited AM vs. WT at 3 and 7 d postbleo in vivo. Recruited AM numbers were also significantly decreased in R213G mice vs. WT at 3 and 7 d postbleo. ChaC glutathione-specific γ-glutamylcyclotransferase 1 (Chac1), a proapoptotic γ-glutamyl cyclotransferase that depletes glutathione, was increased in the R213G recruited AM. Overexpression of Chac1 in vitro induced apoptosis of macrophages and was blocked by administration of cell-permeable glutathione. In summary, we provide new evidence that redistributed EC-SOD accelerates the resolution of inflammation through redox-regulated mechanisms that increase recruited AM apoptosis.-Allawzi, A., McDermott, I., Delaney, C., Nguyen, K., Banimostafa, L., Trumpie, A., Hernandez-Lagunas, L., Riemondy, K., Gillen, A., Hesselberth, J., El Kasmi, K., Sucharov, C. C., Janssen, W. J., Stenmark, K., Bowler, R., Nozik-Grayck, E. Redistribution of EC-SOD resolves bleomycin-induced inflammation via increased apoptosis of recruited alveolar macrophages.
Collapse
Affiliation(s)
- Ayed Allawzi
- Cardiovascular Pulmonary Research Laboratories, Departments of Pediatrics and Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Ivy McDermott
- Cardiovascular Pulmonary Research Laboratories, Departments of Pediatrics and Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Cassidy Delaney
- Cardiovascular Pulmonary Research Laboratories, Departments of Pediatrics and Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Kianna Nguyen
- Cardiovascular Pulmonary Research Laboratories, Departments of Pediatrics and Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Laith Banimostafa
- Cardiovascular Pulmonary Research Laboratories, Departments of Pediatrics and Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Ashley Trumpie
- Cardiovascular Pulmonary Research Laboratories, Departments of Pediatrics and Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Laura Hernandez-Lagunas
- Cardiovascular Pulmonary Research Laboratories, Departments of Pediatrics and Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Kent Riemondy
- RNA Bioscience Initiative, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Austin Gillen
- RNA Bioscience Initiative, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Jay Hesselberth
- RNA Bioscience Initiative, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Karim El Kasmi
- Cardiovascular Pulmonary Research Laboratories, Departments of Pediatrics and Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
- Boehringer Ingelheim Pharma, Biberach, Germany
| | - Carmen C. Sucharov
- Division of Cardiology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA; and
| | | | - Kurt Stenmark
- Cardiovascular Pulmonary Research Laboratories, Departments of Pediatrics and Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Russell Bowler
- Department of Medicine, National Jewish Health, Denver, Colorado, USA
| | - Eva Nozik-Grayck
- Cardiovascular Pulmonary Research Laboratories, Departments of Pediatrics and Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| |
Collapse
|
9
|
Anti-oxidative effects of superoxide dismutase 3 on inflammatory diseases. J Mol Med (Berl) 2019; 98:59-69. [PMID: 31724066 DOI: 10.1007/s00109-019-01845-2] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 09/16/2019] [Accepted: 10/16/2019] [Indexed: 01/16/2023]
Abstract
Free radicals and other oxidants are critical determinants of the cellular signaling pathways involved in the pathogenesis of several human diseases including inflammatory diseases. Numerous studies have demonstrated the protective effects of antioxidant enzymes during inflammation by elimination of free radicals. The superoxide dismutase (SOD), an antioxidant enzyme, plays an essential pathogenic role in the inflammatory diseases by not only catalyzing the conversion of the superoxide to hydrogen peroxide and oxygen but also affecting immune responses. There are three distinct isoforms of SOD, which distribute in different cellular compartments such as cytosolic SOD1, mitochondrial SOD2, and extracellular SOD3. Many studies have investigated the anti-oxidative effects of SOD3 in the inflammatory diseases. Herein, in this review, we focus on the current understanding of SOD3 as a therapeutic protein in inflammatory diseases such as skin, autoimmune, lung, and cardiovascular inflammatory diseases. Moreover, the mechanism(s) by which SOD3 modulates immune responses and signal initiation in the pathogenesis of the diseases will be further discussed.
Collapse
|
10
|
New therapeutic targets for the prevention of infectious acute exacerbations of COPD: role of epithelial adhesion molecules and inflammatory pathways. Clin Sci (Lond) 2019; 133:1663-1703. [PMID: 31346069 DOI: 10.1042/cs20181009] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 06/27/2019] [Accepted: 06/28/2019] [Indexed: 12/15/2022]
Abstract
Chronic respiratory diseases are among the leading causes of mortality worldwide, with the major contributor, chronic obstructive pulmonary disease (COPD) accounting for approximately 3 million deaths annually. Frequent acute exacerbations (AEs) of COPD (AECOPD) drive clinical and functional decline in COPD and are associated with accelerated loss of lung function, increased mortality, decreased health-related quality of life and significant economic costs. Infections with a small subgroup of pathogens precipitate the majority of AEs and consequently constitute a significant comorbidity in COPD. However, current pharmacological interventions are ineffective in preventing infectious exacerbations and their treatment is compromised by the rapid development of antibiotic resistance. Thus, alternative preventative therapies need to be considered. Pathogen adherence to the pulmonary epithelium through host receptors is the prerequisite step for invasion and subsequent infection of surrounding structures. Thus, disruption of bacterial-host cell interactions with receptor antagonists or modulation of the ensuing inflammatory profile present attractive avenues for therapeutic development. This review explores key mediators of pathogen-host interactions that may offer new therapeutic targets with the potential to prevent viral/bacterial-mediated AECOPD. There are several conceptual and methodological hurdles hampering the development of new therapies that require further research and resolution.
Collapse
|
11
|
Zhou Y, Horowitz JC, Naba A, Ambalavanan N, Atabai K, Balestrini J, Bitterman PB, Corley RA, Ding BS, Engler AJ, Hansen KC, Hagood JS, Kheradmand F, Lin QS, Neptune E, Niklason L, Ortiz LA, Parks WC, Tschumperlin DJ, White ES, Chapman HA, Thannickal VJ. Extracellular matrix in lung development, homeostasis and disease. Matrix Biol 2018. [PMID: 29524630 DOI: 10.1016/j.matbio.2018.03.005] [Citation(s) in RCA: 188] [Impact Index Per Article: 26.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The lung's unique extracellular matrix (ECM), while providing structural support for cells, is critical in the regulation of developmental organogenesis, homeostasis and injury-repair responses. The ECM, via biochemical or biomechanical cues, regulates diverse cell functions, fate and phenotype. The composition and function of lung ECM become markedly deranged in pathological tissue remodeling. ECM-based therapeutics and bioengineering approaches represent promising novel strategies for regeneration/repair of the lung and treatment of chronic lung diseases. In this review, we assess the current state of lung ECM biology, including fundamental advances in ECM composition, dynamics, topography, and biomechanics; the role of the ECM in normal and aberrant lung development, adult lung diseases and autoimmunity; and ECM in the regulation of the stem cell niche. We identify opportunities to advance the field of lung ECM biology and provide a set recommendations for research priorities to advance knowledge that would inform novel approaches to the pathogenesis, diagnosis, and treatment of chronic lung diseases.
Collapse
Affiliation(s)
- Yong Zhou
- Division of Pulmonary, Allergy and Critical Care Medicine, University of Alabama at Birmingham, United States.
| | - Jeffrey C Horowitz
- Division of Pulmonary and Critical Care Medicine, University of Michigan, United States.
| | - Alexandra Naba
- Department of Physiology & Biophysics, University of Illinois at Chicago, United States.
| | | | - Kamran Atabai
- Lung Biology Center, University of California, San Francisco, United States.
| | | | | | - Richard A Corley
- Systems Toxicology & Exposure Science, Pacific Northwest National Laboratory, United States.
| | - Bi-Sen Ding
- Weill Cornell Medical College, United States.
| | - Adam J Engler
- Sanford Consortium for Regenerative Medicine, University of California, San Diego, United States.
| | - Kirk C Hansen
- Biochemistry & Molecular Genetics, University of Colorado Denver, United States.
| | - James S Hagood
- Pediatric Respiratory Medicine, University of California San Diego, United States.
| | - Farrah Kheradmand
- Division of Pulmonary and Critical Care, Baylor College of Medicine, United States.
| | - Qing S Lin
- Division of Lung Diseases, National Heart, Lung, and Blood Institute, United States.
| | - Enid Neptune
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins School of Medicine, United States.
| | - Laura Niklason
- Department of Anesthesiology, Yale University, United States.
| | - Luis A Ortiz
- Division of Environmental and Occupational Health, University of Pittsburgh, United States.
| | - William C Parks
- Department of Medicine, Cedars-Sinai Medical Center, United States.
| | - Daniel J Tschumperlin
- Department of Physiology & Biomedical Engineering, Mayo Clinic College of Medicine, United States.
| | - Eric S White
- Division of Pulmonary and Critical Care Medicine, University of Michigan, United States.
| | - Harold A Chapman
- Division of Pulmonary and Critical Care Medicine, University of California, San Francisco, United States.
| | - Victor J Thannickal
- Division of Pulmonary, Allergy and Critical Care Medicine, University of Alabama at Birmingham, United States.
| |
Collapse
|
12
|
Yeo J, Morales DA, Chen T, Crawford EL, Zhang X, Blomquist TM, Levin AM, Massion PP, Arenberg DA, Midthun DE, Mazzone PJ, Nathan SD, Wainz RJ, Nana-Sinkam P, Willey PFS, Arend TJ, Padda K, Qiu S, Federov A, Hernandez DAR, Hammersley JR, Yoon Y, Safi F, Khuder SA, Willey JC. RNAseq analysis of bronchial epithelial cells to identify COPD-associated genes and SNPs. BMC Pulm Med 2018; 18:42. [PMID: 29506519 PMCID: PMC5838965 DOI: 10.1186/s12890-018-0603-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Accepted: 02/23/2018] [Indexed: 01/09/2023] Open
Abstract
Background There is a need for more powerful methods to identify low-effect SNPs that contribute to hereditary COPD pathogenesis. We hypothesized that SNPs contributing to COPD risk through cis-regulatory effects are enriched in genes comprised by bronchial epithelial cell (BEC) expression patterns associated with COPD. Methods To test this hypothesis, normal BEC specimens were obtained by bronchoscopy from 60 subjects: 30 subjects with COPD defined by spirometry (FEV1/FVC < 0.7, FEV1% < 80%), and 30 non-COPD controls. Targeted next generation sequencing was used to measure total and allele-specific expression of 35 genes in genome maintenance (GM) genes pathways linked to COPD pathogenesis, including seven TP53 and CEBP transcription factor family members. Shrinkage linear discriminant analysis (SLDA) was used to identify COPD-classification models. COPD GWAS were queried for putative cis-regulatory SNPs in the targeted genes. Results On a network basis, TP53 and CEBP transcription factor pathway gene pair network connections, including key DNA repair gene ERCC5, were significantly different in COPD subjects (e.g., Wilcoxon rank sum test for closeness, p-value = 5.0E-11). ERCC5 SNP rs4150275 association with chronic bronchitis was identified in a set of Lung Health Study (LHS) COPD GWAS SNPs restricted to those in putative regulatory regions within the targeted genes, and this association was validated in the COPDgene non-hispanic white (NHW) GWAS. ERCC5 SNP rs4150275 is linked (D’ = 1) to ERCC5 SNP rs17655 which displayed differential allelic expression (DAE) in BEC and is an expression quantitative trait locus (eQTL) in lung tissue (p = 3.2E-7). SNPs in linkage (D’ = 1) with rs17655 were predicted to alter miRNA binding (rs873601). A classifier model that comprised gene features CAT, CEBPG, GPX1, KEAP1, TP73, and XPA had pooled 10-fold cross-validation receiver operator characteristic area under the curve of 75.4% (95% CI: 66.3%–89.3%). The prevalence of DAE was higher than expected (p = 0.0023) in the classifier genes. Conclusions GM genes comprised by COPD-associated BEC expression patterns were enriched for SNPs with cis-regulatory function, including a putative cis-rSNP in ERCC5 that was associated with COPD risk. These findings support additional total and allele-specific expression analysis of gene pathways with high prior likelihood for involvement in COPD pathogenesis. Electronic supplementary material The online version of this article (10.1186/s12890-018-0603-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jiyoun Yeo
- Department of Pathology, The University of Toledo College of Medicine, 3000 Arlington Avenue, HEB 219, Toledo, OH, 43614, USA
| | - Diego A Morales
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, The University of Toledo College of Medicine, 3000 Arlington Avenue, HEB 219, Toledo, OH, 43614, USA
| | - Tian Chen
- Department of Mathematics and Statistics, The University of Toledo, 2801 W. Bancroft Street, Toledo, OH, 43606, USA
| | - Erin L Crawford
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, The University of Toledo College of Medicine, 3000 Arlington Avenue, HEB 219, Toledo, OH, 43614, USA
| | - Xiaolu Zhang
- Department of Medicine, The University of Toledo College of Medicine, 3000 Arlington Avenue, Toledo, OH, 43614, USA
| | - Thomas M Blomquist
- Department of Pathology, The University of Toledo College of Medicine, 3000 Arlington Avenue, HEB 219, Toledo, OH, 43614, USA
| | - Albert M Levin
- Department of Biostatistics, Henry Ford Health System, 1 Ford Place Detroit, MI, Detroit, MI, 48202, USA
| | - Pierre P Massion
- Thoracic Program, Vanderbilt Ingram Cancer Center, Nashville, TN, 37232, USA
| | | | - David E Midthun
- Department of Pulmonary and Critical Care Medicine, Mayo Clinic, 200 1st St SW, Rochester, MN, 55905, USA
| | - Peter J Mazzone
- Department of Pulmonary Medicine, Cleveland Clinic, 9500 Euclid Ave, Cleveland, OH, 44195, USA
| | - Steven D Nathan
- Department of Pulmonary Medicine, Inova Fairfax Hospital, 3300 Gallows Road, Falls Church, VA, 22042-3300, USA
| | - Ronald J Wainz
- The Toledo Hospital, 2142 N Cove Blvd, Toledo, OH, 43606, USA
| | - Patrick Nana-Sinkam
- Division of Pulmonary Diseases and Critical Care Medicine, Virginia Commonwealth University, USA, Richmond, VA, 23284-2512, USA.,Ohio State University James Comprehensive Cancer Center and Solove Research Institute, Columbus, OH, USA
| | - Paige F S Willey
- American Enterprise Institute, 1789 Massachusetts Ave NW, Washington, DC, 20036, USA
| | - Taylor J Arend
- The University of Toledo College of Medicine, 3000 Arlington Avenue, Toledo, OH, 43614, USA
| | - Karanbir Padda
- Emory University School of Medicine, 1648 Pierce Dr NE, Atlanta, GA, 30307, USA
| | - Shuhao Qiu
- Department of Medicine, The University of Toledo Medical Center, 3000 Arlington Avenue, Toledo, OH, 43614, USA
| | - Alexei Federov
- Department of Mathematics and Statistics, The University of Toledo, 2801 W. Bancroft Street, Toledo, OH, 43606, USA.,Department of Medicine, The University of Toledo College of Medicine, 3000 Arlington Avenue, Toledo, OH, 43614, USA
| | - Dawn-Alita R Hernandez
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, The University of Toledo College of Medicine, 3000 Arlington Avenue, RHC 0012, Toledo, OH, 43614, USA
| | - Jeffrey R Hammersley
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, The University of Toledo College of Medicine, 3000 Arlington Avenue, RHC 0012, Toledo, OH, 43614, USA
| | - Youngsook Yoon
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, The University of Toledo College of Medicine, 3000 Arlington Avenue, RHC 0012, Toledo, OH, 43614, USA
| | - Fadi Safi
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, The University of Toledo College of Medicine, 3000 Arlington Avenue, RHC 0012, Toledo, OH, 43614, USA
| | - Sadik A Khuder
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, The University of Toledo College of Medicine, 3000 Arlington Avenue, RHC 0012, Toledo, OH, 43614, USA
| | - James C Willey
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, The University of Toledo College of Medicine, 3000 Arlington Avenue, Toledo, OH, 43614, USA.
| |
Collapse
|
13
|
Aghapour M, Raee P, Moghaddam SJ, Hiemstra PS, Heijink IH. Airway Epithelial Barrier Dysfunction in Chronic Obstructive Pulmonary Disease: Role of Cigarette Smoke Exposure. Am J Respir Cell Mol Biol 2018; 58:157-169. [DOI: 10.1165/rcmb.2017-0200tr] [Citation(s) in RCA: 142] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Affiliation(s)
| | - Pourya Raee
- Department of Basic Sciences, Faculty of Veterinary Medicine, University of Tabriz, Tabriz, Iran
| | - Seyed Javad Moghaddam
- Department of Pulmonary Medicine, Division of Internal Medicine, the University of Texas M. D. Anderson Cancer Center, Houston, Texas
| | - Pieter S. Hiemstra
- Department of Pulmonology, Leiden University Medical Center, Leiden, the Netherlands; and
| | - Irene H. Heijink
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| |
Collapse
|
14
|
Hecker L. Mechanisms and consequences of oxidative stress in lung disease: therapeutic implications for an aging populace. Am J Physiol Lung Cell Mol Physiol 2017; 314:L642-L653. [PMID: 29351446 DOI: 10.1152/ajplung.00275.2017] [Citation(s) in RCA: 99] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The rapid expansion of the elderly population has led to the recent epidemic of age-related diseases, including increased incidence and mortality of chronic and acute lung diseases. Numerous studies have implicated aging and oxidative stress in the pathogenesis of various pulmonary diseases; however, despite recent advances in these fields, the specific contributions of aging and oxidative stress remain elusive. This review will discuss the consequences of aging on lung morphology and physiology, and how redox imbalance with aging contributes to lung disease susceptibility. Here, we focus on three lung diseases for which aging is a significant risk factor: acute respiratory distress syndrome (ARDS), chronic obstructive pulmonary disease (COPD), and idiopathic pulmonary fibrosis (IPF). Preclinical and clinical development for redox- and senescence-altering therapeutic strategies are discussed, as well as scientific advancements that may direct current and future therapeutic development. A deeper understanding of how aging impacts normal lung function, redox balance, and injury-repair processes will inspire the development of new therapies to prevent and/or reverse age-associated pulmonary diseases, and ultimately increase health span and longevity. This review is intended to encourage basic, clinical, and translational research that will bridge knowledge gaps at the intersection of aging, oxidative stress, and lung disease to fuel the development of more effective therapeutic strategies for lung diseases that disproportionately afflict the elderly.
Collapse
Affiliation(s)
- Louise Hecker
- Division of Pulmonary, Allergy and Critical Care and Sleep Medicine, University of Arizona , Tucson, Arizona and Southern Arizona Veterans Affairs Health Care System, Tucson, Arizona
| |
Collapse
|
15
|
Call JA, Donet J, Martin KS, Sharma AK, Chen X, Zhang J, Cai J, Galarreta CA, Okutsu M, Du Z, Lira VA, Zhang M, Mehrad B, Annex BH, Klibanov AL, Bowler RP, Laubach VE, Peirce SM, Yan Z. Muscle-derived extracellular superoxide dismutase inhibits endothelial activation and protects against multiple organ dysfunction syndrome in mice. Free Radic Biol Med 2017; 113:212-223. [PMID: 28982599 PMCID: PMC5740866 DOI: 10.1016/j.freeradbiomed.2017.09.029] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Revised: 09/22/2017] [Accepted: 09/28/2017] [Indexed: 12/20/2022]
Abstract
Multiple organ dysfunction syndrome (MODS) is a detrimental clinical complication in critically ill patients with high mortality. Emerging evidence suggests that oxidative stress and endothelial activation (induced expression of adhesion molecules) of vital organ vasculatures are key, early steps in the pathogenesis. We aimed to ascertain the role and mechanism(s) of enhanced extracellular superoxide dismutase (EcSOD) expression in skeletal muscle in protection against MODS induced by endotoxemia. We showed that EcSOD overexpressed in skeletal muscle-specific transgenic mice (TG) redistributes to other peripheral organs through the circulation and enriches at the endothelium of the vasculatures. TG mice are resistant to endotoxemia (induced by lipopolysaccharide [LPS] injection) in developing MODS with significantly reduced mortality and organ damages compared with the wild type littermates (WT). Heterogenic parabiosis between TG and WT mice conferred a significant protection to WT mice, whereas mice with R213G knock-in mutation, a human single nucleotide polymorphism leading to reduced binding EcSOD in peripheral organs, exacerbated the organ damages. Mechanistically, EcSOD inhibits vascular cell adhesion molecule 1 expression and inflammatory leukocyte adhesion to the vascular wall of vital organs, blocking an early step of the pathology in organ damage under endotoxemia. Therefore, enhanced expression of EcSOD in skeletal muscle profoundly protects against MODS by inhibiting endothelial activation and inflammatory cell adhesion, which could be a promising therapy for MODS.
Collapse
Affiliation(s)
- Jarrod A Call
- Center for Skeletal Muscle Research at Robert Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA 22908, USA; Departments of Medicine, University of Virginia, Charlottesville, VA 22908, USA
| | - Jean Donet
- Center for Skeletal Muscle Research at Robert Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA 22908, USA; Departments of Medicine, University of Virginia, Charlottesville, VA 22908, USA
| | - Kyle S Martin
- Departments of Biomedical Engineering, University of Virginia, Charlottesville, VA 22908, USA
| | - Ashish K Sharma
- Departments of Surgery, University of Virginia, Charlottesville, VA 22908, USA
| | - Xiaobin Chen
- Center for Skeletal Muscle Research at Robert Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA 22908, USA; Department of Cardiology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan Province 410008, China
| | - Jiuzhi Zhang
- Center for Skeletal Muscle Research at Robert Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA 22908, USA; Department of Critical Care Medicine and Institute of Critical Care Medicine, First Affiliate Hospital of Dalian Medical University, 222 Zhongshan Road, Dalian, Liaoning Province 116011, China
| | - Jie Cai
- Center for Skeletal Muscle Research at Robert Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA 22908, USA; Department of Infectious Disease, First Affiliate Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, Jiangsu Province 210029, China
| | - Carolina A Galarreta
- Departments of Pediatrics, University of Virginia, Charlottesville, VA 22908, USA
| | - Mitsuharu Okutsu
- Center for Skeletal Muscle Research at Robert Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA 22908, USA; Departments of Medicine, University of Virginia, Charlottesville, VA 22908, USA
| | - Zhongmin Du
- Departments of Medicine, University of Virginia, Charlottesville, VA 22908, USA
| | - Vitor A Lira
- Center for Skeletal Muscle Research at Robert Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA 22908, USA; Departments of Medicine, University of Virginia, Charlottesville, VA 22908, USA
| | - Mei Zhang
- Center for Skeletal Muscle Research at Robert Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA 22908, USA; Departments of Medicine, University of Virginia, Charlottesville, VA 22908, USA
| | - Borna Mehrad
- Departments of Medicine, University of Virginia, Charlottesville, VA 22908, USA
| | - Brian H Annex
- Departments of Medicine, University of Virginia, Charlottesville, VA 22908, USA
| | | | - Russell P Bowler
- Division of Pulmonary Medicine, Department of Medicine, National Jewish Health, Denver, CO, USA
| | - Victor E Laubach
- Departments of Surgery, University of Virginia, Charlottesville, VA 22908, USA
| | - Shayn M Peirce
- Departments of Biomedical Engineering, University of Virginia, Charlottesville, VA 22908, USA
| | - Zhen Yan
- Center for Skeletal Muscle Research at Robert Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA 22908, USA; Departments of Medicine, University of Virginia, Charlottesville, VA 22908, USA; Departments of Pharmacology, University of Virginia, Charlottesville, VA 22908, USA; Departments of Molecular Physiology & Biological Physics, University of Virginia, Charlottesville, VA 22908, USA.
| |
Collapse
|
16
|
Ota F, Kizuka Y, Nakano M, Yamaguchi Y, Kitazume S, Ookawara T, Taniguchi N. Sialylation of extracellular superoxide dismutase (EC-SOD) enhances furin-mediated cleavage and secretion. Glycobiology 2017; 27:1081-1088. [PMID: 29029079 DOI: 10.1093/glycob/cwx087] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Accepted: 09/30/2017] [Indexed: 01/12/2023] Open
Abstract
Extracellular superoxide dismutase (EC-SOD, SOD3) protects tissues against oxidative damage by detoxifying superoxide anions, particularly in the lungs and cardiovascular system. EC-SOD undergoes several posttranslational modifications including N-glycosylation and proteolytic cleavage. While the roles of proteolytic cleavage have been well studied, the structure and function of EC-SOD N-glycans are poorly understood. Here we analyzed glycan structures on native EC-SOD purified from human sera, and identified sialylated biantennary structures. Using glycan maturation-defective CHO mutant cells, we further revealed that the presence of terminal sialic acids in the N-glycans of EC-SOD enhanced both the secretion and furin-mediated C-terminal cleavage of EC-SOD. These results provide new insights into how the posttranslational modifications of EC-SOD control its functions.
Collapse
Affiliation(s)
- Fumi Ota
- Disease Glycomics Team, Systems Glycobiology Research Group, Global Research Cluster, RIKEN, Wako, Saitama 351-0198, Japan
| | - Yasuhiko Kizuka
- Disease Glycomics Team, Systems Glycobiology Research Group, Global Research Cluster, RIKEN, Wako, Saitama 351-0198, Japan
| | - Miyako Nakano
- Graduate School of Advanced Sciences of Matter, Hiroshima University, Higashihiroshima, Hiroshima 739-8530, Japan
| | - Yoshiki Yamaguchi
- Structural Glycobiology Team, Global Research Cluster, RIKEN, Wako, Saitama 351-0198, Japan
| | - Shinobu Kitazume
- Disease Glycomics Team, Systems Glycobiology Research Group, Global Research Cluster, RIKEN, Wako, Saitama 351-0198, Japan
| | - Tomomi Ookawara
- Laboratory of Biochemistry, School of Pharmacy, Hyogo University of Health Sciences, Kobe, Hyogo 650-8530, Japan
| | - Naoyuki Taniguchi
- Disease Glycomics Team, Systems Glycobiology Research Group, Global Research Cluster, RIKEN, Wako, Saitama 351-0198, Japan
| |
Collapse
|
17
|
On the Origin of Superoxide Dismutase: An Evolutionary Perspective of Superoxide-Mediated Redox Signaling. Antioxidants (Basel) 2017; 6:antiox6040082. [PMID: 29084153 PMCID: PMC5745492 DOI: 10.3390/antiox6040082] [Citation(s) in RCA: 91] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Revised: 10/23/2017] [Accepted: 10/27/2017] [Indexed: 12/15/2022] Open
Abstract
The field of free radical biology originated with the discovery of superoxide dismutase (SOD) in 1969. Over the last 5 decades, a plethora of research has been performed in species ranging from bacteria to mammals that has elucidated the molecular reaction, subcellular location, and specific isoforms of SOD. However, while humans have only begun to study this class of enzymes over the past 50 years, it has been estimated that these enzymes have existed for billions of years, and may be some of the original enzymes found in primitive life. As life evolved over this expanse of time, these enzymes have taken on new and different functional roles potentially in contrast to how they were originally derived. Herein, examination of the evolutionary history of these enzymes provides both an explanation and further inquiries into the modern-day role of SOD in physiology and disease.
Collapse
|
18
|
Peh HY, Tan WSD, Chan TK, Pow CW, Foster PS, Wong WSF. Vitamin E isoform γ-tocotrienol protects against emphysema in cigarette smoke-induced COPD. Free Radic Biol Med 2017; 110:332-344. [PMID: 28684161 DOI: 10.1016/j.freeradbiomed.2017.06.023] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Revised: 06/12/2017] [Accepted: 06/30/2017] [Indexed: 12/22/2022]
Abstract
Inflammation and oxidative stress contribute to emphysema in COPD. Although corticosteroids are the standard of care for COPD, they do not reduce oxidative stress, and a subset of patients is steroid-resistant. Vitamin E isoform γ-tocotrienol possesses both anti-inflammatory and anti-oxidative properties that may protect against emphysema. We aimed to establish the therapeutic potential of γ-tocotrienol in cigarette smoke-induced COPD models in comparison with prednisolone. BALB/c mice were exposed to cigarette smoke for 2 weeks or 2 months. γ-Tocotrienol and prednisolone were given orally. Bronchoalveolar lavage (BAL) fluid and lung tissues were assessed for inflammation, oxidative damage, and regulation of transcription factor activities. Emphysema and lung function were also evaluated. γ-Tocotrienol dose-dependently reduced cigarette smoke-induced BAL fluid neutrophil counts and levels of cytokines, chemokines and oxidative damage biomarkers, and pulmonary pro-inflammatory and pro-oxidant gene expression, but restored lung endogenous antioxidant activities. γ-Tocotrienol acted by inhibiting nuclear translocation of STAT3 and NF-κB, and up-regulating Nrf2 activation in the lungs. In mice exposed to 2-month cigarette smoke, γ-tocotrienol ameliorated bronchial epithelium thickening and destruction of alveolar sacs in lungs, and improved lung functions. In comparison with prednisolone, γ-tocotrienol demonstrated better anti-oxidative efficacy, and protection against emphysema and lung function in COPD. We revealed for the first time the anti-inflammatory and antioxidant efficacies of γ-tocotrienol in cigarette smoke-induced COPD models. In addition, γ-tocotrienol was able to attenuate emphysematous lesions and improve lung function in COPD. γ-Tocotrienol may have therapeutic potential for the treatment of COPD.
Collapse
Affiliation(s)
- Hong Yong Peh
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University Health System, Singapore; Immunology Program, Life Science Institute, National University of Singapore, Singapore
| | - W S Daniel Tan
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University Health System, Singapore; Immunology Program, Life Science Institute, National University of Singapore, Singapore
| | - Tze Khee Chan
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University Health System, Singapore; Singapore-MIT Alliance for Research and Technology (SMART), National University of Singapore, Singapore
| | - Chen Wei Pow
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University Health System, Singapore
| | - Paul S Foster
- The Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and The University of Newcastle, Newcastle, New South Wales, Australia
| | - W S Fred Wong
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University Health System, Singapore; Immunology Program, Life Science Institute, National University of Singapore, Singapore.
| |
Collapse
|
19
|
Nørskov MS, Dahl M, Tybjærg-Hansen A. Genetic Variation in GSTP1, Lung Function, Risk of Lung Cancer, and Mortality. J Thorac Oncol 2017; 12:1664-1672. [PMID: 28739440 DOI: 10.1016/j.jtho.2017.07.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Revised: 06/11/2017] [Accepted: 07/06/2017] [Indexed: 12/17/2022]
Abstract
INTRODUCTION Glutathione S-transferase pi 1 metabolizes carcinogens from tobacco smoke in the lung. We tested whether genetically altered glutathione S-transferase pi 1 activity affects lung function and risk for tobacco-related cancer and mortality in the general population. METHODS We genotyped 66,069 individuals from the white general population for two common functional variants in the glutathione S-transferase pi 1 gene (GSTP1)-amino acid isoleucine 105 changed to a valine (Ile105Val) and amino acid alanine 114 changed to a valine (Ala114Val)-and recorded lung function, lung cancer, tobacco-related cancer, and death as outcomes. RESULTS Lung function was increased stepwise with the Ile105Val genotype overall (p < 0.01) and among smokers separately (p < 0.01). Adjusted hazard ratios for lung cancer, tobacco-related cancer, and death were reduced stepwise with the Ile105Val genotype (p < 0.02): Ile105Val homozygotes and heterozygotes versus noncarriers had hazard ratios for lung cancer of 0.64 (0.47-0.89) and 0.93 (0.78-1.11), for tobacco-related cancer of 0.74 (0.60-0.92) and 0.92 (0.81-1.04), and hazard ratios for death of 0.87 (0.80-0.95) and 0.94 (0.89-0.99), respectively. Population prevented fractions of lung cancer, tobacco-related cancer, and death due to Ile105Val homozygosity were 4%, 3% and 2%, respectively. The Ala114Val genotype was associated with reduced mortality (p < 0.01) but not with lung function, lung cancer, or tobacco-related cancer. CONCLUSION GSTP1 Ile105Val was associated with increased lung function, reduced risk for lung cancer and tobacco-related cancer, and reduced all-cause mortality in the general population.
Collapse
Affiliation(s)
- Marianne S Nørskov
- Department of Clinical Biochemistry, Rigshospitalet, Copenhagen, Denmark; Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Morten Dahl
- Department of Clinical Biochemistry, Rigshospitalet, Copenhagen, Denmark; Faculty of Health and Medical Sciences, University of Copenhagen, Denmark; Department of Clinical Biochemistry, Zealand University Hospital, Køge, Denmark
| | - Anne Tybjærg-Hansen
- Department of Clinical Biochemistry, Rigshospitalet, Copenhagen, Denmark; Faculty of Health and Medical Sciences, University of Copenhagen, Denmark; The Copenhagen City Heart Study, Frederiksberg Hospital, Copenhagen, Denmark; The Copenhagen General Population Study, Department of Clinical Biochemistry, Herlev Hospital, Herlev, Denmark.
| |
Collapse
|
20
|
COPD and stroke: are systemic inflammation and oxidative stress the missing links? Clin Sci (Lond) 2017; 130:1039-50. [PMID: 27215677 PMCID: PMC4876483 DOI: 10.1042/cs20160043] [Citation(s) in RCA: 119] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2016] [Accepted: 03/07/2016] [Indexed: 12/12/2022]
Abstract
Chronic obstructive pulmonary disease (COPD) is characterized by progressive airflow limitation and loss of lung function, and is currently the third largest cause of death in the world. It is now well established that cardiovascular-related comorbidities such as stroke contribute to morbidity and mortality in COPD. The mechanisms linking COPD and stroke remain to be fully defined but are likely to be interconnected. The association between COPD and stroke may be largely dependent on shared risk factors such as aging and smoking, or the association of COPD with traditional stroke risk factors. In addition, we propose that COPD-related systemic inflammation and oxidative stress may play important roles by promoting cerebral vascular dysfunction and platelet hyperactivity. In this review, we briefly discuss the pathogenesis of COPD, acute exacerbations of COPD (AECOPD) and cardiovascular comorbidities associated with COPD, in particular stroke. We also highlight and discuss the potential mechanisms underpinning the link between COPD and stroke, with a particular focus on the roles of systemic inflammation and oxidative stress.
Collapse
|
21
|
McGuinness AJA, Sapey E. Oxidative Stress in COPD: Sources, Markers, and Potential Mechanisms. J Clin Med 2017; 6:jcm6020021. [PMID: 28212273 PMCID: PMC5332925 DOI: 10.3390/jcm6020021] [Citation(s) in RCA: 133] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Revised: 02/01/2017] [Accepted: 02/05/2017] [Indexed: 12/13/2022] Open
Abstract
Markers of oxidative stress are increased in chronic obstructive pulmonary disease (COPD) and reactive oxygen species (ROS) are able to alter biological molecules, signaling pathways and antioxidant molecule function, many of which have been implicated in the pathogenesis of COPD. However, the involvement of ROS in the development and progression of COPD is not proven. Here, we discuss the sources of ROS, and the defences that have evolved to protect against their harmful effects. We address the role that ROS may have in the development and progression of COPD, as well as current therapeutic attempts at limiting the damage they cause. Evidence has indicated that the function of several key cells appears altered in COPD patients, and expression levels of important oxidant and antioxidant molecules may be abnormal. Therapeutic trials attempting to restore equilibrium to these molecules have not impacted upon all facets of disease and whilst the theory behind ROS influence in COPD appears sound, current models testing relevant pathways to tissue damage are limited. The heterogeneity seen in COPD patients presents a challenge to our understanding, and further research is essential to identify potential targets and stratified COPD patient populations where ROS therapies may be maximally efficacious.
Collapse
Affiliation(s)
| | - Elizabeth Sapey
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham B15 2TT, UK.
| |
Collapse
|
22
|
Jun S, Dory L. Allele-Specific Effects on Extracellular Superoxide Dismutase Synthesis and Secretion. ACTA ACUST UNITED AC 2017. [DOI: 10.4236/jbise.2017.104011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
23
|
Ota F, Kizuka Y, Kitazume S, Adachi T, Taniguchi N. N-Glycosylation is essential for the secretion of extracellular superoxide dismutase. FEBS Lett 2016; 590:3357-3367. [PMID: 27567024 DOI: 10.1002/1873-3468.12378] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Revised: 08/22/2016] [Accepted: 08/23/2016] [Indexed: 01/25/2023]
Abstract
Extracellular superoxide dismutase (EC-SOD or SOD3) protects against various oxidative stress-related diseases by scavenging reactive superoxides in the extracellular space. It is the only SOD isozyme that is secreted and glycosylated (at asparagine 89). However, the physiological roles of its glycosylation are poorly understood. In this study, we found that the glycosylation site on EC-SOD is well conserved and that a glycosylation-deficient EC-SOD mutant retains its enzymatic activity, but is not secreted. This impairment in secretion may, in part, be due to the ability of the mutants to form unusual higher order oligomers. Our findings reveal that the glycan modification is a key regulator of EC-SOD secretion and contributes to the understanding of the roles of glycans in EC-SOD-related diseases.
Collapse
Affiliation(s)
- Fumi Ota
- Systems Glycobiology Research Group, RIKEN-Max Plank Joint Research Center for Systems Chemical Biology, Global Research Cluster, RIKEN, Wako, Saitama, Japan
| | - Yasuhiko Kizuka
- Systems Glycobiology Research Group, RIKEN-Max Plank Joint Research Center for Systems Chemical Biology, Global Research Cluster, RIKEN, Wako, Saitama, Japan
| | - Shinobu Kitazume
- Systems Glycobiology Research Group, RIKEN-Max Plank Joint Research Center for Systems Chemical Biology, Global Research Cluster, RIKEN, Wako, Saitama, Japan
| | - Tetsuo Adachi
- Laboratory of Clinical Pharmaceutics, Gifu Pharmaceutical University, Gifu, Gifu, Japan
| | - Naoyuki Taniguchi
- Systems Glycobiology Research Group, RIKEN-Max Plank Joint Research Center for Systems Chemical Biology, Global Research Cluster, RIKEN, Wako, Saitama, Japan.
| |
Collapse
|
24
|
Nozik-Grayck E, Woods C, Stearman RS, Venkataraman S, Ferguson BS, Swain K, Bowler RP, Geraci MW, Ihida-Stansbury K, Stenmark KR, McKinsey TA, Domann FE. Histone deacetylation contributes to low extracellular superoxide dismutase expression in human idiopathic pulmonary arterial hypertension. Am J Physiol Lung Cell Mol Physiol 2016; 311:L124-34. [PMID: 27233998 PMCID: PMC4967185 DOI: 10.1152/ajplung.00263.2015] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Accepted: 05/17/2016] [Indexed: 01/14/2023] Open
Abstract
Epigenetic mechanisms, including DNA methylation and histone acetylation, regulate gene expression in idiopathic pulmonary arterial hypertension (IPAH). These mechanisms can modulate expression of extracellular superoxide dismutase (SOD3 or EC-SOD), a key vascular antioxidant enzyme, and loss of vascular SOD3 worsens outcomes in animal models of pulmonary arterial hypertension. We hypothesized that SOD3 gene expression is decreased in patients with IPAH due to aberrant DNA methylation and/or histone deacetylation. We used lung tissue and pulmonary artery smooth muscle cells (PASMC) from subjects with IPAH at transplantation and from failed donors (FD). Lung SOD3 mRNA expression and activity was decreased in IPAH vs. FD. In contrast, mitochondrial SOD (Mn-SOD or SOD2) protein expression was unchanged and intracellular SOD activity was unchanged. Using bisulfite sequencing in genomic lung or PASMC DNA, we found the methylation status of the SOD3 promoter was similar between FD and IPAH. Furthermore, treatment with 5-aza-2'-deoxycytidine did not increase PASMC SOD3 mRNA, suggesting DNA methylation was not responsible for PASMC SOD3 expression. Though total histone deacetylase (HDAC) activity, histone acetyltransferase (HAT) activity, acetylated histones, and acetylated SP1 were similar between IPAH and FD, treatment with two selective class I HDAC inhibitors increased SOD3 only in IPAH PASMC. Class I HDAC3 siRNA also increased SOD3 expression. Trichostatin A, a pan-HDAC inhibitor, decreased proliferation in IPAH, but not in FD PASMC. These data indicate that histone deacetylation, specifically via class I HDAC3, decreases SOD3 expression in PASMC and HDAC inhibitors may protect IPAH in part by increasing PASMC SOD3 expression.
Collapse
Affiliation(s)
- Eva Nozik-Grayck
- Department of Pediatrics, University of Colorado Anschutz Medical Center, Aurora, Colorado; Cardiovascular Pulmonary Research, University of Colorado Anschutz Medical Center, Aurora, Colorado;
| | - Crystal Woods
- Department of Pediatrics, University of Colorado Anschutz Medical Center, Aurora, Colorado; Cardiovascular Pulmonary Research, University of Colorado Anschutz Medical Center, Aurora, Colorado
| | - Robert S Stearman
- Department of Medicine, Indiana University, Indianapolis, Indiana; and
| | - Sujatha Venkataraman
- Department of Pediatrics, University of Colorado Anschutz Medical Center, Aurora, Colorado
| | - Bradley S Ferguson
- Department of Medicine, University of Colorado Anschutz Medical Center, Aurora, Colorado
| | - Kalin Swain
- Department of Pediatrics, University of Colorado Anschutz Medical Center, Aurora, Colorado; Cardiovascular Pulmonary Research, University of Colorado Anschutz Medical Center, Aurora, Colorado
| | - Russell P Bowler
- Department of Medicine, National Jewish Hospital, Denver, Colorado
| | - Mark W Geraci
- Department of Medicine, Indiana University, Indianapolis, Indiana; and
| | - Kaori Ihida-Stansbury
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Kurt R Stenmark
- Department of Pediatrics, University of Colorado Anschutz Medical Center, Aurora, Colorado; Cardiovascular Pulmonary Research, University of Colorado Anschutz Medical Center, Aurora, Colorado; Department of Medicine, University of Colorado Anschutz Medical Center, Aurora, Colorado
| | - Timothy A McKinsey
- Department of Medicine, University of Colorado Anschutz Medical Center, Aurora, Colorado
| | | |
Collapse
|
25
|
Margelidon-Cozzolino V, Chbini K, Freymond N, Devouassoux G, Belaaouaj A, Pacheco Y. [COPD: An early disease]. REVUE DE PNEUMOLOGIE CLINIQUE 2016; 72:49-60. [PMID: 26657351 PMCID: PMC7126852 DOI: 10.1016/j.pneumo.2015.08.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/02/2014] [Accepted: 08/16/2015] [Indexed: 05/04/2023]
Abstract
This general review deals with the mechanisms which underlie the genetic factors in COPD. Many cellular and biochemical mechanisms occur in bronchial inflammation. We present the experimental models of COPD, insisting on the importance of oxydative stress, and on recent knowledge about the lung microbiome. Starting from this pathophysiology basis, we show how various genetic targets are able to interfere with the disease model. Thanks to these genetic targets, new markers in exhaled breath condensates and new drug targets are rising.
Collapse
Affiliation(s)
- V Margelidon-Cozzolino
- Service de pneumologie A, centre hospitalier de Lyon Sud, hospices civils de Lyon, faculté de médecine, université Claude-Bernard Lyon 1, 69310 Pierre-Bénite, France.
| | - K Chbini
- Service de cardiologie, CHU Mohammed VI, faculté de médecine et de pharmacie, université Cadi Ayyad, Marrakech, Maroc
| | - N Freymond
- Service de pneumologie A, centre hospitalier de Lyon Sud, hospices civils de Lyon, 69310 Pierre-Bénite, France
| | - G Devouassoux
- Service de pneumologie, hôpital de la Croix Rousse, hospices civils de Lyon, faculté de médecine Lyon Sud, université Claude-Bernard Lyon 1, 69005 Lyon, France
| | - A Belaaouaj
- Inserm 1111, faculté de médecine Lyon Sud, chemin du Grand-Revoyet, 69310 Pierre-Bénite, France
| | - Y Pacheco
- Service de pneumologie A, centre hospitalier de Lyon Sud, hospices civils de Lyon, faculté de médecine Lyon Sud, université Claude-Bernard Lyon 1, 69310 Pierre-Bénite, France
| |
Collapse
|
26
|
Zhang SJ, Jiang JX, Ren QQ, Xie QM, Xiong YK. Effects of the inhalation of the m3 receptor antagonist bencycloquidium bromide in a mouse cigarette smoke-induced airway inflammation model. Drug Dev Res 2015; 76:123-31. [PMID: 25958838 DOI: 10.1002/ddr.21248] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2015] [Accepted: 02/28/2015] [Indexed: 11/09/2022]
Abstract
Bencycloquidium bromide (BCQB), a novel M3 receptor antagonist, alleviates airway hyperresponsiveness, inflammation, and airway remodeling in a murine model of asthma. The aim of this study was to investigate the anti-inflammatory activity of inhaled BCQB in a cigarette smoke (CS)-induced model of acute lung inflammation. Mice exposed to CS developed chronic obstructive pulmonary disease (COPD). Inhalation of BCQB suppressed the accumulation of neutrophils and macrophages in airways and lung and also inhibited the CS-induced increases in mRNA levels of keratinocyte-derived chemokine, monocyte chemotactic protein-1, tumor necrosis factor-alpha, and interleukin-1β in lung and protein expression levels in bronchoalveolar lavage fluid. Moreover, BCQB (300 μg/ml) inhibited the CS-induced changes in superoxide dismutase and myeloperoxidase activities in the lungs. Our study suggests that BCQB might be a potential therapy for inflammation in CS-induced pulmonary diseases, including COPD.
Collapse
Affiliation(s)
- Shui-Juan Zhang
- Department of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Jun-Xia Jiang
- Department of Pharmacology, Zhejiang University School of Medicine, Hangzhou, China, 310058
| | - Qian-Qian Ren
- Department of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Qiang-Min Xie
- Department of Pharmacology, Zhejiang University School of Medicine, Hangzhou, China, 310058
- Department of Medicine, Laboratory Animal Center of Zhejiang University, Hangzhou, China, 310058
| | - Yao-Kang Xiong
- Department of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| |
Collapse
|
27
|
Ganguly K, Martin TM, Concel VJ, Upadhyay S, Bein K, Brant KA, George L, Mitra A, Thimraj TA, Fabisiak JP, Vuga LJ, Fattman C, Kaminski N, Schulz H, Leikauf GD. Secreted phosphoprotein 1 is a determinant of lung function development in mice. Am J Respir Cell Mol Biol 2015; 51:637-51. [PMID: 24816281 DOI: 10.1165/rcmb.2013-0471oc] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Secreted phosphoprotein 1 (Spp1) is located within quantitative trait loci associated with lung function that was previously identified by contrasting C3H/HeJ and JF1/Msf mouse strains that have extremely divergent lung function. JF1/Msf mice with diminished lung function had reduced lung SPP1 transcript and protein during the peak stage of alveologenesis (postnatal day [P]14-P28) as compared with C3H/HeJ mice. In addition to a previously identified genetic variant that altered runt-related transcription factor 2 (RUNX2) binding in the Spp1 promoter, we identified another promoter variant in a putative RUNX2 binding site that increased the DNA protein binding. SPP1 induced dose-dependent mouse lung epithelial-15 cell proliferation. Spp1((-/-)) mice have decreased specific total lung capacity/body weight, higher specific compliance, and increased mean airspace chord length (Lm) compared with Spp1((+/+)) mice. Microarray analysis revealed enriched gene ontogeny categories, with numerous genes associated with lung development and/or respiratory disease. Insulin-like growth factor 1, Hedgehog-interacting protein, wingless-related mouse mammary tumor virus integration site 5A, and NOTCH1 transcripts decreased in the lung of P14 Spp1((-/-)) mice as determined by quantitative RT-PCR analysis. SPP1 promotes pneumocyte growth, and mice lacking SPP1 have smaller, more compliant lungs with enlarged airspace (i.e., increased Lm). Microarray analysis suggests a dysregulation of key lung developmental transcripts in gene-targeted Spp1((-/-)) mice, particularly during the peak phase of alveologenesis. In addition to its known roles in lung disease, this study supports SPP1 as a determinant of lung development in mice.
Collapse
Affiliation(s)
- Koustav Ganguly
- 1 Department of Environmental and Occupational Health, Graduate School of Public Health
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Dani C, Poggi C. The role of genetic polymorphisms in antioxidant enzymes and potential antioxidant therapies in neonatal lung disease. Antioxid Redox Signal 2014; 21:1863-80. [PMID: 24382101 PMCID: PMC4203110 DOI: 10.1089/ars.2013.5811] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
SIGNIFICANCE Oxidative stress is involved in the development of newborn lung diseases, such as bronchopulmonary dysplasia and persistent pulmonary hypertension of the newborn. The activity of antioxidant enzymes (AOEs), which is impaired as a result of prematurity and oxidative injury, may be further affected by specific genetic polymorphisms or an unfavorable combination of more of them. RECENT ADVANCES Genetic polymorphisms of superoxide dismutase and catalase were recently demonstrated to be protective or risk factors for the main complications of prematurity. A lot of research focused on the potential of different antioxidant strategies in the prevention and treatment of lung diseases of the newborn, providing promising results in experimental models. CRITICAL ISSUES The effect of different genetic polymorphisms on protein synthesis and activity has been poorly detailed in the newborn, hindering to derive conclusive results from the observed associations with adverse outcomes. Therapeutic strategies that aimed at enhancing the activity of AOEs were poorly studied in clinical settings and partially failed to produce clinical benefits. FUTURE DIRECTIONS The clarification of the effects of genetic polymorphisms on the proteomics of the newborn is mandatory, as well as the assessment of a larger number of polymorphisms with a possible correlation with adverse outcome. Moreover, antioxidant treatments should be carefully translated to clinical settings, after further details on optimal doses, administration techniques, and adverse effects are provided. Finally, the study of genetic polymorphisms could help select a specific high-risk population, who may particularly benefit from targeted antioxidant strategies.
Collapse
Affiliation(s)
- Carlo Dani
- Section of Neonatology, Department of Neurosciences, Psychology, Drug Research and Child Health, Careggi University Hospital , Florence, Italy
| | | |
Collapse
|
29
|
Zhao R, Liang X, Zhao M, Liu SL, Huang Y, Idell S, Li X, Ji HL. Correlation of apical fluid-regulating channel proteins with lung function in human COPD lungs. PLoS One 2014; 9:e109725. [PMID: 25329998 PMCID: PMC4201481 DOI: 10.1371/journal.pone.0109725] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2014] [Accepted: 09/03/2014] [Indexed: 12/22/2022] Open
Abstract
Links between epithelial ion channels and chronic obstructive pulmonary diseases (COPD) are emerging through animal model and in vitro studies. However, clinical correlations between fluid-regulating channel proteins and lung function in COPD remain to be elucidated. To quantitatively measure epithelial sodium channels (ENaC), cystic fibrosis transmembrane conductance regulator (CFTR), and aquaporin 5 (AQP5) proteins in human COPD lungs and to analyze the correlation with declining lung function, quantitative western blots were used. Spearman tests were performed to identify correlations between channel proteins and lung function. The expression of α and β ENaC subunits was augmented and inversely associated with lung function. In contrast, both total and alveolar type I (ATI) and II (ATII)-specific CFTR proteins were reduced. The expression level of CFTR proteins was associated with FEV1 positively. Abundance of AQP5 proteins and extracellular superoxide dismutase (SOD3) was decreased and correlated with spirometry test results and gas exchange positively. Furthermore, these channel proteins were significantly associated with severity of disease. Our study demonstrates that expression of ENaC, AQP5, and CFTR proteins in human COPD lungs is quantitatively associated with lung function and severity of COPD. These apically located fluid-regulating channels may thereby serve as biomarkers and potent druggable targets of COPD.
Collapse
Affiliation(s)
- Runzhen Zhao
- Department of Cellular and Molecular Biology, University of Texas Health Science Center at Tyler, Tyler, Texas, United States of America
- Texas Lung Injury Institute, University of Texas Health Science Center at Tyler, Tyler, Texas, United States of America
| | - Xinrong Liang
- Department of Cellular and Molecular Biology, University of Texas Health Science Center at Tyler, Tyler, Texas, United States of America
| | - Meimi Zhao
- Department of Cellular and Molecular Biology, University of Texas Health Science Center at Tyler, Tyler, Texas, United States of America
| | - Shan-Lu Liu
- Department of Molecular Microbiology and Immunology, Bond Life Sciences Center, University of Missouri, Columbia, Missouri, United States of America
| | - Yao Huang
- Department of Obstetrics and Gynecology, St. Joseph's Hospital and Medical Center, Phoenix, Arizona, United States of America
| | - Steven Idell
- Department of Cellular and Molecular Biology, University of Texas Health Science Center at Tyler, Tyler, Texas, United States of America
- Medicine, University of Texas Health Science Center at Tyler, Tyler, Texas, United States of America
- Texas Lung Injury Institute, University of Texas Health Science Center at Tyler, Tyler, Texas, United States of America
| | - Xiumin Li
- Xinxiang Medical University, Xinxiang, Henan, China
| | - Hong-Long Ji
- Department of Cellular and Molecular Biology, University of Texas Health Science Center at Tyler, Tyler, Texas, United States of America
- Texas Lung Injury Institute, University of Texas Health Science Center at Tyler, Tyler, Texas, United States of America
- * E-mail:
| |
Collapse
|
30
|
Chen X, Xu X, Xiao F. Heterogeneity of chronic obstructive pulmonary disease: from phenotype to genotype. Front Med 2014; 7:425-32. [PMID: 24234678 DOI: 10.1007/s11684-013-0295-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2013] [Accepted: 08/22/2013] [Indexed: 12/31/2022]
Abstract
Chronic obstructive pulmonary disease (COPD) is one of the leading causes of morbidity and mortality throughout the world and is mainly characterized by persistent airflow limitation. Given that multiple systems other than the lung can be impaired in COPD patients, the traditional FEV1/FVC ratio shows many limitations in COPD diagnosis and assessment. Certain heterogeneities are found in terms of clinical manifestations, physiology, imaging findings, and inflammatory reactions in COPD patients; thus, phenotyping can provide effective information for the prognosis and treatment. However, phenotypes are often based on symptoms or pathophysiological impairments in late-stage COPD, and the role of phenotypes in COPD prevention and early diagnosis remains unclear. This shortcoming may be overcome by the potential genotypes defined by the heterogeneities in certain genes. This review briefly describes the heterogeneity of COPD, with focus on recent advances in the correlations between genotypes and phenotypes. The potential roles of these genotypes and phenotypes in the molecular mechanisms and management of COPD are also elucidated.
Collapse
|
31
|
Hartney JM, Stidham T, Goldstrohm DA, Oberley-Deegan RE, Weaver MR, Valnickova-Hansen Z, Scavenius C, Benninger RKP, Leahy KF, Johnson R, Gally F, Kosmider B, Zimmermann AK, Enghild JJ, Nozik-Grayck E, Bowler RP. A common polymorphism in extracellular superoxide dismutase affects cardiopulmonary disease risk by altering protein distribution. ACTA ACUST UNITED AC 2014; 7:659-66. [PMID: 25085920 DOI: 10.1161/circgenetics.113.000504] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
BACKGROUND The enzyme extracellular superoxide dismutase (EC-SOD; SOD3) is a major antioxidant defense in lung and vasculature. A nonsynonomous single-nucleotide polymorphism in EC-SOD (rs1799895) leads to an arginine to glycine amino acid substitution at position 213 (R213G) in the heparin-binding domain. In recent human genetic association studies, this single-nucleotide polymorphism attenuates the risk of lung disease, yet paradoxically increases the risk of cardiovascular disease. METHODS AND RESULTS Capitalizing on the complete sequence homology between human and mouse in the heparin-binding domain, we created an analogous R213G single-nucleotide polymorphism knockin mouse. The R213G single-nucleotide polymorphism did not change enzyme activity, but shifted the distribution of EC-SOD from lung and vascular tissue to extracellular fluid (eg, bronchoalveolar lavage fluid and plasma). This shift reduces susceptibility to lung disease (lipopolysaccharide-induced lung injury) and increases susceptibility to cardiopulmonary disease (chronic hypoxic pulmonary hypertension). CONCLUSIONS We conclude that EC-SOD provides optimal protection when localized to the compartment subjected to extracellular oxidative stress: thus, the redistribution of EC-SOD from the lung and pulmonary circulation to the extracellular fluids is beneficial in alveolar lung disease but detrimental in pulmonary vascular disease. These findings account for the discrepant risk associated with R213G in humans with lung diseases compared with cardiovascular diseases.
Collapse
Affiliation(s)
- John M Hartney
- From the Department of Medicine, National Jewish Health, Denver, CO (J.M.H., D.A.G., R.E.O.-D., M.R.W., K.F.L., F.G., B.K., R.P.B.); Integrated Department of Immunology, University of Colorado, Denver (J.M.H.); Departments of Pediatrics (T.S., R.K.P.B., R.J., E.N.-G.) and Bioengineering (R.K.P.B.), University of Colorado School of Medicine, Aurora; Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark (Z.V.-H., C.S., J.J.E.); and Institut de Biologie du Developpement de Marseille Luminy, Aix-Marseille University, Marseille, France (A.K.Z.)
| | - Timothy Stidham
- From the Department of Medicine, National Jewish Health, Denver, CO (J.M.H., D.A.G., R.E.O.-D., M.R.W., K.F.L., F.G., B.K., R.P.B.); Integrated Department of Immunology, University of Colorado, Denver (J.M.H.); Departments of Pediatrics (T.S., R.K.P.B., R.J., E.N.-G.) and Bioengineering (R.K.P.B.), University of Colorado School of Medicine, Aurora; Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark (Z.V.-H., C.S., J.J.E.); and Institut de Biologie du Developpement de Marseille Luminy, Aix-Marseille University, Marseille, France (A.K.Z.)
| | - David A Goldstrohm
- From the Department of Medicine, National Jewish Health, Denver, CO (J.M.H., D.A.G., R.E.O.-D., M.R.W., K.F.L., F.G., B.K., R.P.B.); Integrated Department of Immunology, University of Colorado, Denver (J.M.H.); Departments of Pediatrics (T.S., R.K.P.B., R.J., E.N.-G.) and Bioengineering (R.K.P.B.), University of Colorado School of Medicine, Aurora; Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark (Z.V.-H., C.S., J.J.E.); and Institut de Biologie du Developpement de Marseille Luminy, Aix-Marseille University, Marseille, France (A.K.Z.)
| | - Rebecca E Oberley-Deegan
- From the Department of Medicine, National Jewish Health, Denver, CO (J.M.H., D.A.G., R.E.O.-D., M.R.W., K.F.L., F.G., B.K., R.P.B.); Integrated Department of Immunology, University of Colorado, Denver (J.M.H.); Departments of Pediatrics (T.S., R.K.P.B., R.J., E.N.-G.) and Bioengineering (R.K.P.B.), University of Colorado School of Medicine, Aurora; Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark (Z.V.-H., C.S., J.J.E.); and Institut de Biologie du Developpement de Marseille Luminy, Aix-Marseille University, Marseille, France (A.K.Z.)
| | - Michael R Weaver
- From the Department of Medicine, National Jewish Health, Denver, CO (J.M.H., D.A.G., R.E.O.-D., M.R.W., K.F.L., F.G., B.K., R.P.B.); Integrated Department of Immunology, University of Colorado, Denver (J.M.H.); Departments of Pediatrics (T.S., R.K.P.B., R.J., E.N.-G.) and Bioengineering (R.K.P.B.), University of Colorado School of Medicine, Aurora; Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark (Z.V.-H., C.S., J.J.E.); and Institut de Biologie du Developpement de Marseille Luminy, Aix-Marseille University, Marseille, France (A.K.Z.)
| | - Zuzana Valnickova-Hansen
- From the Department of Medicine, National Jewish Health, Denver, CO (J.M.H., D.A.G., R.E.O.-D., M.R.W., K.F.L., F.G., B.K., R.P.B.); Integrated Department of Immunology, University of Colorado, Denver (J.M.H.); Departments of Pediatrics (T.S., R.K.P.B., R.J., E.N.-G.) and Bioengineering (R.K.P.B.), University of Colorado School of Medicine, Aurora; Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark (Z.V.-H., C.S., J.J.E.); and Institut de Biologie du Developpement de Marseille Luminy, Aix-Marseille University, Marseille, France (A.K.Z.)
| | - Carsten Scavenius
- From the Department of Medicine, National Jewish Health, Denver, CO (J.M.H., D.A.G., R.E.O.-D., M.R.W., K.F.L., F.G., B.K., R.P.B.); Integrated Department of Immunology, University of Colorado, Denver (J.M.H.); Departments of Pediatrics (T.S., R.K.P.B., R.J., E.N.-G.) and Bioengineering (R.K.P.B.), University of Colorado School of Medicine, Aurora; Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark (Z.V.-H., C.S., J.J.E.); and Institut de Biologie du Developpement de Marseille Luminy, Aix-Marseille University, Marseille, France (A.K.Z.)
| | - Richard K P Benninger
- From the Department of Medicine, National Jewish Health, Denver, CO (J.M.H., D.A.G., R.E.O.-D., M.R.W., K.F.L., F.G., B.K., R.P.B.); Integrated Department of Immunology, University of Colorado, Denver (J.M.H.); Departments of Pediatrics (T.S., R.K.P.B., R.J., E.N.-G.) and Bioengineering (R.K.P.B.), University of Colorado School of Medicine, Aurora; Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark (Z.V.-H., C.S., J.J.E.); and Institut de Biologie du Developpement de Marseille Luminy, Aix-Marseille University, Marseille, France (A.K.Z.)
| | - Katelyn F Leahy
- From the Department of Medicine, National Jewish Health, Denver, CO (J.M.H., D.A.G., R.E.O.-D., M.R.W., K.F.L., F.G., B.K., R.P.B.); Integrated Department of Immunology, University of Colorado, Denver (J.M.H.); Departments of Pediatrics (T.S., R.K.P.B., R.J., E.N.-G.) and Bioengineering (R.K.P.B.), University of Colorado School of Medicine, Aurora; Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark (Z.V.-H., C.S., J.J.E.); and Institut de Biologie du Developpement de Marseille Luminy, Aix-Marseille University, Marseille, France (A.K.Z.)
| | - Richard Johnson
- From the Department of Medicine, National Jewish Health, Denver, CO (J.M.H., D.A.G., R.E.O.-D., M.R.W., K.F.L., F.G., B.K., R.P.B.); Integrated Department of Immunology, University of Colorado, Denver (J.M.H.); Departments of Pediatrics (T.S., R.K.P.B., R.J., E.N.-G.) and Bioengineering (R.K.P.B.), University of Colorado School of Medicine, Aurora; Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark (Z.V.-H., C.S., J.J.E.); and Institut de Biologie du Developpement de Marseille Luminy, Aix-Marseille University, Marseille, France (A.K.Z.)
| | - Fabienne Gally
- From the Department of Medicine, National Jewish Health, Denver, CO (J.M.H., D.A.G., R.E.O.-D., M.R.W., K.F.L., F.G., B.K., R.P.B.); Integrated Department of Immunology, University of Colorado, Denver (J.M.H.); Departments of Pediatrics (T.S., R.K.P.B., R.J., E.N.-G.) and Bioengineering (R.K.P.B.), University of Colorado School of Medicine, Aurora; Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark (Z.V.-H., C.S., J.J.E.); and Institut de Biologie du Developpement de Marseille Luminy, Aix-Marseille University, Marseille, France (A.K.Z.)
| | - Beata Kosmider
- From the Department of Medicine, National Jewish Health, Denver, CO (J.M.H., D.A.G., R.E.O.-D., M.R.W., K.F.L., F.G., B.K., R.P.B.); Integrated Department of Immunology, University of Colorado, Denver (J.M.H.); Departments of Pediatrics (T.S., R.K.P.B., R.J., E.N.-G.) and Bioengineering (R.K.P.B.), University of Colorado School of Medicine, Aurora; Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark (Z.V.-H., C.S., J.J.E.); and Institut de Biologie du Developpement de Marseille Luminy, Aix-Marseille University, Marseille, France (A.K.Z.)
| | - Angela K Zimmermann
- From the Department of Medicine, National Jewish Health, Denver, CO (J.M.H., D.A.G., R.E.O.-D., M.R.W., K.F.L., F.G., B.K., R.P.B.); Integrated Department of Immunology, University of Colorado, Denver (J.M.H.); Departments of Pediatrics (T.S., R.K.P.B., R.J., E.N.-G.) and Bioengineering (R.K.P.B.), University of Colorado School of Medicine, Aurora; Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark (Z.V.-H., C.S., J.J.E.); and Institut de Biologie du Developpement de Marseille Luminy, Aix-Marseille University, Marseille, France (A.K.Z.)
| | - Jan J Enghild
- From the Department of Medicine, National Jewish Health, Denver, CO (J.M.H., D.A.G., R.E.O.-D., M.R.W., K.F.L., F.G., B.K., R.P.B.); Integrated Department of Immunology, University of Colorado, Denver (J.M.H.); Departments of Pediatrics (T.S., R.K.P.B., R.J., E.N.-G.) and Bioengineering (R.K.P.B.), University of Colorado School of Medicine, Aurora; Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark (Z.V.-H., C.S., J.J.E.); and Institut de Biologie du Developpement de Marseille Luminy, Aix-Marseille University, Marseille, France (A.K.Z.)
| | - Eva Nozik-Grayck
- From the Department of Medicine, National Jewish Health, Denver, CO (J.M.H., D.A.G., R.E.O.-D., M.R.W., K.F.L., F.G., B.K., R.P.B.); Integrated Department of Immunology, University of Colorado, Denver (J.M.H.); Departments of Pediatrics (T.S., R.K.P.B., R.J., E.N.-G.) and Bioengineering (R.K.P.B.), University of Colorado School of Medicine, Aurora; Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark (Z.V.-H., C.S., J.J.E.); and Institut de Biologie du Developpement de Marseille Luminy, Aix-Marseille University, Marseille, France (A.K.Z.)
| | - Russell P Bowler
- From the Department of Medicine, National Jewish Health, Denver, CO (J.M.H., D.A.G., R.E.O.-D., M.R.W., K.F.L., F.G., B.K., R.P.B.); Integrated Department of Immunology, University of Colorado, Denver (J.M.H.); Departments of Pediatrics (T.S., R.K.P.B., R.J., E.N.-G.) and Bioengineering (R.K.P.B.), University of Colorado School of Medicine, Aurora; Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark (Z.V.-H., C.S., J.J.E.); and Institut de Biologie du Developpement de Marseille Luminy, Aix-Marseille University, Marseille, France (A.K.Z.).
| |
Collapse
|
32
|
Constantino L, Gonçalves RC, Giombelli VR, Tomasi CD, Vuolo F, Kist LW, de Oliveira GMT, Pasquali MADB, Bogo MR, Mauad T, Horn A, Melo KV, Fernandes C, Moreira JCF, Ritter C, Dal-Pizzol F. Regulation of lung oxidative damage by endogenous superoxide dismutase in sepsis. Intensive Care Med Exp 2014; 2:17. [PMID: 26266917 PMCID: PMC4513028 DOI: 10.1186/2197-425x-2-17] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2014] [Accepted: 04/07/2014] [Indexed: 11/18/2022] Open
Abstract
Background The purpose of this research is to study the relationship between superoxide dismutase (SOD) and lung redox state in an animal model of sepsis. Methods Sepsis was induced in rats by the cecal ligation and perforation model (CLP). After 3, 6, and 12 h, CLP protein content and expression of SOD1, SOD2, and SOD3 were evaluated, and SOD activity was assessed. Oxidative damage was determined by quantifying nitrotyrosine content. Lung localization of SOD3 was performed by immunohistochemistry. The protective effect of a SOD mimetic on oxidative damage, inflammation, and lung permeability was assessed 12 and 24 h after sepsis induction. Results Lung levels of SOD1 decreased 3 and 12 h after sepsis, but SOD2 and SOD3 increased, as well as SOD activity. These alterations were not associated with changes in sod gene expression. Nitrotyrosine levels increased 3 and 12 h after sepsis. The administration of a SOD mimetic decreased nitrotyrosine and proinflammatory cytokine levels and improved lung permeability. Conclusions SOD2 and SOD3 increased after sepsis induction, but this was insufficient to protect the lung. Treatments based on SOD mimetics could have a role in lung injury associated with sepsis.
Collapse
Affiliation(s)
- Larissa Constantino
- Laboratório de Fisiopatologia Experimenta e Instituto Nacional de Ciência e Tecnologia Translacional em Medicina (INCT-TM), Programa de Pós-Graduação de Ciências da Saúde, Universidade do Extremo Sul Catarinense, Avenida Universitária, 1105, Criciúma, SC, 88806-000, Brazil,
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Okutsu M, Call JA, Lira VA, Zhang M, Donet JA, French BA, Martin KS, Peirce-Cottler SM, Rembold CM, Annex BH, Yan Z. Extracellular superoxide dismutase ameliorates skeletal muscle abnormalities, cachexia, and exercise intolerance in mice with congestive heart failure. Circ Heart Fail 2014; 7:519-30. [PMID: 24523418 DOI: 10.1161/circheartfailure.113.000841] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
BACKGROUND Congestive heart failure (CHF) is a leading cause of morbidity and mortality, and oxidative stress has been implicated in the pathogenesis of cachexia (muscle wasting) and the hallmark symptom, exercise intolerance. We have previously shown that a nitric oxide-dependent antioxidant defense renders oxidative skeletal muscle resistant to catabolic wasting. Here, we aimed to identify and determine the functional role of nitric oxide-inducible antioxidant enzyme(s) in protection against cardiac cachexia and exercise intolerance in CHF. METHODS AND RESULTS We demonstrated that systemic administration of endogenous nitric oxide donor S-nitrosoglutathione in mice blocked the reduction of extracellular superoxide dismutase (EcSOD) protein expression, as well as the induction of MAFbx/Atrogin-1 mRNA expression and muscle atrophy induced by glucocorticoid. We further showed that endogenous EcSOD, expressed primarily by type IId/x and IIa myofibers and enriched at endothelial cells, is induced by exercise training. Muscle-specific overexpression of EcSOD by somatic gene transfer or transgenesis (muscle creatine kinase [MCK]-EcSOD) in mice significantly attenuated muscle atrophy. Importantly, when crossbred into a mouse genetic model of CHF (α-myosin heavy chain-calsequestrin), MCK-EcSOD transgenic mice had significant attenuation of cachexia with preserved whole body muscle strength and endurance capacity in the absence of reduced HF. Enhanced EcSOD expression significantly ameliorated CHF-induced oxidative stress, MAFbx/Atrogin-1 mRNA expression, loss of mitochondria, and vascular rarefaction in skeletal muscle. CONCLUSIONS EcSOD plays an important antioxidant defense function in skeletal muscle against cardiac cachexia and exercise intolerance in CHF.
Collapse
Affiliation(s)
- Mitsuharu Okutsu
- From the Departments of Medicine (M.O., J.A.C., V.A.L., M.Z., J.A.D., C.M.R., B.H.A., Z.Y.), Pharmacology (Z.Y.), and Molecular Physiology and Biological Physics (Z.Y.), Center for Skeletal Muscle Research (M.O., J.A.C., V.A.L., M.Z., J.A.D., Z.Y.), Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA; and Department of Biomedical Engineering, University of Virginia, Charlottesville, VA (B.A.F., K.S.M., S.M.P.-C.)
| | - Jarrod A Call
- From the Departments of Medicine (M.O., J.A.C., V.A.L., M.Z., J.A.D., C.M.R., B.H.A., Z.Y.), Pharmacology (Z.Y.), and Molecular Physiology and Biological Physics (Z.Y.), Center for Skeletal Muscle Research (M.O., J.A.C., V.A.L., M.Z., J.A.D., Z.Y.), Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA; and Department of Biomedical Engineering, University of Virginia, Charlottesville, VA (B.A.F., K.S.M., S.M.P.-C.)
| | - Vitor A Lira
- From the Departments of Medicine (M.O., J.A.C., V.A.L., M.Z., J.A.D., C.M.R., B.H.A., Z.Y.), Pharmacology (Z.Y.), and Molecular Physiology and Biological Physics (Z.Y.), Center for Skeletal Muscle Research (M.O., J.A.C., V.A.L., M.Z., J.A.D., Z.Y.), Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA; and Department of Biomedical Engineering, University of Virginia, Charlottesville, VA (B.A.F., K.S.M., S.M.P.-C.)
| | - Mei Zhang
- From the Departments of Medicine (M.O., J.A.C., V.A.L., M.Z., J.A.D., C.M.R., B.H.A., Z.Y.), Pharmacology (Z.Y.), and Molecular Physiology and Biological Physics (Z.Y.), Center for Skeletal Muscle Research (M.O., J.A.C., V.A.L., M.Z., J.A.D., Z.Y.), Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA; and Department of Biomedical Engineering, University of Virginia, Charlottesville, VA (B.A.F., K.S.M., S.M.P.-C.)
| | - Jean A Donet
- From the Departments of Medicine (M.O., J.A.C., V.A.L., M.Z., J.A.D., C.M.R., B.H.A., Z.Y.), Pharmacology (Z.Y.), and Molecular Physiology and Biological Physics (Z.Y.), Center for Skeletal Muscle Research (M.O., J.A.C., V.A.L., M.Z., J.A.D., Z.Y.), Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA; and Department of Biomedical Engineering, University of Virginia, Charlottesville, VA (B.A.F., K.S.M., S.M.P.-C.)
| | - Brent A French
- From the Departments of Medicine (M.O., J.A.C., V.A.L., M.Z., J.A.D., C.M.R., B.H.A., Z.Y.), Pharmacology (Z.Y.), and Molecular Physiology and Biological Physics (Z.Y.), Center for Skeletal Muscle Research (M.O., J.A.C., V.A.L., M.Z., J.A.D., Z.Y.), Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA; and Department of Biomedical Engineering, University of Virginia, Charlottesville, VA (B.A.F., K.S.M., S.M.P.-C.)
| | - Kyle S Martin
- From the Departments of Medicine (M.O., J.A.C., V.A.L., M.Z., J.A.D., C.M.R., B.H.A., Z.Y.), Pharmacology (Z.Y.), and Molecular Physiology and Biological Physics (Z.Y.), Center for Skeletal Muscle Research (M.O., J.A.C., V.A.L., M.Z., J.A.D., Z.Y.), Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA; and Department of Biomedical Engineering, University of Virginia, Charlottesville, VA (B.A.F., K.S.M., S.M.P.-C.)
| | - Shayn M Peirce-Cottler
- From the Departments of Medicine (M.O., J.A.C., V.A.L., M.Z., J.A.D., C.M.R., B.H.A., Z.Y.), Pharmacology (Z.Y.), and Molecular Physiology and Biological Physics (Z.Y.), Center for Skeletal Muscle Research (M.O., J.A.C., V.A.L., M.Z., J.A.D., Z.Y.), Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA; and Department of Biomedical Engineering, University of Virginia, Charlottesville, VA (B.A.F., K.S.M., S.M.P.-C.)
| | - Christopher M Rembold
- From the Departments of Medicine (M.O., J.A.C., V.A.L., M.Z., J.A.D., C.M.R., B.H.A., Z.Y.), Pharmacology (Z.Y.), and Molecular Physiology and Biological Physics (Z.Y.), Center for Skeletal Muscle Research (M.O., J.A.C., V.A.L., M.Z., J.A.D., Z.Y.), Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA; and Department of Biomedical Engineering, University of Virginia, Charlottesville, VA (B.A.F., K.S.M., S.M.P.-C.)
| | - Brian H Annex
- From the Departments of Medicine (M.O., J.A.C., V.A.L., M.Z., J.A.D., C.M.R., B.H.A., Z.Y.), Pharmacology (Z.Y.), and Molecular Physiology and Biological Physics (Z.Y.), Center for Skeletal Muscle Research (M.O., J.A.C., V.A.L., M.Z., J.A.D., Z.Y.), Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA; and Department of Biomedical Engineering, University of Virginia, Charlottesville, VA (B.A.F., K.S.M., S.M.P.-C.)
| | - Zhen Yan
- From the Departments of Medicine (M.O., J.A.C., V.A.L., M.Z., J.A.D., C.M.R., B.H.A., Z.Y.), Pharmacology (Z.Y.), and Molecular Physiology and Biological Physics (Z.Y.), Center for Skeletal Muscle Research (M.O., J.A.C., V.A.L., M.Z., J.A.D., Z.Y.), Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA; and Department of Biomedical Engineering, University of Virginia, Charlottesville, VA (B.A.F., K.S.M., S.M.P.-C.).
| |
Collapse
|
34
|
Vlahos R, Bozinovski S. Recent advances in pre-clinical mouse models of COPD. Clin Sci (Lond) 2014; 126:253-65. [PMID: 24144354 PMCID: PMC3878607 DOI: 10.1042/cs20130182] [Citation(s) in RCA: 122] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2013] [Revised: 07/17/2013] [Accepted: 08/01/2013] [Indexed: 02/06/2023]
Abstract
COPD (chronic obstructive pulmonary disease) is a major incurable global health burden and will become the third largest cause of death in the world by 2020. It is currently believed that an exaggerated inflammatory response to inhaled irritants, in particular cigarette smoke, causes progressive airflow limitation. This inflammation, where macrophages, neutrophils and T-cells are prominent, leads to oxidative stress, emphysema, small airways fibrosis and mucus hypersecretion. The mechanisms and mediators that drive the induction and progression of chronic inflammation, emphysema and altered lung function are poorly understood. Current treatments have limited efficacy in inhibiting chronic inflammation, do not reverse the pathology of disease and fail to modify the factors that initiate and drive the long-term progression of disease. Therefore there is a clear need for new therapies that can prevent the induction and progression of COPD. Animal modelling systems that accurately reflect disease pathophysiology continue to be essential to the development of new therapies. The present review highlights some of the mouse models used to define the cellular, molecular and pathological consequences of cigarette smoke exposure and whether they can be used to predict the efficacy of new therapeutics for COPD.
Collapse
Key Words
- acute exacerbations of chronic obstructive pulmonary disease (aecopd)
- chronic obstructive pulmonary disease (copd)
- emphysema
- inflammation
- skeletal muscle wasting
- smoking
- aecopd, acute exacerbations of copd
- bal, bronchoalveolar lavage
- balf, bal fluid
- copd, chronic obstructive pulmonary disease
- gm-csf, granulocyte/macrophage colony-stimulating factor
- gold, global initiative on chronic obstructive lung disease
- gpx, glutathione peroxidase
- hdac, histone deacetylation
- il, interleukin
- ltb4, leukotriene b4
- mapk, mitogen-activated protein kinase
- mcp-1, monocyte chemotactic protein-1
- mmp, matrix metalloproteinase
- ne, neutrophil elastase
- nf-κb, nuclear factor κb
- nrf2, nuclear erythroid-related factor 2
- o2•−, superoxide radical
- onoo−, peroxynitrite
- pde, phosphodiesterase
- pi3k, phosphoinositide 3-kinase
- ros, reactive oxygen species
- rv, rhinovirus
- slpi, secretory leucocyte protease inhibitor
- sod, superoxide dismutase
- tgf-β, transforming growth factor-β
- timp, tissue inhibitor of metalloproteinases
- tnf-α, tumour necrosis factor-α
- v/q, ventilation/perfusion
Collapse
Affiliation(s)
- Ross Vlahos
- *Lung Health Research Centre, Department of Pharmacology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Steven Bozinovski
- *Lung Health Research Centre, Department of Pharmacology, University of Melbourne, Parkville, VIC 3010, Australia
| |
Collapse
|
35
|
Usher AKH, Stockley RA. The link between chronic periodontitis and COPD: a common role for the neutrophil? BMC Med 2013; 11:241. [PMID: 24229090 PMCID: PMC4225606 DOI: 10.1186/1741-7015-11-241] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2013] [Accepted: 11/01/2013] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND The possible relationship between chronic inflammatory diseases and their co-morbidities has become an increasing focus of research. Both chronic periodontitis and chronic obstructive pulmonary disease are neutrophilic, inflammatory conditions characterized by the loss of local connective tissue. Evidence suggests an association and perhaps a causal link between the two diseases. However, the nature of any relationship between them is unclear, but if pathophysiologically established may have wide-reaching implications for targeted treatments to improve outcomes and prognosis. DISCUSSION There have been a number of epidemiological studies undertaken demonstrating an independent association between chronic periodontitis and chronic obstructive pulmonary disease. However, many of them have significant limitations, and drawing firm conclusions regarding causality may be premature. Although the pathology of both these diseases is complex and involves many cell types, such as CD8 positive cells and macrophages, both conditions are predominantly characterized by neutrophilic inflammation. Increasingly, there is evidence that the two conditions are underpinned by similar pathophysiological processes, especially centered on the functions of the neutrophil. These include a disturbance in protease/anti-protease and redox state balance. The association demonstrated by epidemiological studies, as well as emerging similarities in pathogenesis at the level of the neutrophil, suggest a basis for testing the effects of treatment for one condition upon the severity of the other. SUMMARY Although the evidence of an independent association between chronic periodontitis and chronic obstructive pulmonary disease grows stronger, there remains a lack of definitive studies designed to establish causality and treatment effects. There is a need for future research to be focused on answering these questions.
Collapse
Affiliation(s)
- Adam K H Usher
- Lung Investigation Unit, Queen Elizabeth Hospital, Mindelsohn Way, Edgbaston, Birmingham B15 2WB, UK.
| | | |
Collapse
|
36
|
Tang W, Bentley AR, Kritchevsky SB, Harris TB, Newman AB, Bauer DC, Meibohm B, Cassano PA. Genetic variation in antioxidant enzymes, cigarette smoking, and longitudinal change in lung function. Free Radic Biol Med 2013; 63:304-12. [PMID: 23688726 PMCID: PMC4060265 DOI: 10.1016/j.freeradbiomed.2013.05.016] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2012] [Revised: 04/08/2013] [Accepted: 05/10/2013] [Indexed: 12/25/2022]
Abstract
Antioxidant enzymes play an important role in the defense against oxidative stress in the lung and in the pathogenesis of chronic obstructive pulmonary disease (COPD). Sequence variation in genes encoding antioxidant enzymes may alter susceptibility to COPD by affecting longitudinal change in lung function in adults. We genotyped 384 sequence variants in 56 candidate genes in 1281 African American and 1794 European American elderly adults in the Health, Aging, and Body Composition study. Single-marker associations and gene-by-smoking interactions with rate of change in FEV₁ and FEV₁/FVC were evaluated using linear mixed-effects models, stratified by race/ethnicity. In European Americans, rs17883901 in GCLC was statistically significantly associated with rate of change in FEV₁/FVC; the recessive genotype (TT) was associated with a 0.9% per year steeper decline (P = 4.50 × 10(-5)). Statistically significant gene-by-smoking interactions were observed for variants in two genes in European Americans: the minor allele of rs2297765 in mGST3 attenuated the accelerated decline in FEV₁/FVC in smokers by 0.45% per year (P = 1.13 × 10(-4)); for participants with greater baseline smoking pack-years, the minor allele of rs2073192 in IDH3B was associated with an accelerated decline in FEV₁/FVC (P = 2.10 × 10(-4)). For both genes, nominally significant interactions (P < 0.01) were observed at the gene level in African Americans (P = 0.007 and 4.60 × 10(-4), respectively). Nominally significant evidence of association was observed for variants in SOD3 and GLRX2 in multiple analyses. This study identifies two novel genes associated with longitudinal lung function phenotypes in both African and European Americans and confirms a prior finding for GCLC. These findings suggest novel mechanisms and molecular targets for future research and advance the understanding of genetic determinants of lung function and COPD risk.
Collapse
Affiliation(s)
- Wenbo Tang
- Division of Nutritional Sciences, Cornell University, Ithaca, NY 14853, USA
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Afzal S, Lange P, Bojesen SE, Freiberg JJ, Nordestgaard BG. Plasma 25-hydroxyvitamin D, lung function and risk of chronic obstructive pulmonary disease. Thorax 2013; 69:24-31. [DOI: 10.1136/thoraxjnl-2013-203682] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
|
38
|
|
39
|
Abstract
A genetic contribution to develop chronic obstructive pulmonary disease (COPD) is well established. However, the specific genes responsible for enhanced risk or host differences in susceptibility to smoke exposure remain poorly understood. The goal of this review is to provide a comprehensive literature overview on the genetics of COPD, highlight the most promising findings during the last few years, and ultimately provide an updated COPD gene list. Candidate gene studies on COPD and related phenotypes indexed in PubMed before January 5, 2012 are tabulated. An exhaustive list of publications for any given gene was looked for. This well-documented COPD candidate-gene list is expected to serve many purposes for future replication studies and meta-analyses as well as for reanalyzing collected genomic data in the field. In addition, this review summarizes recent genetic loci identified by genome-wide association studies on COPD, lung function, and related complications. Assembling resources, integrative genomic approaches, and large sample sizes of well-phenotyped subjects is part of the path forward to elucidate the genetic basis of this debilitating disease.
Collapse
Affiliation(s)
- Yohan Bossé
- Centre de recherche Institut universitaire de cardiologie et de pneumologie de Québec, Quebec, Canada.
| |
Collapse
|
40
|
Giusti B, Vestrini A, Poggi C, Magi A, Pasquini E, Abbate R, Dani C. Genetic polymorphisms of antioxidant enzymes as risk factors for oxidative stress-associated complications in preterm infants. Free Radic Res 2012; 46:1130-9. [PMID: 22574884 DOI: 10.3109/10715762.2012.692787] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
We aimed to identify specific polymorphisms of genes encoding for superoxide dismutase (SOD) 1 (cytoplasmic Cu/ZnSOD), SOD2 (mitochondrial MnSOD), SOD3 (extracellular Cu/ZnSOD) and CAT in a cohort of preterm infants and correlate their presence to the development of respiratory distress syndrome (RDS), bronchopulmonary dysplasia (BPD), intraventricular haemorrhage (IVH) and retinopathy of prematurity (ROP). We carried out a retrospective study to evaluate the allele frequency and the genotype distribution of polymorphisms of SODs and CAT in a population of preterm neonates (n = 152) with a gestational age ≤ 28 weeks according to the presence or absence of RDS, BPD, IVH and ROP. Moreover, we evaluated through the haplotype reconstruction analysis whether combinations of the selected polymorphisms are related to the occurrence of RDS, BPD, IVH and ROP. We found that rs8192287 SOD3 polymorphism is an independent protective factor for all grade IVH, while rs4880 and rs5746136 SOD2 polymorphisms are associated with a lower gestational age (rs4880, rs5746136) and birth weight (rs4880). Haplotypes reconstruction showed that SOD1 (GG) decreased the risk of RDS, IVH and ROP; SOD2 (GT) increased the risk of BPD and decreased the risk of RDS, IVH and ROP; SOD3 (TGC) decreased the risk of BPD and IVH; and 4) CAT (CTC) decreased the risk of RDS. The rs8192287 SOD3 polymorphism is per se an independent predictor of a decreased risk of developing IVH. Different SOD2 polymorphisms are associated per se with a lower gestational age and/or birth weight, and haplotypes of SOD1, SOD3 and CAT genes may be independent protecting or risk markers for prematurity complications.
Collapse
Affiliation(s)
- Betti Giusti
- Department of Medical and Surgical Critical Care, University of Florence, Atherothrombotic Diseases Centre, Careggi University Hospital of Florence, Florence, Italy
| | | | | | | | | | | | | |
Collapse
|
41
|
Crawford A, Fassett RG, Geraghty DP, Kunde DA, Ball MJ, Robertson IK, Coombes JS. Relationships between single nucleotide polymorphisms of antioxidant enzymes and disease. Gene 2012; 501:89-103. [PMID: 22525041 DOI: 10.1016/j.gene.2012.04.011] [Citation(s) in RCA: 124] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2012] [Revised: 02/21/2012] [Accepted: 04/05/2012] [Indexed: 12/18/2022]
Abstract
The presence and progression of numerous diseases have been linked to deficiencies in antioxidant systems. The relationships between single nucleotide polymorphisms (SNPs) arising from specific antioxidant enzymes and diseases associated with elevated oxidative stress have been studied with the rationale that they may be useful in screening for diseases. The purpose of this narrative review is to analyse evidence from these studies. The antioxidant enzyme SNPs selected for analysis are based on those most frequently investigated in relation to diseases in humans: superoxide dismutase (SOD2) Ala16Val (80 studies), glutathione peroxidise (GPx1) Pro197Leu (24 studies) and catalase C-262T (22 studies). Although the majority of evidence supports associations between the SOD2 Ala16Val SNP and diseases such as breast, prostate and lung cancers, diabetes and cardiovascular disease, the presence of the SOD2 Ala16Val SNP confers only a small, clinically insignificant reduction (if any) in the risk of these diseases. Other diseases such as bladder cancer, liver disease, nervous system pathologies and asthma have not been consistently related to this SOD SNP genotype. The GPx1 Pro197Leu and catalase C-262T SNP genotypes have been associated with breast cancer, but only in a small number of studies. Thus, currently available evidence suggests antioxidant enzyme SNP genotypes are not useful for screening for diseases in humans.
Collapse
Affiliation(s)
- Amanda Crawford
- School of Human Life Sciences, University of Tasmania, Newnham, Launceston, Tasmania 7248, Australia
| | | | | | | | | | | | | |
Collapse
|
42
|
Rahman I. Pharmacological antioxidant strategies as therapeutic interventions for COPD. Biochim Biophys Acta Mol Basis Dis 2011; 1822:714-28. [PMID: 22101076 DOI: 10.1016/j.bbadis.2011.11.004] [Citation(s) in RCA: 74] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2011] [Revised: 11/01/2011] [Accepted: 11/02/2011] [Indexed: 10/15/2022]
Abstract
Cigarette/tobacco smoke/biomass fuel-induced oxidative and aldehyde/carbonyl stress are intimately associated with the progression and exacerbation of chronic obstructive pulmonary disease (COPD). Therefore, targeting systemic and local oxidative stress with antioxidants/redox modulating agents, or boosting the endogenous levels of antioxidants are likely to have beneficial effects in the treatment/management of COPD. Various antioxidant agents, such as thiol molecules (glutathione and mucolytic drugs, such as N-acetyl-L-cysteine and N-acystelyn, erdosteine, fudosteine, ergothioneine, and carbocysteine), have been reported to modulate various cellular and biochemical aspects of COPD. These antioxidants have been found to scavenge and detoxify free radicals and oxidants, regulate of glutathione biosynthesis, control nuclear factor-kappaB (NF-kappaB) activation, and hence inhibiting inflammatory gene expression. Synthetic molecules, such as specific spin traps like α-phenyl-N-tert-butyl nitrone, a catalytic antioxidant (ECSOD mimetic), porphyrins (AEOL 10150 and AEOL 10113), and a superoxide dismutase mimetic M40419, iNOS and myeloperoxidase inhibitors, lipid peroxidation inhibitors/blockers edaravone, and lazaroids/tirilazad have also been shown to have beneficial effects by inhibiting cigarette smoke-induced inflammatory responses and other carbonyl/oxidative stress-induced cellular alterations. A variety of oxidants, free radicals, and carbonyls/aldehydes are implicated in the pathogenesis of COPD, it is therefore, possible that therapeutic administration or supplementation of multiple antioxidants and/or boosting the endogenous levels of antioxidants will be beneficial in the treatment of COPD. This review discusses various novel pharmacological approaches adopted to enhance lung antioxidant levels, and various emerging beneficial and/or prophylactic effects of antioxidant therapeutics in halting or intervening the progression of COPD. This article is part of a Special Issue entitled: Antioxidants and Antioxidant Treatment in Disease.
Collapse
Affiliation(s)
- Irfan Rahman
- Department of Environmental Medicine, Lung Biology and Disease Program, University of Rochester Medical Center, NY 14642, USA.
| |
Collapse
|
43
|
Teoh-Fitzgerald MLT, Fitzgerald MP, Jensen TJ, Futscher BW, Domann FE. Genetic and epigenetic inactivation of extracellular superoxide dismutase promotes an invasive phenotype in human lung cancer by disrupting ECM homeostasis. Mol Cancer Res 2011; 10:40-51. [PMID: 22064654 DOI: 10.1158/1541-7786.mcr-11-0501] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Extracellular superoxide dismutase (EcSOD) is an important superoxide scavenger in the lung in which its loss, sequence variation, or abnormal expression contributes to lung diseases; however, the role of EcSOD in lung cancer has yet to be studied. We hypothesized that EcSOD loss could affect malignant progression in lung, and could be either genetic or epigenetic in nature. To test this, we analyzed EcSOD expression, gene copy number, promoter methylation, and chromatin accessibility in normal lung and carcinoma cells. We found that normal airway epithelial cells expressed abundant EcSOD and had an unmethylated promoter, whereas EcSOD-negative lung cancer cells displayed aberrant promoter hypermethylation and decreased chromatin accessibility. 5-aza-dC induced EcSOD suggesting that cytosine methylation was causal, in part, to silencing. In 48/50 lung tumors, EcSOD mRNA was significantly lower as early as stage I, and the EcSOD promoter was hypermethylated in 8/10 (80%) adenocarcinomas compared with 0/5 normal lung samples. In addition, 20% of the tumors showed loss of heterozygosity (LOH) of EcSOD. Reexpression of EcSOD attenuated the malignant phenotype of lung carcinoma cells by significantly decreasing invasion and survival. Finally, EcSOD decreased heparanase and syndecan-1 mRNAs in part by reducing NF-κB. By contrast, MnSOD and CuZnSOD showed no significant changes in lung tumors and had no effect on heparanase expression. Taken together, the loss of EcSOD expression is unique among the superoxide dismutases in lung cancer and is the result of EcSOD promoter methylation and LOH, suggesting that its early loss may contribute to ECM remodeling and malignant progression.
Collapse
Affiliation(s)
- Melissa L T Teoh-Fitzgerald
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, Carver College of Medicine and The Holden Comprehensive Cancer Center, The University of Iowa, Iowa City, Iowa 52242, USA
| | | | | | | | | |
Collapse
|
44
|
Fischer BM, Pavlisko E, Voynow JA. Pathogenic triad in COPD: oxidative stress, protease-antiprotease imbalance, and inflammation. Int J Chron Obstruct Pulmon Dis 2011; 6:413-21. [PMID: 21857781 PMCID: PMC3157944 DOI: 10.2147/copd.s10770] [Citation(s) in RCA: 196] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2011] [Indexed: 01/07/2023] Open
Abstract
Patients with chronic obstructive pulmonary disease (COPD) exhibit dominant features of chronic bronchitis, emphysema, and/or asthma, with a common phenotype of airflow obstruction. COPD pulmonary physiology reflects the sum of pathological changes in COPD, which can occur in large central airways, small peripheral airways, and the lung parenchyma. Quantitative or high-resolution computed tomography is used as a surrogate measure for assessment of disease progression. Different biological or molecular markers have been reported that reflect the mechanistic or pathogenic triad of inflammation, proteases, and oxidants and correspond to the different aspects of COPD histopathology. Similar to the pathogenic triad markers, genetic variations or polymorphisms have also been linked to COPD-associated inflammation, protease–antiprotease imbalance, and oxidative stress. Furthermore, in recent years, there have been reports identifying aging-associated mechanistic markers as downstream consequences of the pathogenic triad in the lungs from COPD patients. For this review, the authors have limited their discussion to a review of mechanistic markers and genetic variations and their association with COPD histopathology and disease status.
Collapse
Affiliation(s)
- Bernard M Fischer
- Department of Pediatrics, Duke University Medical Center, Durham, NC 27710, USA.
| | | | | |
Collapse
|
45
|
Regan EA, Mazur W, Meoni E, Toljamo T, Millar J, Vuopala K, Bowler RP, Rahman I, Nicks ME, Crapo JD, Kinnula VL. Smoking and COPD increase sputum levels of extracellular superoxide dismutase. Free Radic Biol Med 2011; 51:726-32. [PMID: 21621610 DOI: 10.1016/j.freeradbiomed.2011.05.008] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2011] [Revised: 04/28/2011] [Accepted: 05/09/2011] [Indexed: 02/02/2023]
Abstract
Extracellular superoxide dismutase (ECSOD) is the major superoxide-scavenging enzyme in the lung. Certain ECSOD polymorphisms are protective against COPD. We postulated that smokers and COPD subjects would have altered levels of ECSOD in the lung, airway secretions, and/or plasma. Lung tissue ECSOD was evaluated from nonsmokers, smokers, and subjects with mild to very severe COPD by Western blot, immunohistochemistry, and ELISA. ECSOD levels in plasma, bronchoalveolar lavage fluid (BALF), and induced-sputum supernatants were analyzed by ELISA and correlated with smoking history and disease status. Immunohistochemistry identified ECSOD in extracellular matrix around bronchioles, arteries, and alveolar walls, with decreases seen in the interstitium and vessels of severe COPD subjects using digital image analysis. Plasma ECSOD did not differ between COPD subjects and controls nor based on smoking status. ECSOD levels in induced sputum supernatants were elevated in current smokers and especially in COPD subjects compared to nonsmokers, whereas corresponding changes could not be seen in the BALF. ECSOD expression was reduced around vessels and bronchioles in COPD lungs. Substantial increases in sputum ECSOD in smokers and COPD is interpreted as an adaptive response to increased oxidative stress and may be a useful biomarker of disease activity in COPD.
Collapse
Affiliation(s)
- Elizabeth A Regan
- Department of Medicine, National Jewish Health, Denver, CO 80206, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Lakhdar R, Denden S, Kassab A, Leban N, Knani J, Lefranc G, Miled A, Chibani JB, Khelil AH. Update in chronic obstructive pulmonary disease: role of antioxidant and metabolizing gene polymorphisms. Exp Lung Res 2011; 37:364-75. [PMID: 21721950 DOI: 10.3109/01902148.2011.580416] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Chronic obstructive pulmonary disease (COPD) is characterized by systemic and local chronic inflammation and oxidative stress. The sources of the increased oxidative stress in COPD patients derive from the increased burden of inhaled oxidants such as cigarette smoke and other forms of particulate or gaseous air pollution and from the increase in reactive oxygen species (ROS) generated by several inflammatory, immune, and structural airways cells. There is increasing evidence that genetic factors may also contribute to the pathogenesis if COPD, particularly antioxidant genes, which may confer a susceptibility to environmental insults such as cigarette smoke and thereafter development of COPD. Consequently, heme oxygenase-1 (HO-1), superoxide dismutase (SOD), catalase (CAT), glutathione S-transferase (GST), microsomal epoxide hydrolase (EPHX1), and cytochrome P450 (CYP) genetic polymorphisms may have an important role in COPD pathogenesis. In this review the authors summarized the most recent findings dealing with these antioxidant genes contributing to the free radical neutralization and xenobiotic enzymes playing a role in different phases of cell detoxification reactions related to the redox status imbalance in COPD, with an emphasis on their possible roles in disease progression.
Collapse
Affiliation(s)
- Ramzi Lakhdar
- Biochemistry and Molecular Biology Laboratory, Faculty of Pharmacy, Monastir, Tunisia.
| | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Lee J, Dahl M, Nordestgaard BG. Genetically lowered microsomal epoxide hydrolase activity and tobacco-related cancer in 47,000 individuals. Cancer Epidemiol Biomarkers Prev 2011; 20:1673-82. [PMID: 21653646 DOI: 10.1158/1055-9965.epi-10-1165] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Two functional polymorphisms of the microsomal epoxide hydrolase (mEH) gene (EPHX1), Tyr113His (rs1051740) and His139Arg (rs2234922), have variably been found to influence susceptibility to various cancer forms. We tested whether genetically lowered mEH activity affects risk of developing cancer in the general population. METHODS We genotyped 47,089 individuals from the Danish general population for the Tyr113His and His139Arg polymorphisms in the EPHX1 gene and divided them into groups with predicted fast, intermediate, and slow mEH activity. Using Cox proportional hazards models, we calculated HRs for 26 individual cancer diagnoses and for groups of any cancer, tobacco-related cancers, estrogen-related female cancers, and other cancers. RESULTS Of the 47,089 individuals, 7,590 experienced a cancer event, and of these, 1,466 were tobacco-related. After multifactorial adjustment, the HRs (95% CI) for tobacco-related cancer were 1.1 (0.8-1.5) and 1.5 (1.1-2.0) in individuals with intermediate and slow mEH activity versus individuals with the fast phenotype (P(trend) = 0.003). The corresponding HRs among ever-smokers were 1.1 (0.8-1.5) and 1.5 (1.1-2.0; P(trend) = 0.003), whereas HRs among never-smokers did not differ from 1.0. CONCLUSIONS Our results indicate that genetically lowered mEH activity is associated with increased risk of developing tobacco-related cancer among smokers in the general population; however, additional studies are needed to confirm our findings. IMPACT To our knowledge, this is the largest study to investigate the association of mEH phenotype and genotype with tobacco-related cancers combined in the general population.
Collapse
Affiliation(s)
- Julie Lee
- Department of Clinical Biochemistry, Herlev Hospital, Herlev, Denmark
| | | | | |
Collapse
|
48
|
Thomsen M, Nordestgaard BG, Tybjaerg-Hansen A, Dahl M. Scavenger receptor AI/II truncation, lung function and COPD: a large population-based study. J Intern Med 2011; 269:340-8. [PMID: 21077973 DOI: 10.1111/j.1365-2796.2010.02308.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
OBJECTIVES The scavenger receptor A-I/II (SRA-I/II) on alveolar macrophages is involved in recognition and clearance of modified lipids and inhaled particulates. A rare variant of the SRA-I/II gene, Arg293X, truncates the distal collagen-like domain, which is essential for ligand recognition. We tested whether the Arg293X variant is associated with reduced lung function and risk of chronic obstructive pulmonary disease (COPD) in the general population. METHODS We genotyped 48,741 individuals from the adult Danish general population for Arg293X, and recorded lung function and spirometry-defined COPD. RESULTS Arg293X homozygotes (n = 5) and heterozygotes (n = 587), compared with noncarriers (n = 48,149), had a 6% and 1% reduction in predicted percentage of forced vital capacity (FVC % predicted) (P = 0.05) and a nonsignificant 7% and 1% reduction in predicted percentage of forced expiratory volume in one second (FEV(1) % predicted) (P = 0.06), respectively. The Arg293X genotype interacted with gender (P = 0.004) and α(1) -antitrypsin MZ heterozygosity (P = 0.049), but not with superoxide dismutase-3 E1I1 heterozygosity (P = 0.11) in determining FEV(1) % predicted. Amongst men, FEV(1) % predicted and FVC % predicted were both reduced by 4% (P = 0.0004 and P = 0.0003, respectively) in Arg293X heterozygotes compared with noncarriers. Corresponding values were 14% (P = 0.03) and 11% (P = 0.04) amongst MZ heterozygotes, and 9% (P = 0.03) and 8% (P = 0.04) amongst E1I1 heterozygotes, compared with noncarriers. Lung function did not differ between Arg293X heterozygotes and noncarriers amongst females or individuals without MZ and E1I1. Arg293X heterozygosity was associated with spirometry-defined COPD amongst men [odds ratio (95% confidence interval): 1.7 (1.1-2.4)], but not with COPD in the whole cohort or in any other subgroup. CONCLUSIONS SRAI/II Arg293X heterozygotes have reduced lung function and increased COPD risk amongst men. They also have reduced lung function amongst individuals heterozygous for the α(1)-antitrypsin MZ and superoxide dismutase-3 E1I1 genotypes.
Collapse
Affiliation(s)
- M Thomsen
- Department of Clinical Biochemistry, Herlev Hospital, Copenhagen University Hospital, Copenhagen, Denmark
| | | | | | | |
Collapse
|
49
|
Yao H, Rahman I. Current concepts on oxidative/carbonyl stress, inflammation and epigenetics in pathogenesis of chronic obstructive pulmonary disease. Toxicol Appl Pharmacol 2011; 254:72-85. [PMID: 21296096 DOI: 10.1016/j.taap.2009.10.022] [Citation(s) in RCA: 172] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2008] [Revised: 10/04/2009] [Accepted: 10/04/2009] [Indexed: 12/17/2022]
Abstract
Chronic obstructive pulmonary disease (COPD) is a global health problem. The current therapies for COPD are poorly effective and the mainstays of pharmacotherapy are bronchodilators. A better understanding of the pathobiology of COPD is critical for the development of novel therapies. In the present review, we have discussed the roles of oxidative/aldehyde stress, inflammation/immunity, and chromatin remodeling in the pathogenesis of COPD. An imbalance of oxidants/antioxidants caused by cigarette smoke and other pollutants/biomass fuels plays an important role in the pathogenesis of COPD by regulating redox-sensitive transcription factors (e.g., NF-κB), autophagy and unfolded protein response leading to chronic lung inflammatory response. Cigarette smoke also activates canonical/alternative NF-κB pathways and their upstream kinases leading to sustained inflammatory response in lungs. Recently, epigenetic regulation has been shown to be critical for the development of COPD because the expression/activity of enzymes that regulate these epigenetic modifications have been reported to be abnormal in airways of COPD patients. Hence, the significant advances made in understanding the pathophysiology of COPD as described herein will identify novel therapeutic targets for intervention in COPD.
Collapse
Affiliation(s)
- Hongwei Yao
- Department of Environmental Medicine, Lung Biology and Disease Program, University of Rochester Medical Center, Rochester, NY 1464, USA
| | | |
Collapse
|
50
|
Tollefson AK, Oberley-Deegan RE, Butterfield KT, Nicks ME, Weaver MR, Remigio LK, Decsesznak J, Chu H, Bratton DL, Riches DW, Bowler RP. Endogenous enzymes (NOX and ECSOD) regulate smoke-induced oxidative stress. Free Radic Biol Med 2010; 49:1937-46. [PMID: 20887783 PMCID: PMC3780970 DOI: 10.1016/j.freeradbiomed.2010.09.022] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2010] [Revised: 09/15/2010] [Accepted: 09/22/2010] [Indexed: 01/25/2023]
Abstract
Chronic obstructive pulmonary disease (COPD) is the fourth leading cause of death in the United States and the incidence is increasing as the population ages. Cigarette smoking is the primary risk factor; however, only a minority of smokers develop the disease. Inhalation of cigarette smoke introduces an abundance of free radicals into the lungs, causing oxidative stress and inflammation. We hypothesized that after the initial burst of oxidative stress associated with cigarette smoke exposure, a sustained source of endogenous free radical production is modulated by the antioxidant enzyme extracellular superoxide dismutase (ECSOD) and the superoxide-generating complex NADPH oxidase (NOX). Primary mouse macrophages exposed to cigarette smoke extract exhibited increased oxidative stress as indicated by fluorogenic dyes and isoprostane concentration, which was suppressed in the presence of both a superoxide dismutase mimetic and a NOX inhibitor. Similarly, primary macrophages isolated from ECSOD-overexpressing mice or NOX-deficient mice showed reduced oxidative stress in response to cigarette smoke treatment. In addition, both reduced glutathione and cytokines (MIP2 and IFNγ) were increased in bronchoalveolar lavage fluid of wild-type mice exposed to cigarette smoke but not in ECSOD-overexpressing or NOX-deficient mice. These data suggest that the mechanisms underlying the host defense against cigarette smoke-induced oxidative damage and subsequent development of COPD may include endogenous oxidases and antioxidant enzymes.
Collapse
Affiliation(s)
| | | | | | - Michael E. Nicks
- Department of Medicine, National Jewish Health, Denver, CO 80206, USA
| | - Michael R. Weaver
- Department of Medicine, National Jewish Health, Denver, CO 80206, USA
| | - Linda K. Remigio
- Department of Medicine, National Jewish Health, Denver, CO 80206, USA
| | | | - H.W. Chu
- Department of Medicine, National Jewish Health, Denver, CO 80206, USA
- University of Colorado at Denver and Health Sciences Center, Denver, CO 80262, USA
| | - Donna L. Bratton
- University of Colorado at Denver and Health Sciences Center, Denver, CO 80262, USA
| | - David W. Riches
- Department of Medicine, National Jewish Health, Denver, CO 80206, USA
- University of Colorado at Denver and Health Sciences Center, Denver, CO 80262, USA
| | - Russell P. Bowler
- Department of Medicine, National Jewish Health, Denver, CO 80206, USA
- University of Colorado at Denver and Health Sciences Center, Denver, CO 80262, USA
| |
Collapse
|