51
|
He B, Zhang W, Qiao J, Peng Z, Chai X. Melatonin protects against COPD by attenuating apoptosis and endoplasmic reticulum stress via upregulating SIRT1 expression in rats. Can J Physiol Pharmacol 2019; 97:386-391. [PMID: 30673309 DOI: 10.1139/cjpp-2018-0529] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The apoptosis of bronchial and alveolar epithelial cells plays a key role in chronic obstructive pulmonary disease (COPD). The endoplasmic reticulum (ER) stress induced by cigarette smoke contributes to apoptosis. Previous studies demonstrated that melatonin prevented the development of COPD. In addition, silent information regulator 1 (SIRT1) had a protective effect against COPD. However, it remains unclear whether SIRT1 is involved in the protection of melatonin against COPD. In this study, 32 male Wistar rats were randomly assigned to 4 groups: Control, COPD, COPD + Mel, and COPD + Mel + EX527. Rats were challenged with cigarette smoke and lipopolysaccharide with or without melatonin or EX527 (a selective inhibitor of SIRT1). The lung histopathology, apoptotic index, as well as the protein expressions of cleaved caspase-3, SIRT1, C/EBP homologous protein, and caspase-12 in the lung tissues were measured. These results demonstrated that melatonin attenuated apoptosis and ER stress in the lung tissues of rats with COPD. In addition, melatonin increased SIRT1 expression in lung tissues of rats with COPD, while inhibition of SIRT1 by EX527 upregulated ER stress and abolished the protective effect of melatonin against apoptosis. In conclusion, these findings suggested that melatonin protected against COPD by attenuating apoptosis and ER stress via upregulating SIRT1 expression in rats.
Collapse
Affiliation(s)
- Baimei He
- a Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, 410008, China.,b Department of Geriatric Medicine, Xiangya Hospital, Central South University, Changsha, 410008, China.,c National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Wenxuan Zhang
- d Department of Emergency Medicine, Second Xiangya Hospital, Central South University, Changsha, 410011, China.,e Emergency Medicine and Difficult Diseases Institute, Central South University, Changsha, 410011, China
| | - Jianfeng Qiao
- f Clinical Nursing Teaching and Research Section, Second Xiangya Hospital, Central South University, Changsha, 410011, China
| | - Zhenyu Peng
- d Department of Emergency Medicine, Second Xiangya Hospital, Central South University, Changsha, 410011, China.,e Emergency Medicine and Difficult Diseases Institute, Central South University, Changsha, 410011, China
| | - Xiangping Chai
- d Department of Emergency Medicine, Second Xiangya Hospital, Central South University, Changsha, 410011, China.,e Emergency Medicine and Difficult Diseases Institute, Central South University, Changsha, 410011, China
| |
Collapse
|
52
|
Sanders KA, Delker DA, Huecksteadt T, Beck E, Wuren T, Chen Y, Zhang Y, Hazel MW, Hoidal JR. RAGE is a Critical Mediator of Pulmonary Oxidative Stress, Alveolar Macrophage Activation and Emphysema in Response to Cigarette Smoke. Sci Rep 2019; 9:231. [PMID: 30659203 PMCID: PMC6338799 DOI: 10.1038/s41598-018-36163-z] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Accepted: 11/12/2018] [Indexed: 02/07/2023] Open
Abstract
The receptor for advanced glycation end products (RAGE), a cell membrane receptor, recognizes ligands produced by cigarette smoke (CS) and has been implicated in the pathogenesis of COPD. We demonstrate that deletion or pharmacologic inhibition of RAGE prevents development of CS-induced emphysema. To identify molecular pathways by which RAGE mediates smoking related lung injury we performed unbiased gene expression profiling of alveolar macrophages (AM) obtained from RAGE null and C57BL/6 WT mice exposed to CS for one week or four months. Pathway analysis of RNA expression identified a number of genes integral to the pathogenesis of COPD impacted by the absence of RAGE. Altered expression of antioxidant response genes and lung protein 4-HNE immunostaining suggest attenuated oxidative stress in the RAGE null mice despite comparable CS exposure and lung leukocyte burden as the WT mice. Reduced endoplasmic reticulum stress in response to CS exposure also was observed in the AM from RAGE null mice. These findings provide novel insight into the sources of oxidative stress, macrophage activation, and the pathogenesis of lung disease due to CS exposure.
Collapse
Affiliation(s)
- Karl A Sanders
- Division of Respiratory, Critical Care, and Occupational Pulmonary Medicine, University of Utah, Salt Lake City, Utah, USA
- Department of Internal Medicine, University of Utah, Salt Lake City, Utah, USA
- George E. Wahlen Department of Veterans Affairs Medical Center, Salt Lake City, Utah, USA
| | - Don A Delker
- Division of Gastroenterology, Hepatology, and Nutrition, University of Utah, Salt Lake City, Utah, USA
| | - Tom Huecksteadt
- George E. Wahlen Department of Veterans Affairs Medical Center, Salt Lake City, Utah, USA
| | - Emily Beck
- Division of Respiratory, Critical Care, and Occupational Pulmonary Medicine, University of Utah, Salt Lake City, Utah, USA
- Department of Internal Medicine, University of Utah, Salt Lake City, Utah, USA
- George E. Wahlen Department of Veterans Affairs Medical Center, Salt Lake City, Utah, USA
| | - Tanna Wuren
- George E. Wahlen Department of Veterans Affairs Medical Center, Salt Lake City, Utah, USA
| | - Yuntian Chen
- Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuxia Zhang
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Mark W Hazel
- Division of Gastroenterology, Hepatology, and Nutrition, University of Utah, Salt Lake City, Utah, USA
| | - John R Hoidal
- Division of Respiratory, Critical Care, and Occupational Pulmonary Medicine, University of Utah, Salt Lake City, Utah, USA.
- Department of Internal Medicine, University of Utah, Salt Lake City, Utah, USA.
- George E. Wahlen Department of Veterans Affairs Medical Center, Salt Lake City, Utah, USA.
| |
Collapse
|
53
|
Dickens JA, Malzer E, Chambers JE, Marciniak SJ. Pulmonary endoplasmic reticulum stress-scars, smoke, and suffocation. FEBS J 2019; 286:322-341. [PMID: 29323786 DOI: 10.1111/febs.14381] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Revised: 12/11/2017] [Accepted: 01/08/2018] [Indexed: 12/14/2022]
Abstract
Protein misfolding within the endoplasmic reticulum (ER stress) can be a cause or consequence of pulmonary disease. Mutation of proteins restricted to the alveolar type II pneumocyte can lead to inherited forms of pulmonary fibrosis, but even sporadic cases of pulmonary fibrosis appear to be strongly associated with activation of the unfolded protein response and/or the integrated stress response. Inhalation of smoke can impair protein folding and may be an important cause of pulmonary ER stress. Similarly, tissue hypoxia can lead to impaired protein homeostasis (proteostasis). But the mechanisms linking smoke and hypoxia to ER stress are only partially understood. In this review, we will examine the role of ER stress in the pathogenesis of lung disease by focusing on fibrosis, smoke, and hypoxia.
Collapse
Affiliation(s)
- Jennifer A Dickens
- Cambridge Institute for Medical Research (CIMR), University of Cambridge, UK
| | - Elke Malzer
- Cambridge Institute for Medical Research (CIMR), University of Cambridge, UK
| | - Joseph E Chambers
- Cambridge Institute for Medical Research (CIMR), University of Cambridge, UK
| | - Stefan J Marciniak
- Cambridge Institute for Medical Research (CIMR), University of Cambridge, UK
| |
Collapse
|
54
|
Kammerl IE, Caniard A, Merl-Pham J, Ben-Nissan G, Mayr CH, Mossina A, Geerlof A, Eickelberg O, Hauck SM, Sharon M, Meiners S. Dissecting the molecular effects of cigarette smoke on proteasome function. J Proteomics 2018; 193:1-9. [PMID: 30557664 DOI: 10.1016/j.jprot.2018.12.015] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Revised: 12/11/2018] [Accepted: 12/13/2018] [Indexed: 01/02/2023]
Abstract
Proteasome dysfunction is emerging as a novel pathomechanism for the development of chronic obstructive pulmonary disease (COPD), a major leading cause of death in the world. Cigarette smoke, one of the main risk factors for COPD, impairs proteasome function in vitro and in vivo. In the present study, we dissected the molecular changes induced by cigarette smoke on the proteasome in lung epithelial cells and mouse lungs. 26S proteasome pull-down, MS interactome, and stoichiometry analyses indicated that 26S proteasome complexes become instable in cigarette smoke-treated lung epithelial cells as well as in lungs of mice after three day smoke exposure. The interactome of the 26S was clearly altered in mouse lungs upon smoke exposure but not in cells after 24 h of smoke exposure. Using native MS analysis of purified 20S proteasomes, we observed some destabilization of 20S complexes purified from cigarette smoke-exposed cells in the absence of any dominant and inhibitory modification of proteasomal proteins. Taken together, our results suggest that cigarette smoke induces minor but detectable changes in the stability of 20S and 26S proteasome complexes which might contribute to imbalanced proteostasis in a chronic setting as observed in chronic lung diseases associated with cigarette smoking.
Collapse
Affiliation(s)
- Ilona E Kammerl
- Comprehensive Pneumology Center (CPC), University Hospital, Ludwig-Maximilians University, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Anne Caniard
- Comprehensive Pneumology Center (CPC), University Hospital, Ludwig-Maximilians University, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Juliane Merl-Pham
- Research Unit Protein Science, Helmholtz Zentrum München, Munich, Germany
| | - Gili Ben-Nissan
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, Israel
| | - Christoph H Mayr
- Comprehensive Pneumology Center (CPC), University Hospital, Ludwig-Maximilians University, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Alessandra Mossina
- Comprehensive Pneumology Center (CPC), University Hospital, Ludwig-Maximilians University, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Arie Geerlof
- Protein Expression and Purification Facility (PEPF), Institute of Structural Biology, Helmholtz Zentrum München, Neuherberg, Germany
| | - Oliver Eickelberg
- Division of Respiratory and Critical Care Medicine, University of Colorado, Aurora, USA
| | - Stefanie M Hauck
- Research Unit Protein Science, Helmholtz Zentrum München, Munich, Germany
| | - Michal Sharon
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, Israel
| | - Silke Meiners
- Comprehensive Pneumology Center (CPC), University Hospital, Ludwig-Maximilians University, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany.
| |
Collapse
|
55
|
Ding HB, Liu KX, Huang JF, Wu DW, Chen JY, Chen QS. RETRACTED: Protective effect of exogenous hydrogen sulfide on pulmonary artery endothelial cells by suppressing endoplasmic reticulum stress in a rat model of chronic obstructive pulmonary disease. Biomed Pharmacother 2018; 105:734-741. [PMID: 29908494 DOI: 10.1016/j.biopha.2018.05.131] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Revised: 04/24/2018] [Accepted: 05/27/2018] [Indexed: 01/08/2023] Open
Abstract
This article has been retracted: please see Elsevier Policy on Article Withdrawal (http://www.elsevier.com/locate/withdrawalpolicy). This article has been retracted at the request of the Editor-in-Chief. An Expression of Concern for this article was previously published while an investigation was conducted (see related editorial: https://doi.org/10.1016/j.biopha.2022.113812). This retraction notice supersedes the Expression of Concern published earlier. Concern was raised about the reliability of the Western blot data in Figure 4A, which appear to represent a distinct phenotype as found in many other publications, as detailed here: https://pubpeer.com/publications/029A84E50BD071A2088140723E3CF0; and here: https://docs.google.com/spreadsheets/d/1r0MyIYpagBc58BRF9c3luWNlCX8VUvUuPyYYXzxWvgY/edit#gid=262337249. Independent analysis confirmed the presence of suspected image duplications between the Western blots in Figure 4A and those contained in Yan et al (2017). The journal requested the corresponding author comment on these concerns and provide the associated raw data. The authors did not respond to this request and therefore the Editor-in-Chief decided to retract the article.
Collapse
Affiliation(s)
- Hai-Bo Ding
- Division of Respiratory and Critical Care Medicine, First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, Fujian Province, PR China.
| | - Kai-Xiong Liu
- Division of Respiratory and Critical Care Medicine, First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, Fujian Province, PR China
| | - Jie-Feng Huang
- Division of Respiratory and Critical Care Medicine, First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, Fujian Province, PR China
| | - Da-Wen Wu
- Division of Respiratory and Critical Care Medicine, First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, Fujian Province, PR China
| | - Jun-Ying Chen
- Central Lab, First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, Fujian Province, PR China
| | - Qing-Shi Chen
- Division of Respiratory and Critical Care Medicine, First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, Fujian Province, PR China
| |
Collapse
|
56
|
Tsai MJ, Chang WA, Jian SF, Chang KF, Sheu CC, Kuo PL. Possible mechanisms mediating apoptosis of bronchial epithelial cells in chronic obstructive pulmonary disease - A next-generation sequencing approach. Pathol Res Pract 2018; 214:1489-1496. [PMID: 30115538 DOI: 10.1016/j.prp.2018.08.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2018] [Revised: 07/24/2018] [Accepted: 08/06/2018] [Indexed: 12/16/2022]
Abstract
PURPOSE Chronic obstructive pulmonary disease (COPD) is a chronic inflammatory airway disease characterized by persistent airflow limitation. Apoptosis of pulmonary structural cells contributes to pulmonary destruction and dysfunction. This study aimed to explore the possible mechanisms underlying decreased cell proliferation and increased apoptosis of bronchial epithelial cells of COPD. MATERIALS AND METHODS The expression profiles of mRNAs and microRNAs in bronchial epithelial cells from a COPD patient and a normal subject were identified using next-generation sequencing (NGS) and analyzed using bioinformatic tools. RESULTS We identified 233 significantly upregulated and 204 significantly downregulated genes in COPD bronchial epithelial cells. The PI3K-Akt pathway was one of the most important dysregulated pathways in bronchial epithelial cells. We further identified that 3 genes involved in the PI3K-Akt signaling pathway, including IL6, F2R, and FGFR3, might be associated with inhibition of cell proliferation in bronchial epithelial cells, while 5 genes involved in the PI3K-Akt signaling pathway, including TLR4, IL6, F2R, FGFR3, and FGFR1, might be associated with apoptosis of bronchial epithelial cells. FGFR1 was also a predicted target for some up-regulated miRNAs in COPD bronchial epithelial cells, including hsa-miR-195-5p, hsa-miR-424-5p, and hsa-miR-6724-5p. CONCLUSION Our findings suggest PI3K-Akt signaling pathway plays an important role in COPD. We observed altered expression of apoptosis and cell proliferation-related genes that might contribute to the pathogenesis of COPD.
Collapse
Affiliation(s)
- Ming-Ju Tsai
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, No.100, Shih-Chuan 1st Rd., Kaohsiung, 807, Taiwan; Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, No. 100, Tz-You 1st Rd., Kaohsiung, 807, Taiwan; Department of Internal Medicine, School of Medicine, College of Medicine, Kaohsiung Medical University, No.100, Shih-Chuan 1st Rd., Kaohsiung, 807, Taiwan; Department of Respiratory Therapy, School of Medicine, College of Medicine, Kaohsiung Medical University, No.100, Shih-Chuan 1st Rd., Kaohsiung, 807, Taiwan.
| | - Wei-An Chang
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, No.100, Shih-Chuan 1st Rd., Kaohsiung, 807, Taiwan; Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, No. 100, Tz-You 1st Rd., Kaohsiung, 807, Taiwan.
| | - Shu-Fang Jian
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, No.100, Shih-Chuan 1st Rd., Kaohsiung, 807, Taiwan.
| | | | - Chau-Chyun Sheu
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, No.100, Shih-Chuan 1st Rd., Kaohsiung, 807, Taiwan; Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, No. 100, Tz-You 1st Rd., Kaohsiung, 807, Taiwan; Department of Internal Medicine, School of Medicine, College of Medicine, Kaohsiung Medical University, No.100, Shih-Chuan 1st Rd., Kaohsiung, 807, Taiwan; Department of Respiratory Therapy, School of Medicine, College of Medicine, Kaohsiung Medical University, No.100, Shih-Chuan 1st Rd., Kaohsiung, 807, Taiwan.
| | - Po-Lin Kuo
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, No.100, Shih-Chuan 1st Rd., Kaohsiung, 807, Taiwan.
| |
Collapse
|
57
|
Chen ACH, Burr L, McGuckin MA. Oxidative and endoplasmic reticulum stress in respiratory disease. Clin Transl Immunology 2018; 7:e1019. [PMID: 29928501 PMCID: PMC5999202 DOI: 10.1002/cti2.1019] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Revised: 05/01/2018] [Accepted: 05/03/2018] [Indexed: 01/01/2023] Open
Abstract
Oxidative stress and endoplasmic reticulum (ER) stress are related states that can occur in cells as part of normal physiology but occur frequently in diseases involving inflammation. In this article, we review recent findings relating to the role of oxidative and ER stress in the pathophysiology of acute and chronic nonmalignant diseases of the lung, including infections, cystic fibrosis, idiopathic pulmonary fibrosis and asthma. We also explore the potential of drugs targeting oxidative and ER stress pathways to alleviate disease.
Collapse
Affiliation(s)
- Alice C-H Chen
- Diamantina Institute Faculty of Medicine The University of Queensland Brisbane QLD Australia.,Department of Cell and Molecular Therapy Royal Prince Alfred Hospital Sydney NSW Australia
| | - Lucy Burr
- Department of Respiratory Medicine Mater Adult Hospital and Mater Research Institute - The University of Queensland Raymond Tce, South Brisbane QLD Australia
| | - Michael A McGuckin
- Inflammatory Disease Biology and Therapeutics Group Translational Research Institute Mater Research Institute - The University of Queensland Brisbane QLD Australia
| |
Collapse
|
58
|
The Unfolded Protein Response in Chronic Obstructive Pulmonary Disease. Ann Am Thorac Soc 2018; 13 Suppl 2:S138-45. [PMID: 27115948 DOI: 10.1513/annalsats.201506-320kv] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Accumulation of nonfunctional and potentially cytotoxic, misfolded proteins in chronic obstructive pulmonary disease (COPD) is believed to contribute to lung cell apoptosis, inflammation, and autophagy. Because of its fundamental role as a quality control system in protein metabolism, the "unfolded protein response" (UPR) is of potential importance in the pathogenesis of COPD. The UPR comprises a series of transcriptional, translational, and post-translational processes that decrease protein synthesis while enhancing protein folding capacity and protein degradation. Several studies have suggested that the UPR contributes to lung cell apoptosis and lung inflammation in at least some subjects with human COPD. However, information on the prevalence of the UPR in subjects with COPD, the lung cells that manifest a UPR, and the role of the UPR in the pathogenesis of COPD is extremely limited and requires additional study.
Collapse
|
59
|
Bodas M, Mazur S, Min T, Vij N. Inhibition of histone-deacetylase activity rescues inflammatory cystic fibrosis lung disease by modulating innate and adaptive immune responses. Respir Res 2018; 19:2. [PMID: 29301535 PMCID: PMC5755330 DOI: 10.1186/s12931-017-0705-8] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Accepted: 12/15/2017] [Indexed: 12/25/2022] Open
Abstract
Background Chronic lung disease resulting from dysfunctional cystic fibrosis transmembrane conductance regulator (CFTR) and NFκB-mediated neutrophilic-inflammation forms the basis of CF-related mortality. Here we aimed to evaluate if HDAC inhibition controls Pseudomonas-aeruginosa-lipopolysaccharide (Pa-LPS) induced airway inflammation and CF-lung disease. Methods For in vitro experiments, HEK293-cells were transfected with IL-8 or NFκB-firefly luciferase, and SV40-renilla- luciferase reporter constructs or ΔF508-CFTR-pCEP, followed by treatment with suberoylanilide hydroxamic acid (SAHA), Trichostatin-A (TSA) and/or TNFα. For murine studies, Cftr+/+ or Cftr−/− mice (n = 3) were injected/instilled with Pa-LPS and/or treated with SAHA or vehicle control. The progression of lung disease was monitored by quantifying changes in inflammatory markers (NFκB), cytokines (IL-6/IL-10), neutrophil activity (MPO, myeloperoxidase and/or NIMP-R14) and T-reg numbers. Results SAHA treatment significantly (p < 0.05) suppresses TNFα-induced NFκB and IL-8 reporter activities in HEK293-cells. Moreover, SAHA, Tubacin (selective HDAC6-inhibitor) or HDAC6-shRNAs controls CSE-induced ER-stress activities (p < 0.05). In addition, SAHA restores trafficking of misfolded-ΔF508-CFTR, by inducing protein levels of both B and C forms of CFTR. Murine studies using Cftr+/+ or Cftr−/− mice verified that SAHA controls Pa-LPS induced IL-6 levels, and neutrophil (MPO levels and/or NIMP-R14), NFκB-(inflammation) and Nrf2 (oxidative-stress marker) activities, while promoting FoxP3+ T-reg activity. Conclusion In summary, SAHA-mediated HDAC inhibition modulates innate and adaptive immune responses involved in pathogenesis and progression of inflammatory CF-lung disease. Electronic supplementary material The online version of this article (10.1186/s12931-017-0705-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Manish Bodas
- College of Medicine, Central Michigan University, 2630 Denison Drive, Room# 120 (Office) & 126-127 (Lab), Mt Pleasant, MI, USA.,Department of Pediatrics and Pulmonary Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Steven Mazur
- Department of Pediatrics and Pulmonary Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD, USA.,National Institute of Allergy and Infectious Diseases, National Institutes of Health, Integrated Research Facility at Fort Detrick, Fort Detrick, Frederick, MD, USA
| | - Taehong Min
- Department of Pediatrics and Pulmonary Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Genentech, 1 DNA Way, San Francisco, CA, USA
| | - Neeraj Vij
- College of Medicine, Central Michigan University, 2630 Denison Drive, Room# 120 (Office) & 126-127 (Lab), Mt Pleasant, MI, USA. .,Department of Pediatrics and Pulmonary Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD, USA. .,VIJ Biotech LLC, Baltimore, Maryland, USA.
| |
Collapse
|
60
|
Abstract
Aging is characterized by progressive deterioration of physiological integrity, decline in homeostasis, and degeneration of the tissues that occurs after the reproductive phase of life is complete, leading to impaired function. This deterioration is an important risk factor for chronic lung pathologies such as chronic obstructive pulmonary disease (COPD). COPD is a disease that develops gradually. Emphysematous changes in the lung take years to develop after exposure to cigarette smoke; hence, the vast majority of patients are elderly. There has been a dramatic increase in the life expectancy of the general population, resulting in an increased burden of chronic lung diseases. There is growing evidence that molecular mechanisms involved in aging may also play a role in COPD pathogenesis. Recently, the nine hallmarks of aging were identified. In this article, we will review the nine hallmarks of aging and how each hallmark contributes to the pathogenesis of COPD.
Collapse
Affiliation(s)
- Shweta P Kukrety
- Department of Internal Medicine, Creighton University, Omaha, NE, USA
| | - Jai D Parekh
- Department of Internal Medicine, Creighton University, Omaha, NE, USA
| | - Kristina L Bailey
- Department of Internal Medicine, University of Nebraska Medical Center; Research Service, Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, NE, USA
| |
Collapse
|
61
|
Brandsma CA, de Vries M, Costa R, Woldhuis RR, Königshoff M, Timens W. Lung ageing and COPD: is there a role for ageing in abnormal tissue repair? Eur Respir Rev 2017; 26:26/146/170073. [PMID: 29212834 DOI: 10.1183/16000617.0073-2017] [Citation(s) in RCA: 108] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Accepted: 09/20/2017] [Indexed: 11/05/2022] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is the fourth leading cause of death worldwide, with increasing prevalence, in particular in the elderly. COPD is characterised by abnormal tissue repair resulting in (small) airways disease and emphysema. There is accumulating evidence that ageing hallmarks are prominent features of COPD. These ageing hallmarks have been described in different subsets of COPD patients, in different lung compartments and also in a variety of cell types, and thus might contribute to different COPD phenotypes. A better understanding of the main differences and similarities between normal lung ageing and the pathology of COPD may improve our understanding of the mechanisms driving COPD pathology, in particular in those patients that develop the most severe form of COPD at a relatively young age, i.e. severe early-onset COPD patients.In this review, after introducing the main concepts of lung ageing and COPD pathology, we focus on the role of (abnormal) ageing in lung remodelling and repair in COPD. We discuss the current evidence for the involvement of ageing hallmarks in these pathological features of COPD. We also highlight potential novel treatment strategies and opportunities for future research based on our current knowledge of abnormal lung ageing in COPD.
Collapse
Affiliation(s)
- Corry-Anke Brandsma
- University of Groningen, University Medical Center Groningen, Dept of Pathology and Medical Biology, Groningen, The Netherlands .,University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD (GRIAC), Groningen, The Netherlands
| | - Maaike de Vries
- University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD (GRIAC), Groningen, The Netherlands.,University of Groningen, University Medical Center Groningen, Dept of Epidemiology, Groningen, The Netherlands
| | - Rita Costa
- Comprehensive Pneumology Center, Helmholtz Zentrum München, University Hospital of the Ludwig Maximilians University, Munich, Germany
| | - Roy R Woldhuis
- University of Groningen, University Medical Center Groningen, Dept of Pathology and Medical Biology, Groningen, The Netherlands.,University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD (GRIAC), Groningen, The Netherlands
| | - Melanie Königshoff
- Comprehensive Pneumology Center, Helmholtz Zentrum München, University Hospital of the Ludwig Maximilians University, Munich, Germany.,Division of Pulmonary Sciences and Critical Care Medicine, Dept of Medicine, University of Colorado, Denver, CO, USA.,Both authors contributed equally
| | - Wim Timens
- University of Groningen, University Medical Center Groningen, Dept of Pathology and Medical Biology, Groningen, The Netherlands.,University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD (GRIAC), Groningen, The Netherlands.,Both authors contributed equally
| |
Collapse
|
62
|
Lin F, Liao C, Sun Y, Zhang J, Lu W, Bai Y, Liao Y, Li M, Ni X, Hou Y, Qi Y, Chen Y. Hydrogen Sulfide Inhibits Cigarette Smoke-Induced Endoplasmic Reticulum Stress and Apoptosis in Bronchial Epithelial Cells. Front Pharmacol 2017; 8:675. [PMID: 29033840 PMCID: PMC5625329 DOI: 10.3389/fphar.2017.00675] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Accepted: 09/08/2017] [Indexed: 01/23/2023] Open
Abstract
Background: Apoptosis of lung structural cells contributes to the process of lung damage and remodeling in chronic obstructive pulmonary disease (COPD). Our previous studies demonstrated that exogenous hydrogen sulfide (H2S) can reduce the lung tissue pathology score, anti-inflammation and anti-oxidation effects in COPD, but the effect of H2S in regulating cigarette smoke (CS) induced bronchial epithelial cell apoptosis and the underlying mechanisms are not clear. Objectives: To investigate the effect of H2S on CS induced endoplasmic reticulum stress (ERS) and bronchial epithelial cell apoptosis. Methods: Male Sprague–Dawley rats randomly divided into four groups for treatment: control, CS, NaHS + CS, and propargylglycine (PPG) + CS. The rats in the CS group were exposed to CS generated from 20 commercial unfiltered cigarettes for 4 h/day, 7 days/week for 4 months. Since the beginning of the third month, freshly prepared NaHS (14 μmol/kg) and PPG (37.5 mg/kg) were intraperitoneally administered 30 min before CS-exposure in the NaHS and PPG groups. 16HBE cells were pretreated with Taurine (10 mM), 5 mmol/L 4-phenylbutyric acid (4-PBA) or NaHS (100, 200, and 400 μM) for 30 min, and then cells were exposed to 40 μmol/L nicotine for 72 h. ERS markers (GRP94, GRP78) and ERS-mediated apoptosis markers 4-C/EBP homologous protein (CHOP), caspase-3 and caspase-12 were assessed in rat lung tissues and human bronchial epithelial cells. The apoptotic bronchial epithelial cells were detected by Hoechst staining in vitro and TUNEL staining in vivo. Results: In CS exposed rats, peritoneal injection of NaHS significantly inhibited CS induced overexpression ERS-mediated apoptosis markers and upregulation of apoptotic rate in rat lungs, and inhibiting the endogenous H2S production by peritoneal injection of PPG exacerbated these effects. In the nicotine-exposed bronchial epithelial cells, appropriate concentration of NaHS and ERS inhibitors taurine and 4-PBA inhibited nicotine-induced upregulation of apoptotic rate and overexpression of ERS-mediated apoptosis markers. Conclusion: H2S inhibited lung tissue damage by attenuating CS induced ERS in rat lung and exogenous H2S attenuated nicotine induced ERS-mediated apoptosis in bronchial epithelial cells.
Collapse
Affiliation(s)
- Fan Lin
- Department of Respiratory and Critical Care Medicine, Peking University Third Hospital, Beijing, China.,Department of Respiratory Medicine, Zhejiang Provincial People's Hospital, Hangzhou, China
| | - Chengcheng Liao
- Department of Respiratory and Critical Care Medicine, Peking University Third Hospital, Beijing, China
| | - Yun Sun
- Department of Respiratory and Critical Care Medicine, Peking University Third Hospital, Beijing, China
| | - Jinsheng Zhang
- Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Peking University Health Science Center, Beijing, China
| | - Weiwei Lu
- Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Peking University Health Science Center, Beijing, China
| | - Yu Bai
- Department of Respiratory and Critical Care Medicine, Peking University Third Hospital, Beijing, China
| | - Yixuan Liao
- Department of Respiratory and Critical Care Medicine, Peking University Third Hospital, Beijing, China
| | - Minxia Li
- Department of Respiratory and Critical Care Medicine, Peking University Third Hospital, Beijing, China
| | - Xianqiang Ni
- Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Peking University Health Science Center, Beijing, China
| | - Yuelong Hou
- Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Peking University Health Science Center, Beijing, China
| | - Yongfen Qi
- Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Peking University Health Science Center, Beijing, China
| | - Yahong Chen
- Department of Respiratory and Critical Care Medicine, Peking University Third Hospital, Beijing, China
| |
Collapse
|
63
|
Bodas M, Silverberg D, Walworth K, Brucia K, Vij N. Augmentation of S-Nitrosoglutathione Controls Cigarette Smoke-Induced Inflammatory-Oxidative Stress and Chronic Obstructive Pulmonary Disease-Emphysema Pathogenesis by Restoring Cystic Fibrosis Transmembrane Conductance Regulator Function. Antioxid Redox Signal 2017; 27:433-451. [PMID: 28006950 PMCID: PMC5564030 DOI: 10.1089/ars.2016.6895] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
AIMS Cigarette smoke (CS)-mediated acquired cystic fibrosis transmembrane conductance regulator (CFTR)-dysfunction, autophagy-impairment, and resulting inflammatory-oxidative/nitrosative stress leads to chronic obstructive pulmonary disease (COPD)-emphysema pathogenesis. Moreover, nitric oxide (NO) signaling regulates lung function decline, and low serum NO levels that correlates with COPD severity. Hence, we aim to evaluate here the effects and mechanism(s) of S-nitrosoglutathione (GSNO) augmentation in regulating inflammatory-oxidative stress and COPD-emphysema pathogenesis. RESULTS Our data shows that cystic fibrosis transmembrane conductance regulator (CFTR) colocalizes with aggresome bodies in the lungs of COPD subjects with increasing emphysema severity (Global Initiative for Chronic Obstructive Lung Disease [GOLD] I - IV) compared to nonemphysema controls (GOLD 0). We further demonstrate that treatment with GSNO or S-nitrosoglutathione reductase (GSNOR)-inhibitor (N6022) significantly inhibits cigarette smoke extract (CSE; 5%)-induced decrease in membrane CFTR expression by rescuing it from ubiquitin (Ub)-positive aggresome bodies (p < 0.05). Moreover, GSNO restoration significantly (p < 0.05) decreases CSE-induced reactive oxygen species (ROS) activation and autophagy impairment (decreased accumulation of ubiquitinated proteins in the insoluble protein fractions and restoration of autophagy flux). In addition, GSNO augmentation inhibits protein misfolding as CSE-induced colocalization of ubiquitinated proteins and LC3B (in autophagy bodies) is significantly reduced by GSNO/N6022 treatment. We verified using the preclinical COPD-emphysema murine model that chronic CS (Ch-CS)-induced inflammation (interleukin [IL]-6/IL-1β levels), aggresome formation (perinuclear coexpression/colocalization of ubiquitinated proteins [Ub] and p62 [impaired autophagy marker], and CFTR), oxidative/nitrosative stress (p-Nrf2, inducible nitric oxide synthase [iNOS], and 3-nitrotyrosine expression), apoptosis (caspase-3/7 activity), and alveolar airspace enlargement (Lm) are significantly (p < 0.05) alleviated by augmenting airway GSNO levels. As a proof of concept, we demonstrate that GSNO augmentation suppresses Ch-CS-induced perinuclear CFTR protein accumulation (p < 0.05), which restores both acquired CFTR dysfunction and autophagy impairment, seen in COPD-emphysema subjects. INNOVATION GSNO augmentation alleviates CS-induced acquired CFTR dysfunction and resulting autophagy impairment. CONCLUSION Overall, we found that augmenting GSNO levels controls COPD-emphysema pathogenesis by reducing CS-induced acquired CFTR dysfunction and resulting autophagy impairment and chronic inflammatory-oxidative stress. Antioxid. Redox Signal. 27, 433-451.
Collapse
Affiliation(s)
- Manish Bodas
- 1 College of Medicine, Central Michigan University , Mt. Pleasant, Michigan
| | - David Silverberg
- 1 College of Medicine, Central Michigan University , Mt. Pleasant, Michigan
| | - Kyla Walworth
- 1 College of Medicine, Central Michigan University , Mt. Pleasant, Michigan
| | - Kathryn Brucia
- 1 College of Medicine, Central Michigan University , Mt. Pleasant, Michigan
| | - Neeraj Vij
- 1 College of Medicine, Central Michigan University , Mt. Pleasant, Michigan.,2 Department of Pediatrics and Pulmonary Medicine, The Johns Hopkins University School of Medicine , Baltimore, Maryland
| |
Collapse
|
64
|
Govindaraju VK, Bodas M, Vij N. Cigarette smoke induced autophagy-impairment regulates AMD pathogenesis mechanisms in ARPE-19 cells. PLoS One 2017; 12:e0182420. [PMID: 28767736 PMCID: PMC5540403 DOI: 10.1371/journal.pone.0182420] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Accepted: 07/18/2017] [Indexed: 12/01/2022] Open
Abstract
Age related macular degeneration (AMD) is one of the leading causes of blindness. Genetics, environmental insult, and age-related factors all play a key role in altering proteostasis, the homeostatic process regulating protein synthesis, degradation and processing. These factors also play a role in the pathogenesis of AMD and it has been well established that cigarette smoking (CS) initiates AMD pathogenic mechanisms. The primary goal of this study is to elucidate whether CS can induce proteostasis/autophagy-impairment in retinal pigment epithelial (RPE) cells. In our preliminary analysis, it was found that cigarette smoke extract (CSE) induces accumulation of ubiquitinated proteins in the insoluble protein fraction (p < 0.01), which was subsequently mitigated through cysteamine (p < 0.01) or fisetin (p < 0.05) treatment. Further, it was verified that these CSE induced ubiquitinated proteins accumulated in the peri-nuclear spaces (p<0.05) that were cleared- off with cysteamine (p < 0.05) or fisetin (p < 0.05). Moreover, CSE-induced aggresome-formation (LC3B-GFP and Ub-RFP co-localization) and autophagy-flux impairment was significantly (p<0.01) mitigated by cysteamine (p<0.05) or fisetin (p<0.05) treatment, indicating the restoration of CSE-mediated autophagy-impairment. CSE treatment was also found to induce intracellular reactive oxygen species (ROS, p < 0.001) while impacting cell viability (p < 0.001), which was quantified using CMH2DCFDA-dye (ROS) and MTS (proliferation) or propodium iodide staining (cell viability) assays, respectively. Moreover, cysteamine and fisetin treatment ameliorated CS-mediated ROS production (p < 0.05) and diminished cell viability (p < 0.05). Lastly, CSE was found to induce cellular senescence (p < 0.001), which was significantly ameliorated by cysteamine (p < 0.001) or fisetin (p < 0.001). In conclusion, our study indicates that CS induced proteostasis/autophagy-impairment regulates mechanisms associated with AMD pathogenesis. Moreover, autophagy-inducing drugs such as cysteamine or fisetin can ameliorate AMD pathogenesis mechanisms that warrant further investigation in pre-clinical murine models.
Collapse
Affiliation(s)
- Viren Kumar Govindaraju
- College of Medicine, Central Michigan University, Mt Pleasant, Michigan, United States of America
| | - Manish Bodas
- College of Medicine, Central Michigan University, Mt Pleasant, Michigan, United States of America
| | - Neeraj Vij
- College of Medicine, Central Michigan University, Mt Pleasant, Michigan, United States of America
- Department of Pediatrics and Pulmonary Medicine, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| |
Collapse
|
65
|
Bodas M, Patel N, Silverberg D, Walworth K, Vij N. Master Autophagy Regulator Transcription Factor EB Regulates Cigarette Smoke-Induced Autophagy Impairment and Chronic Obstructive Pulmonary Disease-Emphysema Pathogenesis. Antioxid Redox Signal 2017; 27:150-167. [PMID: 27835930 PMCID: PMC5510670 DOI: 10.1089/ars.2016.6842] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Revised: 11/06/2016] [Accepted: 11/11/2016] [Indexed: 12/14/2022]
Abstract
AIMS Recent studies have shown that cigarette smoke (CS)-induced oxidative stress impairs autophagy, resulting in aggresome-formation that correlates with severity of chronic obstructive pulmonary disease (COPD)-emphysema, although the specific step in autophagy pathway that is impaired is unknown. Hence, in this study, we aimed to evaluate the role of master autophagy transcription factor EB (TFEB) in CS-induced COPD-emphysema pathogenesis. RESULTS We first observed that TFEB accumulates in perinuclear spaces as aggresome-bodies in COPD lung tissues of tobacco smokers and severe emphysema subjects, compared with non-emphysema or nonsmoker controls. Next, Beas2b cells and C57BL/6 mice were exposed to either cigarette smoke extract (CSE) or subchronic-CS (sc-CS), followed by treatment with potent TFEB-inducing drug, gemfibrozil (GEM, or fisetin as an alternate), to experimentally verify the role of TFEB in COPD. Our in vitro results indicate that GEM/fisetin-mediated TFEB induction significantly (p < 0.05) decreases CSE-induced autophagy-impairment (Ub/LC3B reporter and autophagy flux assay) and resulting aggresome-formation (Ub/p62 coexpression/accumulation; immunoblotting and staining) by controlling reactive oxygen species (ROS) activity. Intriguingly, we observed that CS induces TFEB accumulation in the insoluble protein fractions of Beas2b cells, which shows a partial rescue with GEM treatment. Moreover, TFEB knockdown induces oxidative stress, autophagy-impairment, and senescence, which can all be mitigated by GEM-mediated TFEB induction. Finally, in vivo studies were used to verify that CS-induced autophagy-impairment (increased Ub, p62, and valosin-containing protein in the insoluble protein fractions of lung/cell lysates), inflammation (interleukin-6 [IL-6] levels in bronchoalveolar lavage fluid and iNOS expression in lung sections), apoptosis (caspase-3/7), and resulting emphysema (hematoxylin and eosin [H&E]) can be controlled by GEM-mediated TFEB induction (p < 0.05). INNOVATION CS exposure impairs autophagy in COPD-emphysema by inducing perinuclear localization of master autophagy regulator, TFEB, to aggresome-bodies. CONCLUSION TFEB-inducing drug(s) can control CS-induced TFEB/autophagy-impairment and COPD-emphysema pathogenesis. Antioxid. Redox Signal. 27, 150-167.
Collapse
Affiliation(s)
- Manish Bodas
- College of Medicine, Central Michigan University, Mount Pleasant, Michigan
| | - Neel Patel
- College of Medicine, Central Michigan University, Mount Pleasant, Michigan
| | - David Silverberg
- College of Medicine, Central Michigan University, Mount Pleasant, Michigan
| | - Kyla Walworth
- College of Medicine, Central Michigan University, Mount Pleasant, Michigan
| | - Neeraj Vij
- College of Medicine, Central Michigan University, Mount Pleasant, Michigan
- Department of Pediatrics and Pulmonary Medicine, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
66
|
Bahmed K, Messier EM, Zhou W, Tuder RM, Freed CR, Chu HW, Kelsen SG, Bowler RP, Mason RJ, Kosmider B. DJ-1 Modulates Nuclear Erythroid 2-Related Factor-2-Mediated Protection in Human Primary Alveolar Type II Cells in Smokers. Am J Respir Cell Mol Biol 2017; 55:439-49. [PMID: 27093578 DOI: 10.1165/rcmb.2015-0304oc] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Cigarette smoke (CS) is a main source of oxidative stress and a key risk factor for emphysema, which consists of alveolar wall destruction. Alveolar type (AT) II cells are in the gas exchange regions of the lung. We isolated primary ATII cells from deidentified organ donors whose lungs were not suitable for transplantation. We analyzed the cell injury obtained from nonsmokers, moderate smokers, and heavy smokers. DJ-1 protects cells from oxidative stress and induces nuclear erythroid 2-related factor-2 (Nrf2) expression, which activates the antioxidant defense system. In ATII cells isolated from moderate smokers, we found DJ-1 expression by RT-PCR, and Nrf2 and heme oxygenase (HO)-1 translocation by Western blotting and immunocytofluorescence. In ATII cells isolated from heavy smokers, we detected Nrf2 and HO-1 cytoplasmic localization. Moreover, we found high oxidative stress, as detected by 4-hydroxynonenal (4-HNE) (immunoblotting), inflammation by IL-8 and IL-6 levels by ELISA, and apoptosis by terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) assay in ATII cells obtained from heavy smokers. Furthermore, we detected early DJ-1 and late Nrf2 expression after ATII cell treatment with CS extract. We also overexpressed DJ-1 by adenovirus construct and found that this restored Nrf2 and HO-1 expression and induced nuclear translocation in heavy smokers. Moreover, DJ-1 overexpression also decreased ATII cell apoptosis caused by CS extract in vitro. Our results indicate that DJ-1 activates the Nrf2-mediated antioxidant defense system. Furthermore, DJ-1 overexpression can restore the impaired Nrf2 pathway, leading to ATII cell protection in heavy smokers. This suggests a potential therapeutic strategy for targeting DJ-1 in CS-related lung diseases.
Collapse
Affiliation(s)
- Karim Bahmed
- 1 Department of Thoracic Medicine and Surgery.,2 Center for Inflammation, Translational and Clinical Lung Research, and
| | - Elise M Messier
- 3 Department of Medicine, National Jewish Health, Denver, Colorado
| | - Wenbo Zhou
- 4 Department of Medicine, Division of Clinical Pharmacology, University of Colorado Denver, Aurora, Colorado; and
| | - Rubin M Tuder
- 5 Department of Medicine, Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado School of Medicine, Aurora, Colorado
| | - Curt R Freed
- 4 Department of Medicine, Division of Clinical Pharmacology, University of Colorado Denver, Aurora, Colorado; and
| | - Hong Wei Chu
- 3 Department of Medicine, National Jewish Health, Denver, Colorado
| | - Steven G Kelsen
- 1 Department of Thoracic Medicine and Surgery.,2 Center for Inflammation, Translational and Clinical Lung Research, and
| | - Russell P Bowler
- 3 Department of Medicine, National Jewish Health, Denver, Colorado
| | - Robert J Mason
- 3 Department of Medicine, National Jewish Health, Denver, Colorado
| | - Beata Kosmider
- 1 Department of Thoracic Medicine and Surgery.,2 Center for Inflammation, Translational and Clinical Lung Research, and.,6 Department of Physiology, Temple University, Philadelphia, Pennsylvania.,3 Department of Medicine, National Jewish Health, Denver, Colorado
| |
Collapse
|
67
|
Lin L, Yin Y, Hou G, Han D, Kang J, Wang Q. Ursolic acid attenuates cigarette smoke-induced emphysema in rats by regulating PERK and Nrf2 pathways. Pulm Pharmacol Ther 2017; 44:111-121. [PMID: 28347799 DOI: 10.1016/j.pupt.2017.03.014] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2016] [Revised: 02/28/2017] [Accepted: 03/23/2017] [Indexed: 12/30/2022]
Abstract
OBJECTIVE Ursolic acid (UA) is widely distributed in natural plants to against oxidation, virus, inflammation, tumor, and has been widely used in the pharmaceutical and cosmetics. However, its effect on emphysema of chronic obstructive pulmonary disease (COPD) is unknown. Unfolded protein response is involved in pathogenesis of COPD through PERK pathway. Nuclear erythroid-related factor 2 (Nrf2) regulates antioxidant defensive mechanism in COPD. This study was to explore effect and mechanism of UA on cigarette smoke (CS)-induced rat emphysema. MATERIALS AND METHODS 50 Wistar rats were divided into 5 groups (n = 10 each): rats were exposed to CS for 12 weeks in absence (CS group) or presence of UA at different doses. Control group was treated with UA vehicle only. Histopathology, apoptosis, key protein expression of PERK and Nrf2 pathway were determined in lung tissues. Oxidative stress levels in lung were represented by 8-OHdG, MDA and GSH levels. RESULTS Emphysema-related pathology, based on inter-alveolar wall distance and alveolar density, was less severe in UA groups than in CS group. Compared with CS group, UA treatment down-regulated PERK pathway protein expression, up-regulated expression of Bcl-2 and down-regulated expression of Bax, Cleaved-Caspase3 and Cleaved-Caspase12. Moreover, UA decreased number of apoptotic cells in rat lungs. UA also up-regulated protein expression of Nrf2/ARE pathway and GSH level, decreased expression of oxidant stress factor 8-OHdG and MDA. These improvements were in accordance with attenuation of severity of emphysema. CONCLUSIONS UA attenuates CS-induced rat emphysema by down-regulating PERK pathway to alleviate CS-induced apoptosis in lung, and up-regulating Nrf2 pathway to improve cigarette smoke-induced oxidant stress in rat lungs.
Collapse
Affiliation(s)
- Li Lin
- Institute of Respiratory Disease, The First Hospital of China Medical University, Shenyang 110001, China
| | - Yan Yin
- Institute of Respiratory Disease, The First Hospital of China Medical University, Shenyang 110001, China
| | - Gang Hou
- Institute of Respiratory Disease, The First Hospital of China Medical University, Shenyang 110001, China
| | - Dan Han
- Institute of Respiratory Disease, The First Hospital of China Medical University, Shenyang 110001, China
| | - Jian Kang
- Institute of Respiratory Disease, The First Hospital of China Medical University, Shenyang 110001, China
| | - Qiuyue Wang
- Institute of Respiratory Disease, The First Hospital of China Medical University, Shenyang 110001, China.
| |
Collapse
|
68
|
Wan ES, Li Y, Lao T, Qiu W, Jiang Z, Mancini JD, Owen CA, Clish C, DeMeo DL, Silverman EK, Zhou X. Metabolomic profiling in a Hedgehog Interacting Protein (Hhip) murine model of chronic obstructive pulmonary disease. Sci Rep 2017; 7:2504. [PMID: 28566717 PMCID: PMC5451406 DOI: 10.1038/s41598-017-02701-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Accepted: 04/13/2017] [Indexed: 01/22/2023] Open
Abstract
Genetic variants annotated to the hedgehog interacting protein (HHIP) are robustly associated with chronic obstructive pulmonary disease (COPD). Hhip haploinsufficiency in mice leads to increased susceptibility towards the development of emphysema following exposure to chronic cigarette smoke (CS). To explore the molecular pathways which contribute to increased susceptibility, we performed metabolomic profiling using high performance liquid chromatography tandem mass spectroscopy (LC/MS-MS) on plasma, urine, and lung tissue of Hhip+/− heterozygotes and wild type (Hhip+/+) C57/BL6 mice exposed to either room-air or CS for six months. Univariate comparisons between groups were made with a combined fold change ≥2 and Student’s t-test p-value < 0.05 to denote significance; associations with mean alveolar chord length (MACL), a quantitative measure of emphysema, and gene-by-environment interactions were examined using empiric Bayes-mediated linear models. Decreased urinary excretion of cotinine despite comparable plasma levels was observed in Hhip+/− heterozygotes; a strong gene-by-smoking association was also observed. Correlations between MACL and markers of oxidative stress such as urinary methionine sulfoxide were observed in Hhip+/− but not in Hhip+/+ mice. Metabolite set enrichment analyses suggest reduced antioxidant capacity and alterations in macronutrient metabolism contribute to increased susceptibility to chronic CS-induced oxidative stress in Hhip haploinsufficiency states.
Collapse
Affiliation(s)
- Emily S Wan
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, MA, USA. .,Division of Pulmonary and Critical Care, Brigham and Women's Hospital, Boston, MA, USA. .,Pulmonary Section, VA Boston Healthcare System, Jamaica Plain, MA, USA.
| | - Yan Li
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Taotao Lao
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Weiliang Qiu
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Zhiqiang Jiang
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - John D Mancini
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Caroline A Owen
- Division of Pulmonary and Critical Care, Brigham and Women's Hospital, Boston, MA, USA.,Lovelace Respiratory Research Institute, Albuquerque, NM, USA
| | | | - Dawn L DeMeo
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, MA, USA.,Division of Pulmonary and Critical Care, Brigham and Women's Hospital, Boston, MA, USA
| | - Edwin K Silverman
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, MA, USA.,Division of Pulmonary and Critical Care, Brigham and Women's Hospital, Boston, MA, USA
| | - Xiaobo Zhou
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, MA, USA
| |
Collapse
|
69
|
Dalle-Donne I, Colombo G, Gornati R, Garavaglia ML, Portinaro N, Giustarini D, Bernardini G, Rossi R, Milzani A. Protein Carbonylation in Human Smokers and Mammalian Models of Exposure to Cigarette Smoke: Focus on Redox Proteomic Studies. Antioxid Redox Signal 2017; 26:406-426. [PMID: 27393565 DOI: 10.1089/ars.2016.6772] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
SIGNIFICANCE Oxidative stress is one mechanism whereby tobacco smoking affects human health, as reflected by increased levels of several biomarkers of oxidative stress/damage isolated from tissues and biological fluids of active and passive smokers. Many investigations of cigarette smoke (CS)-induced oxidative stress/damage have been carried out in mammalian animal and cellular models of exposure to CS. Animal models allow the investigation of many parameters that are similar to those measured in human smokers. In vitro cell models may provide new information on molecular and functional differences between cells of smokers and nonsmokers. Recent Advances: Over the past decade or so, a growing number of researches highlighted that CS induces protein carbonylation in different tissues and body fluids of smokers as well as in in vivo and in vitro models of exposure to CS. CRITICAL ISSUES We review recent findings on protein carbonylation in smokers and models thereof, focusing on redox proteomic studies. We also discuss the relevance and limitations of these models of exposure to CS and critically assess the congruence between the smoker's condition and laboratory models. FUTURE DIRECTIONS The identification of protein targets is crucial for understanding the mechanism(s) by which carbonylated proteins accumulate and potentially affect cellular functions. Recent progress in redox proteomics allows the enrichment, identification, and characterization of specific oxidative protein modifications, including carbonylation. Therefore, redox proteomics can be a powerful tool to gain new insights into the onset and/or progression of CS-related diseases and to develop strategies to prevent and/or treat them. Antioxid. Redox Signal. 26, 406-426.
Collapse
Affiliation(s)
| | - Graziano Colombo
- 1 Department of Biosciences, Università degli Studi di Milano , Milan, Italy
| | - Rosalba Gornati
- 2 Department of Biotechnology and Life Sciences, University of Insubria , Varese, Italy
| | - Maria L Garavaglia
- 1 Department of Biosciences, Università degli Studi di Milano , Milan, Italy
| | - Nicola Portinaro
- 3 Department of Medical Biotechnology and Translational Medicine, Università degli Studi di Milano and Pediatric Orthopaedic Unit, Humanitas Clinical and Research Center , Rozzano (Milan), Italy
| | | | - Giovanni Bernardini
- 2 Department of Biotechnology and Life Sciences, University of Insubria , Varese, Italy
| | - Ranieri Rossi
- 4 Department of Life Sciences, University of Siena , Siena, Italy
| | - Aldo Milzani
- 1 Department of Biosciences, Università degli Studi di Milano , Milan, Italy
| |
Collapse
|
70
|
Patel N, Trumph CD, Bodas M, Vij N. Role of second-hand smoke (SHS)-induced proteostasis/autophagy impairment in pediatric lung diseases. Mol Cell Pediatr 2017; 4:3. [PMID: 28150141 PMCID: PMC5289127 DOI: 10.1186/s40348-017-0069-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2016] [Accepted: 01/02/2017] [Indexed: 12/11/2022] Open
Abstract
Background Exposure to second-hand tobacco smoke (SHS) is one of the prime risk factors for chronic lung disease development. Smoking during pregnancy may lead to birth defects in the newborn that include pulmonary dysfunction, increased susceptibility to opportunistic pathogens, or initiation of childhood respiratory manifestations such as bronchopulmonary dysplasia (BPD). Moreover, exposure to SHS in early childhood can have negative impact on lung health, although the exact mechanisms are unclear. Autophagy is a crucial proteostatic mechanism modulated by cigarette smoke (CS) in adult lungs. Here, we sought to investigate whether SHS exposure impairs autophagy in pediatric lungs. Methods Pregnant C57BL/6 mice were exposed to room air or SHS for 14 days. The newborn pups were subsequently exposed to room air or SHS (5 h/day) for 1 or 14 days, and lungs were harvested. Soluble and insoluble protein fractions isolated from pediatric mice lungs were subjected to immunoblotting for ubiquitin (Ub), p62, VCP, HIF-1α, and β-actin. Results Our data shows that short-term exposure to SHS (1 or 14 days) leads to proteostasis and autophagy-impairment as evident by significant increase in accumulation of ubiquitinated proteins (Ub), p62 (impaired-autophagy marker) and valosin-containing protein (VCP) in the insoluble protein fractions of pediatric mice lungs. Moreover, increased HIF-1α levels in SHS-exposed mice lungs points towards a novel mechanism for SHS-induced lung disease initiation in the pediatric population. Validating the in vivo studies, we demonstrate that treatment of human bronchial epithelial cells (Beas2b cells) with the proteasome inhibitor (MG-132) induces HIF-1α expression that is controlled by co-treatment with autophagy-inducing drug, cysteamine. Conclusions SHS-exposure induced proteostasis/autophagy impairment can mediate the initiation of chronic lung disease in pediatric subjects. Hence, our data warrants the evaluation of proteostasis/autophagy-inducing drugs, such as cysteamine, as a potential therapeutic intervention strategy for SHS-induced pediatric lung diseases.
Collapse
Affiliation(s)
- Neel Patel
- College of Medicine, Central Michigan University, College of Medicine Research Building, 2630 Denison Drive, Room# 120, Room# 120 (Office) and 126-127 (Lab), Mt Pleasant, MI, 48859, USA
| | - Christopher D Trumph
- College of Medicine, Central Michigan University, College of Medicine Research Building, 2630 Denison Drive, Room# 120, Room# 120 (Office) and 126-127 (Lab), Mt Pleasant, MI, 48859, USA
| | - Manish Bodas
- College of Medicine, Central Michigan University, College of Medicine Research Building, 2630 Denison Drive, Room# 120, Room# 120 (Office) and 126-127 (Lab), Mt Pleasant, MI, 48859, USA
| | - Neeraj Vij
- College of Medicine, Central Michigan University, College of Medicine Research Building, 2630 Denison Drive, Room# 120, Room# 120 (Office) and 126-127 (Lab), Mt Pleasant, MI, 48859, USA. .,Department of Pediatric Respiratory Science, The Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
71
|
Cervical Cancer Cell Line Secretome Highlights the Roles of Transforming Growth Factor-Beta-Induced Protein ig-h3, Peroxiredoxin-2, and NRF2 on Cervical Carcinogenesis. BIOMED RESEARCH INTERNATIONAL 2017; 2017:4180703. [PMID: 28261610 PMCID: PMC5316418 DOI: 10.1155/2017/4180703] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Revised: 11/16/2016] [Accepted: 11/24/2016] [Indexed: 02/07/2023]
Abstract
Cancer cells acquire unique secretome compositions that contribute to tumor development and metastasis. The aim of our study was to elucidate the biological processes involved in cervical cancer, by performing a proteomic analysis of the secretome from the following informative cervical cell lines: SiHa (HPV16+), HeLa (HPV18+), C33A (HPV-), and HCK1T (normal). Proteins were analyzed by 2D gel electrophoresis coupled to MALDI-TOF-MS. Enrichment of secreted proteins with characteristic profiles for each cell line was followed by the identification of differentially expressed proteins. Particularly, transforming growth factor-beta-induced protein ig-h3 (Beta ig-h3) and peroxiredoxin-2 (PRDX2) overexpression in the secretome of cancer cell lines was detected and confirmed by Western blot. Bioinformatics analysis identified the transcription factor NRF2 as a regulator of differentially expressed proteins in the cervical cancer secretome. NRF2 levels were measured by both Western blot and Multiple Reaction Monitoring (MRM) in the total cell extract of the four cell lines. NRF2 was upregulated in SiHa and C33A compared to HCK1T. In conclusion, the secreted proteins identified in cervical cancer cell lines indicate that aberrant NRF2-mediated oxidative stress response (OSR) is a prominent feature of cervical carcinogenesis.
Collapse
|
72
|
Salubrinal attenuates right ventricular hypertrophy and dysfunction in hypoxic pulmonary hypertension of rats. Vascul Pharmacol 2016; 87:190-198. [DOI: 10.1016/j.vph.2016.09.009] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Revised: 08/16/2016] [Accepted: 09/23/2016] [Indexed: 11/23/2022]
|
73
|
Schumacher FR, Schubert S, Hannus M, Sönnichsen B, Ittrich C, Kreideweiss S, Kurz T, Rippmann JF. RNAi Screen for NRF2 Inducers Identifies Targets That Rescue Primary Lung Epithelial Cells from Cigarette Smoke Induced Radical Stress. PLoS One 2016; 11:e0166352. [PMID: 27832175 PMCID: PMC5104413 DOI: 10.1371/journal.pone.0166352] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Accepted: 10/27/2016] [Indexed: 12/30/2022] Open
Abstract
Chronic Obstructive Pulmonary Disease (COPD) is a highly prevalent condition characterized by inflammation and progressive obstruction of the airways. At present, there is no treatment that suppresses the chronic inflammation of the disease, and COPD patients often succumb to the condition. Excessive oxidative stress caused by smoke inhalation is a major driving force of the disease. The transcription factor NRF2 is a critical player in the battle against oxidative stress and its function is impaired in COPD. Increasing NRF2 activity may therefore be a viable therapeutic option for COPD treatment. We show that down regulation of KEAP1, a NRF2 inhibitor, protects primary human lung epithelial cells from cigarette-smoke-extract (CSE) induced cell death in an established in vitro model of radical stress. To identify new potential drug targets with a similar effect, we performed a siRNA screen of the ‘druggable’ genome using a NRF2 transcriptional reporter cell line. This screen identified multiple genes that when down regulated increased NRF2 transcriptional activity and provided a survival benefit in the in vitro model. Our results suggest that inhibiting components of the ubiquitin-proteasome system will have the strongest effects on NRF2 transcriptional activity by increasing NRF2 levels. We also find that down regulation of the small GTPase Rab28 or the Estrogen Receptor ESRRA provide a survival benefit. Rab28 knockdown increased NRF2 protein levels, indicating that Rab28 may regulate NRF2 proteolysis. Conversely ESRRA down regulation increased NRF2 transcriptional activity without affecting NRF2 levels, suggesting a proteasome-independent mechanism.
Collapse
Affiliation(s)
- Frances-Rose Schumacher
- MRC Protein Phosphorylation and Ubiquitylation Unit, The Sir James Black Center, University of Dundee, Dow Street, Dundee, DD1 5EH, United Kingdom
| | | | | | | | - Carina Ittrich
- Boehringer Ingelheim Pharma GmbH & Co. KG, Research Germany, 8400, Biberach a. d. Riss, Germany
| | - Stefan Kreideweiss
- Boehringer Ingelheim Pharma GmbH & Co. KG, Research Germany, 8400, Biberach a. d. Riss, Germany
| | - Thimo Kurz
- MRC Protein Phosphorylation and Ubiquitylation Unit, The Sir James Black Center, University of Dundee, Dow Street, Dundee, DD1 5EH, United Kingdom
- * E-mail:
| | - Jörg F. Rippmann
- Boehringer Ingelheim Pharma GmbH & Co. KG, Research Germany, 8400, Biberach a. d. Riss, Germany
| |
Collapse
|
74
|
Hubbs AF, Fluharty KL, Edwards RJ, Barnabei JL, Grantham JT, Palmer SM, Kelly F, Sargent LM, Reynolds SH, Mercer RR, Goravanahally MP, Kashon ML, Honaker JC, Jackson MC, Cumpston AM, Goldsmith WT, McKinney W, Fedan JS, Battelli LA, Munro T, Bucklew-Moyers W, McKinstry K, Schwegler-Berry D, Friend S, Knepp AK, Smith SL, Sriram K. Accumulation of Ubiquitin and Sequestosome-1 Implicate Protein Damage in Diacetyl-Induced Cytotoxicity. THE AMERICAN JOURNAL OF PATHOLOGY 2016; 186:2887-2908. [PMID: 27643531 PMCID: PMC5222965 DOI: 10.1016/j.ajpath.2016.07.018] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/23/2015] [Revised: 07/09/2016] [Accepted: 07/21/2016] [Indexed: 01/12/2023]
Abstract
Inhaled diacetyl vapors are associated with flavorings-related lung disease, a potentially fatal airway disease. The reactive α-dicarbonyl group in diacetyl causes protein damage in vitro. Dicarbonyl/l-xylulose reductase (DCXR) metabolizes diacetyl into acetoin, which lacks this α-dicarbonyl group. To investigate the hypothesis that flavorings-related lung disease is caused by in vivo protein damage, we correlated diacetyl-induced airway damage in mice with immunofluorescence for markers of protein turnover and autophagy. Western immunoblots identified shifts in ubiquitin pools. Diacetyl inhalation caused dose-dependent increases in bronchial epithelial cells with puncta of both total ubiquitin and K63-ubiquitin, central mediators of protein turnover. This response was greater in Dcxr-knockout mice than in wild-type controls inhaling 200 ppm diacetyl, further implicating the α-dicarbonyl group in protein damage. Western immunoblots demonstrated decreased free ubiquitin in airway-enriched fractions. Transmission electron microscopy and colocalization of ubiquitin-positive puncta with lysosomal-associated membrane proteins 1 and 2 and with the multifunctional scaffolding protein sequestosome-1 (SQSTM1/p62) confirmed autophagy. Surprisingly, immunoreactive SQSTM1 also accumulated in the olfactory bulb of the brain. Olfactory bulb SQSTM1 often congregated in activated microglial cells that also contained olfactory marker protein, indicating neuronophagia within the olfactory bulb. This suggests the possibility that SQSTM1 or damaged proteins may be transported from the nose to the brain. Together, these findings strongly implicate widespread protein damage in the etiology of flavorings-related lung disease.
Collapse
Affiliation(s)
- Ann F Hubbs
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Centers for Disease Control and Prevention, Morgantown, West Virginia.
| | - Kara L Fluharty
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Centers for Disease Control and Prevention, Morgantown, West Virginia
| | - Rebekah J Edwards
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Centers for Disease Control and Prevention, Morgantown, West Virginia; Department of Forensic and Investigative Science, West Virginia University, Morgantown, West Virginia
| | - Jamie L Barnabei
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Centers for Disease Control and Prevention, Morgantown, West Virginia; College of Veterinary Medicine, University of Georgia, Athens, Georgia
| | - John T Grantham
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Centers for Disease Control and Prevention, Morgantown, West Virginia; School of Medicine, West Virginia University, Morgantown, West Virginia
| | - Scott M Palmer
- Duke University School of Medicine, Durham, North Carolina
| | - Francine Kelly
- Duke University School of Medicine, Durham, North Carolina
| | - Linda M Sargent
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Centers for Disease Control and Prevention, Morgantown, West Virginia
| | - Steven H Reynolds
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Centers for Disease Control and Prevention, Morgantown, West Virginia
| | - Robert R Mercer
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Centers for Disease Control and Prevention, Morgantown, West Virginia
| | - Madhusudan P Goravanahally
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Centers for Disease Control and Prevention, Morgantown, West Virginia; Centers for Neuroscience, West Virginia University, Morgantown, West Virginia
| | - Michael L Kashon
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Centers for Disease Control and Prevention, Morgantown, West Virginia
| | - John C Honaker
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Centers for Disease Control and Prevention, Morgantown, West Virginia
| | - Mark C Jackson
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Centers for Disease Control and Prevention, Morgantown, West Virginia
| | - Amy M Cumpston
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Centers for Disease Control and Prevention, Morgantown, West Virginia
| | - William T Goldsmith
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Centers for Disease Control and Prevention, Morgantown, West Virginia
| | - Walter McKinney
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Centers for Disease Control and Prevention, Morgantown, West Virginia
| | - Jeffrey S Fedan
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Centers for Disease Control and Prevention, Morgantown, West Virginia
| | - Lori A Battelli
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Centers for Disease Control and Prevention, Morgantown, West Virginia
| | - Tiffany Munro
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Centers for Disease Control and Prevention, Morgantown, West Virginia
| | - Winnie Bucklew-Moyers
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Centers for Disease Control and Prevention, Morgantown, West Virginia
| | - Kimberly McKinstry
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Centers for Disease Control and Prevention, Morgantown, West Virginia
| | - Diane Schwegler-Berry
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Centers for Disease Control and Prevention, Morgantown, West Virginia
| | - Sherri Friend
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Centers for Disease Control and Prevention, Morgantown, West Virginia
| | - Alycia K Knepp
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Centers for Disease Control and Prevention, Morgantown, West Virginia; Department of Forensic and Investigative Science, West Virginia University, Morgantown, West Virginia
| | - Samantha L Smith
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Centers for Disease Control and Prevention, Morgantown, West Virginia; Department of Forensic and Investigative Science, West Virginia University, Morgantown, West Virginia
| | - Krishnan Sriram
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Centers for Disease Control and Prevention, Morgantown, West Virginia
| |
Collapse
|
75
|
Vij N. Nano-based rescue of dysfunctional autophagy in chronic obstructive lung diseases. Expert Opin Drug Deliv 2016; 14:483-489. [PMID: 27561233 DOI: 10.1080/17425247.2016.1223040] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
INTRODUCTION ΔF508-CFTR (cystic fibrosis transmembrane conductance regulator) is a common CF-mutation that is known to induce oxidative-inflammatory stress through activation of reactive oxygen species (ROS), which induces autophagy-impairment resulting in accumulation of CFTR in aggresome-bodies. Cysteamine, the reduced form of cystamine, is a FDA-approved drug that has anti-oxidant, anti-bacterial, and mucolytic properties. This drug has been shown in a recent clinical trial to decrease lung inflammation and improve lung function in CF patients by potentially restoring autophagy and allowing CFTR to be trafficked to the cell membrane. Areas covered: The delivery of cysteamine to airway epithelia of chronic subjects prerequisite the need for a delivery system to allow rescue of dysfunctional autophagy. Expert opinion: We anticipate based on our ongoing studies that PLGA-PEG- or Dendrimer-mediated cysteamine delivery could allow sustained airway delivery over standard cysteamine tablets or delay release capsules that are currently used for systemic treatment. In addition, proposed nano-based autophagy induction strategy can also allow rescue of cigarette smoke (CS) induced acquired-CFTR dysfunction seen in chronic obstructive pulmonary disease (COPD)-emphysema subjects. The CS induced acquired-CFTR dysfunction involves CFTR-accumulation in aggresome-bodies that can be rescued by an autophagy-inducing antioxidant drug, cysteamine. Moreover, chronic CS-exposure generates ROS that induces overall protein-misfolding and aggregation of ubiquitinated-proteins as aggresome-bodies via autophagy-impairment that can be also be resolved by treatment with autophagy-inducing antioxidant drug, cysteamine.
Collapse
Affiliation(s)
- Neeraj Vij
- a College of Medicine , Central Michigan University , Mount Pleasant , MI , USA.,b Department of Pediatric Respiratory Sciences , The Johns Hopkins School of Medicine , Baltimore , MD , USA
| |
Collapse
|
76
|
Bodas M, Van Westphal C, Carpenter-Thompson R, K Mohanty D, Vij N. Nicotine exposure induces bronchial epithelial cell apoptosis and senescence via ROS mediated autophagy-impairment. Free Radic Biol Med 2016; 97:441-453. [PMID: 27394171 DOI: 10.1016/j.freeradbiomed.2016.06.017] [Citation(s) in RCA: 90] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Revised: 05/30/2016] [Accepted: 06/17/2016] [Indexed: 12/15/2022]
Abstract
Waterpipe smoking and e-cigarette vaping, the non-combustible sources of inhaled nicotine exposure are increasingly becoming popular and marketed as safer alternative to cigarette smoking. Hence, this study was designed to investigate the impact of inhaled nicotine exposure on disease causing COPD-emphysema mechanisms. For in vitro studies, human bronchial epithelial cells (Beas2b) were treated with waterpipe smoke extract (WPSE, 5%), nicotine (5mM), and/or cysteamine (250μM, an autophagy inducer and anti-oxidant drug), for 6hrs. We observed significantly (p<0.05) increased ubiquitinated protein-accumulation in the insoluble protein fractions of Beas2b cells treated with WPSE or nicotine that could be rescued by cysteamine treatment, suggesting aggresome-formation and autophagy-impairment. Moreover, our data also demonstrate that both WPSE and nicotine exposure significantly (p<0.05) elevates Ub-LC3β co-localization to aggresome-bodies while inducing Ub-p62 co-expression/accumulation, verifying autophagy-impairment. We also found that WPSE and nicotine exposure impacts Beas2b cell viability by significantly (p<0.05) inducing cellular apoptosis/senescence via ROS-activation, as it could be controlled by cysteamine, which is known to have an anti-oxidant property. For murine studies, C57BL/6 mice were administered with inhaled nicotine (intranasal, 500μg/mouse/day for 5 days), as an experimental model of non-combustible nicotine exposure. The inhaled nicotine exposure mediated oxidative-stress induces autophagy-impairment in the murine lungs as seen by significant (p<0.05, n=4) increase in the expression levels of nitrotyrosine protein-adduct (oxidative-stress marker, soluble-fraction) and Ub/p62/VCP (impaired-autophagy marker, insoluble-fraction). Overall, our data shows that nicotine, a common component of WPS, e-cigarette vapor and cigarette smoke, induces bronchial epithelial cell apoptosis and senescence via ROS mediated autophagy-impairment as a potential mechanism for COPD-emphysema pathogenesis.
Collapse
Affiliation(s)
- Manish Bodas
- College of Medicine, Central Michigan University, Mt Pleasant, MI, United States
| | - Colin Van Westphal
- College of Medicine, Central Michigan University, Mt Pleasant, MI, United States
| | | | - Dillip K Mohanty
- Department of Chemistry and Biochemistry, Central Michigan University, Mt Pleasant, MI, United States
| | - Neeraj Vij
- College of Medicine, Central Michigan University, Mt Pleasant, MI, United States; Department of Pediatrics and Pulmonary Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD, United States.
| |
Collapse
|
77
|
Fratta Pasini AM, Ferrari M, Stranieri C, Vallerio P, Mozzini C, Garbin U, Zambon G, Cominacini L. Nrf2 expression is increased in peripheral blood mononuclear cells derived from mild-moderate ex-smoker COPD patients with persistent oxidative stress. Int J Chron Obstruct Pulmon Dis 2016; 11:1733-43. [PMID: 27555763 PMCID: PMC4968670 DOI: 10.2147/copd.s102218] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Inadequacy of antioxidant nuclear factor-E2-related factor 2 (Nrf2) and endoplasmic reticulum stress-mediated unfolded protein response has been implicated in severe chronic obstructive pulmonary disease (COPD) and cigarette smoking-induced emphysema. As evidence suggests that the ability to upregulate Nrf2 expression may influence the progression of COPD and no data exist up to now in ex-smokers with mild-moderate COPD, this study was first aimed to evaluate Nrf2 and unfolded protein response expression in peripheral blood mononuclear cells (PBMC) of mild-moderate ex-smokers with COPD compared to smoking habit-matched non-COPD subjects. Then, we tested whether oxidative stress persists after cigarette smoking cessation and whether the concentrations of oxidized phospholipids (oxidation products of the phospholipid 1-palmitoyl-2-arachidonyl-sn-glycero-3-phosphorylcholine [oxPAPC]) in the PBMC of the same subjects may have a causative role in determining the upregulation of Nrf2. The expression (mRNA and protein) of Nrf2 and of its related gene heme oxygenase-1 was significantly increased in COPD group without differences in the unfolded protein response. Plasma malondialdehyde, the circulating marker of oxidative stress, and oxPAPC in PBMC were significantly higher in COPD than in non-COPD subjects. The fact that the expression of p47phox, a subunit of NADPH oxidase, was increased in PBMC of COPD patients and that it was directly correlated with oxPAPC may indicate that oxPAPC may be one of the determinants of oxidative stress-induced Nrf2 upregulation. Finally, we also demonstrated that lung function inversely correlated with plasma malondialdehyde and with Nrf2 and heme oxygenase-1 mRNA expression in all subjects. Our results indicate that mild-moderate ex-smokers with COPD may be able to counteract oxidative stress by increasing the expression of Nrf2/antioxidant-response elements. Because Nrf2 failure significantly contributes to the development of COPD, our findings suggest that the possibility to prevent Nrf2 reduction may open a new scenario in helping to prevent the oxidative stress-associated lung function decline.
Collapse
Affiliation(s)
| | - Marcello Ferrari
- Department of Medicine, Unit of Respiratory Diseases, University of Verona, Verona, Italy
| | | | | | | | - Ulisse Garbin
- Department of Medicine, Section of Internal Medicine
| | | | | |
Collapse
|
78
|
Walworth K, Bodas M, Campbell RJ, Swanson D, Sharma A, Vij N. Dendrimer-Based Selective Proteostasis-Inhibition Strategy to Control NSCLC Growth and Progression. PLoS One 2016; 11:e0158507. [PMID: 27434122 PMCID: PMC4951140 DOI: 10.1371/journal.pone.0158507] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Accepted: 06/16/2016] [Indexed: 02/06/2023] Open
Abstract
Elevated valosin containing protein (VCP/p97) levels promote the progression of non-small cell lung carcinoma (NSCLC). Although many VCP inhibitors are available, most of these therapeutic compounds have low specificity for targeted tumor cell delivery. Hence, the primary aim of this study was to evaluate the in vitro efficacy of dendrimer-encapsulated potent VCP-inhibitor drug in controlling non-small cell lung carcinoma (NSCLC) progression. The VCP inhibitor(s) (either in their pure form or encapsulated in generation-4 PAMAM-dendrimer with hydroxyl surface) were tested for their in vitro efficacy in modulating H1299 (NSCLC cells) proliferation, migration, invasion, apoptosis and cell cycle progression. Our results show that VCP inhibition by DBeQ was significantly more potent than NMS-873 as evident by decreased cell proliferation (p<0.0001, MTT-assay) and migration (p<0.05; scratch-assay), and increased apoptosis (p<0.05; caspase-3/7-assay) as compared to untreated control cells. Next, we found that dendrimer-encapsulated DBeQ (DDNDBeQ) treatment increased ubiquitinated-protein accumulation in soluble protein-fraction (immunoblotting) of H1299 cells as compared to DDN-control, implying the effectiveness of DBeQ in proteostasis-inhibition. We verified by immunostaining that DDNDBeQ treatment increases accumulation of ubiquitinated-proteins that co-localizes with an ER-marker, KDEL. We observed that proteostasis-inhibition with DDNDBeQ, significantly decreased cell migration rate (scratch-assay and transwell-invasion) as compared to the control-DDN treatment (p<0.05). Moreover, DDNDBeQ treatment showed a significant decrease in cell proliferation (p<0.01, MTT-assay) and increased caspase-3/7 mediated apoptotic cell death (p<0.05) as compared to DDN-control. This was further verified by cell cycle analysis (propidium-iodide-staining) that demonstrated significant cell cycle arrest in the G2/M-phase (p<0.001) by DDNDBeQ treatment as compared to control-DDN. Moreover, we confirmed by clonogenic-assay that DDNDBeQ treatment significantly (p<0.001) inhibits H1299 colony-formation as compared to control/DDN. Overall, encapsulation of potent VCP-inhibitor DBeQ into a dendrimer allows selective VCP-mediated proteostasis-inhibition for controlling NSCLC-tumor growth and progression to allow tumor-targeted sustained drug delivery.
Collapse
Affiliation(s)
- Kyla Walworth
- College of Medicine, Central Michigan University, Mount Pleasant, Michigan, United States of America
| | - Manish Bodas
- College of Medicine, Central Michigan University, Mount Pleasant, Michigan, United States of America
| | - Ryan John Campbell
- College of Medicine, Central Michigan University, Mount Pleasant, Michigan, United States of America
| | - Doug Swanson
- Department of Chemistry and Biochemistry, Central Michigan University, Mount Pleasant, Michigan, United States of America
| | - Ajit Sharma
- Department of Chemistry and Biochemistry, Central Michigan University, Mount Pleasant, Michigan, United States of America
| | - Neeraj Vij
- College of Medicine, Central Michigan University, Mount Pleasant, Michigan, United States of America
- Department of Pediatric Respiratory Sciences, The Johns Hopkins School of Medicine, Baltimore, Maryland, United States of America
- * E-mail: ;
| |
Collapse
|
79
|
Vij N, Chandramani-Shivalingappa P, Van Westphal C, Hole R, Bodas M. Cigarette smoke-induced autophagy impairment accelerates lung aging, COPD-emphysema exacerbations and pathogenesis. Am J Physiol Cell Physiol 2016; 314:C73-C87. [PMID: 27413169 DOI: 10.1152/ajpcell.00110.2016] [Citation(s) in RCA: 201] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Cigarette-smoke (CS) exposure and aging are the leading causes of chronic obstructive pulmonary disease (COPD)-emphysema development, although the molecular mechanism that mediates disease pathogenesis remains poorly understood. Our objective was to investigate the impact of CS exposure and aging on autophagy and the pathophysiological changes associated with lung aging (senescence) and emphysema progression. Beas2b cells, C57BL/6 mice, and human (GOLD 0-IV) lung tissues were used to determine the central mechanism involved in CS/age-related COPD-emphysema pathogenesis. Beas2b cells and murine lungs exposed to cigarette smoke extract (CSE)/CS showed a significant ( P < 0.05) accumulation of poly-ubiquitinated proteins and impaired autophagy marker, p62, in aggresome bodies. Moreover, treatment with the autophagy-inducing antioxidant drug cysteamine significantly ( P < 0.001) decreased CSE/CS-induced aggresome bodies. We also found a significant ( P < 0.001) increase in levels of aggresome bodies in the lungs of smokers and COPD subjects in comparison to nonsmoker controls. Furthermore, the presence and levels of aggresome bodies statistically correlated with severity of emphysema and alveolar senescence. In addition to CS exposure, lungs from old mice also showed accumulation of aggresome bodies, suggesting this as a common mechanism to initiate cellular senescence and emphysema. Additionally, Beas2b cells and murine lungs exposed to CSE/CS showed cellular apoptosis and senescence, which were both controlled by cysteamine treatment. In parallel, we evaluated the impact of CS on pulmonary exacerbation, using mice exposed to CS and/or infected with Pseudomonas aeruginosa ( Pa), and confirmed cysteamine's potential as an autophagy-inducing antibacterial drug, based on its ability to control CS-induced pulmonary exacerbation ( Pa-bacterial counts) and resulting inflammation. CS induced autophagy impairment accelerates lung aging and COPD-emphysema exacerbations and pathogenesis.
Collapse
Affiliation(s)
- Neeraj Vij
- College of Medicine, Central Michigan University, Mt. Pleasant, Michigan.,Department of Pediatric Respiratory Science, The Johns Hopkins University School of Medicine , Baltimore, Maryland
| | | | - Colin Van Westphal
- College of Medicine, Central Michigan University, Mt. Pleasant, Michigan
| | - Rachel Hole
- College of Medicine, Central Michigan University, Mt. Pleasant, Michigan
| | - Manish Bodas
- College of Medicine, Central Michigan University, Mt. Pleasant, Michigan
| |
Collapse
|
80
|
Fu Q, Jiang Y, Zhang D, Liu X, Guo J, Zhao J. Valosin-containing protein (VCP) promotes the growth, invasion, and metastasis of colorectal cancer through activation of STAT3 signaling. Mol Cell Biochem 2016; 418:189-98. [PMID: 27344168 PMCID: PMC4927615 DOI: 10.1007/s11010-016-2746-6] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Accepted: 06/17/2016] [Indexed: 01/14/2023]
Abstract
Valosin-containing protein (VCP) was previously shown to exhibit high expression in colorectal cancer (CRC) tissues as compared with that in normal tissues; however, the role of VCP in human CRC cells has remained to be elucidated. Two colorectal cancer cell lines HCT116 and RKO were used in the experiment. We introduced lentiviral constructs expressing VCP to infect RKO cells and lenti-shRNA targeting VCP into HCT116 cells, respectively. Cell proliferation, invasion, apoptosis, and cell cycle arrest were subsequently examined by MTT assay, transwell chamber assay, flow cytometry, and western blot analysis, respectively. Furthermore, a subcutaneous tumor mouse model and lung metastasis model was used to investigate the effects of VCP on the growth and metastasis of CRC cells in vivo. VCP knockdown was shown to inhibit cell proliferation, chemoresistance and invasion, and induce apoptosis in the HCT116 CRC cells, whereas VCP over-expression suppressed apoptosis and chemoresponse, promoted proliferation and invasion of the RKO CRC cells. In addition, in the subcutaneous tumor and lung metastasis mouse model, VCP knockdown in HCT116 cells suppressed carcinogenesis and metastasis in vivo. The findings of the present study indicated that VCP is very important for the proliferation and metastasis of CRC; therefore, targeting VCP and its downstream targets may represent novel therapies for the treatment of CRC.
Collapse
Affiliation(s)
- Qianfeng Fu
- Department of Oncology, The First Affiliated Hospital of Harbin Medical University, No. 23 Youzheng Street, Harbin, 150001, Heilongjiang, China
| | - Yuling Jiang
- Department of Clinical Laborotary, The First Affiliated Hospital of Harbin Medical University, No. 23 Youzheng Street, Harbin, 150001, Heilongjiang, China
| | - Daxin Zhang
- Department of Oncology, The First Affiliated Hospital of Harbin Medical University, No. 23 Youzheng Street, Harbin, 150001, Heilongjiang, China
| | - Xiuli Liu
- Department of Oncology, The First Affiliated Hospital of Harbin Medical University, No. 23 Youzheng Street, Harbin, 150001, Heilongjiang, China
| | - Junfeng Guo
- Department of Oncology, The First Affiliated Hospital of Harbin Medical University, No. 23 Youzheng Street, Harbin, 150001, Heilongjiang, China
| | - Jinlong Zhao
- Department of Oncology, The First Affiliated Hospital of Harbin Medical University, No. 23 Youzheng Street, Harbin, 150001, Heilongjiang, China.
| |
Collapse
|
81
|
Emerging role of immunoproteasomes in pathophysiology. Immunol Cell Biol 2016; 94:812-820. [PMID: 27192937 DOI: 10.1038/icb.2016.50] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Revised: 05/15/2016] [Accepted: 05/16/2016] [Indexed: 11/08/2022]
Abstract
The immunoproteasome is a proteasome variant that is found only in jawed vertebrates. It is responsible for degrading intracellular proteins to generate a major source of peptides with substantial major histocompatibility complex I binding affinity. The immunoproteasome also has roles in T-cell survival, differentiation and proliferation in various pathological conditions. In humans, any alteration in the expression, assembly or function of the immunoproteasome can lead to cancer, autoimmune disorders or inflammatory diseases. Although the roles of the immunoproteasome in cancer and neurodegenerative disorders have been extensively studied, its significance in other disease conditions has only recently become known. Therefore, there is renewed interest in the development of drugs, vaccines and biomarkers that target the immunoproteasome. The current review highlights the involvement of this complex in disease pathology in addition to the advances made in immunoproteasome research.
Collapse
|
82
|
Kammerl IE, Dann A, Mossina A, Brech D, Lukas C, Vosyka O, Nathan P, Conlon TM, Wagner DE, Overkleeft HS, Prasse A, Rosas IO, Straub T, Krauss-Etschmann S, Königshoff M, Preissler G, Winter H, Lindner M, Hatz R, Behr J, Heinzelmann K, Yildirim AÖ, Noessner E, Eickelberg O, Meiners S. Impairment of Immunoproteasome Function by Cigarette Smoke and in Chronic Obstructive Pulmonary Disease. Am J Respir Crit Care Med 2016; 193:1230-41. [PMID: 26756824 DOI: 10.1164/rccm.201506-1122oc] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
RATIONALE Patients with chronic obstructive pulmonary disease (COPD) and in particular smokers are more susceptible to respiratory infections contributing to acute exacerbations of disease. The immunoproteasome is a specialized type of proteasome destined to improve major histocompatibility complex (MHC) class I-mediated antigen presentation for the resolution of intracellular infections. OBJECTIVES To characterize immunoproteasome function in COPD and its regulation by cigarette smoke. METHODS Immunoproteasome expression and activity were determined in bronchoalveolar lavage (BAL) and lungs of human donors and patients with COPD or idiopathic pulmonary fibrosis (IPF), as well as in cigarette smoke-exposed mice. Smoke-mediated alterations of immunoproteasome activity and MHC I surface expression were analyzed in human blood-derived macrophages. Immunoproteasome-specific MHC I antigen presentation was evaluated in spleen and lung immune cells that had been smoke-exposed in vitro or in vivo. MEASUREMENTS AND MAIN RESULTS Immunoproteasome and MHC I mRNA expression was reduced in BAL cells of patients with COPD and in isolated alveolar macrophages of patients with COPD or IPF. Exposure of immune cells to cigarette smoke extract in vitro reduced immunoproteasome activity and impaired immunoproteasome-specific MHC I antigen presentation. In vivo, acute cigarette smoke exposure dynamically regulated immunoproteasome function and MHC I antigen presentation in mouse BAL cells. End-stage COPD lungs showed markedly impaired immunoproteasome activities. CONCLUSIONS We here show that the activity of the immunoproteasome is impaired by cigarette smoke resulting in reduced MHC I antigen presentation. Regulation of immunoproteasome function by cigarette smoke may thus alter adaptive immune responses and add to prolonged infections and exacerbations in COPD and IPF.
Collapse
Affiliation(s)
- Ilona E Kammerl
- 1 Comprehensive Pneumology Center, University Hospital, Ludwig-Maximilians University, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Angela Dann
- 1 Comprehensive Pneumology Center, University Hospital, Ludwig-Maximilians University, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Alessandra Mossina
- 1 Comprehensive Pneumology Center, University Hospital, Ludwig-Maximilians University, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Dorothee Brech
- 2 Institute of Molecular Immunology, Helmholtz Zentrum München, Munich, Germany
| | - Christina Lukas
- 1 Comprehensive Pneumology Center, University Hospital, Ludwig-Maximilians University, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Oliver Vosyka
- 1 Comprehensive Pneumology Center, University Hospital, Ludwig-Maximilians University, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Petra Nathan
- 1 Comprehensive Pneumology Center, University Hospital, Ludwig-Maximilians University, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Thomas M Conlon
- 3 Comprehensive Pneumology Center, Institute of Lung Biology and Disease, Helmholtz Zentrum München, Member of the DZL, Neuherberg, Germany
| | - Darcy E Wagner
- 2 Institute of Molecular Immunology, Helmholtz Zentrum München, Munich, Germany
| | - Hermen S Overkleeft
- 4 Department of Bio-organic Synthesis, Leiden University, Leiden, the Netherlands
| | - Antje Prasse
- 5 Department of Pneumology, Hannover Medical School, Hannover, Germany
| | - Ivan O Rosas
- 6 Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Tobias Straub
- 7 Biomedical Center, Bioinformatics Unit, Ludwig-Maximilians University, Munich, Germany
| | - Susanne Krauss-Etschmann
- 8 Division of Experimental Asthma Research, Research Center Borstel, Airway Research Center North, Member of the DZL, Borstel, Germany
- 9 Institute of Experimental Medicine, Christian-Albrechts-Universität zu Kiel, Germany
| | - Melanie Königshoff
- 1 Comprehensive Pneumology Center, University Hospital, Ludwig-Maximilians University, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Gerhard Preissler
- 10 Thoraxchirurgisches Zentrum, Klinik für Allgemeine-, Viszeral-, Transplantations-, Gefäß- und Thoraxchirurgie, Klinikum Großhadern, Ludwig-Maximilians-Universität, Member of the DZL, Munich, Germany
| | - Hauke Winter
- 10 Thoraxchirurgisches Zentrum, Klinik für Allgemeine-, Viszeral-, Transplantations-, Gefäß- und Thoraxchirurgie, Klinikum Großhadern, Ludwig-Maximilians-Universität, Member of the DZL, Munich, Germany
| | - Michael Lindner
- 11 Asklepios Fachkliniken München-Gauting, Gauting, Germany; and
| | - Rudolf Hatz
- 10 Thoraxchirurgisches Zentrum, Klinik für Allgemeine-, Viszeral-, Transplantations-, Gefäß- und Thoraxchirurgie, Klinikum Großhadern, Ludwig-Maximilians-Universität, Member of the DZL, Munich, Germany
- 11 Asklepios Fachkliniken München-Gauting, Gauting, Germany; and
| | - Jürgen Behr
- 1 Comprehensive Pneumology Center, University Hospital, Ludwig-Maximilians University, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
- 11 Asklepios Fachkliniken München-Gauting, Gauting, Germany; and
- 12 Medizinische Klinik und Poliklinik V, Klinikum der Ludwig-Maximilians-Universität, Member of the DZL, Munich, Germany
| | - Katharina Heinzelmann
- 1 Comprehensive Pneumology Center, University Hospital, Ludwig-Maximilians University, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Ali Ö Yildirim
- 3 Comprehensive Pneumology Center, Institute of Lung Biology and Disease, Helmholtz Zentrum München, Member of the DZL, Neuherberg, Germany
| | - Elfriede Noessner
- 2 Institute of Molecular Immunology, Helmholtz Zentrum München, Munich, Germany
| | - Oliver Eickelberg
- 1 Comprehensive Pneumology Center, University Hospital, Ludwig-Maximilians University, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Silke Meiners
- 1 Comprehensive Pneumology Center, University Hospital, Ludwig-Maximilians University, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
| |
Collapse
|
83
|
Leuenberger C, Schuoler C, Bye H, Mignan C, Rechsteiner T, Hillinger S, Opitz I, Marsland B, Faiz A, Hiemstra PS, Timens W, Camici GG, Kohler M, Huber LC, Brock M. MicroRNA-223 controls the expression of histone deacetylase 2: a novel axis in COPD. J Mol Med (Berl) 2016; 94:725-34. [PMID: 26864305 DOI: 10.1007/s00109-016-1388-1] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2015] [Revised: 12/07/2015] [Accepted: 01/08/2016] [Indexed: 01/14/2023]
Abstract
UNLABELLED Reduced activity of histone deacetylase 2 (HDAC2) has been described in patients with chronic obstructive pulmonary disease (COPD), but the mechanisms resulting in decreased expression of this important epigenetic modifier remain unknown. Here, we employed several in vitro experiments to address the role of microRNAs (miRNAs) on the regulation of HDAC2 in endothelial cells. Manipulation of miRNA levels in human pulmonary artery endothelial cells (HPAEC) was achieved by using electroporation with anti-miRNAs and miRNA mimics. Target prediction software identified miR-223 as a potential repressor of HDAC2. In subsequent stimulation experiments using inflammatory cytokines known to be increased in patients with COPD, miR-223 was found to be significantly induced. Functional analysis demonstrated that overexpression of miR-223 decreased HDAC2 expression and activity in HPAEC. Conversely, HDAC2 expression and activity was preserved in anti-miR-223-treated cells. Direct miRNA-target interaction was confirmed by reporter gene assay. In a next step, reduced expression of HDAC2 was found to increase the levels of the chemokine fractalkine (CX3CL1). In vivo studies confirmed elevated expression levels of miR-223 in mice exposed to cigarette smoke and in emphysematous lung tissue from LPS-treated mice. Moreover, a significant inverse correlation of miR-223 and HDAC2 expression was found in two independent cohorts of COPD patients. These data emphasize that miR-223, the most prevalent miRNA in COPD, controls expression and activity of HDAC2 in pulmonary cells, which, in turn, might alter the expression profile of chemokines. This pathway provides a novel pathogenic link between dysregulated miRNA expression and epigenetic activity in COPD. KEY MESSAGES Histone deacetylase 2 is directly targeted by miR-223. Levels of miR-223 are induced by interleukin-1β and tumor necrosis factor-α. miR-223 controls the expression of fractalkine by targeting histone deacetylase 2. miR-223 levels are increased in COPD mouse models. miR-223 levels inversely correlate with HDAC2 expression in COPD patients.
Collapse
Affiliation(s)
- Caroline Leuenberger
- Division of Pulmonology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Claudio Schuoler
- Division of Pulmonology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
- Institute of Veterinary Physiology, University of Zurich and Zurich Center for Integrative Human Physiology (ZIHP), Zurich, Switzerland
| | - Hannah Bye
- Division of Pulmonology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Célia Mignan
- Division of Pulmonology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Thomas Rechsteiner
- Division of Pulmonology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Sven Hillinger
- Division of Thoracic Surgery, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Isabelle Opitz
- Division of Thoracic Surgery, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Benjamin Marsland
- Faculty of Biology and Medicine, Service de Pneumologie, University of Lausanne, Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, Switzerland
| | - Alen Faiz
- Department of Pulmonary Diseases, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Pieter S Hiemstra
- Department of Pulmonary Diseases, Leiden University Medical Center, Leiden, The Netherlands
| | - Wim Timens
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Giovanni G Camici
- Center for Molecular Cardiology, Division of Cardiology, University Hospital of Zurich, University of Zurich, Zurich, Switzerland
| | - Malcolm Kohler
- Division of Pulmonology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Lars C Huber
- Division of Pulmonology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Matthias Brock
- Division of Pulmonology, University Hospital Zurich, University of Zurich, Zurich, Switzerland.
| |
Collapse
|
84
|
Panday A, Inda ME, Bagam P, Sahoo MK, Osorio D, Batra S. Transcription Factor NF-κB: An Update on Intervention Strategies. Arch Immunol Ther Exp (Warsz) 2016; 64:463-483. [PMID: 27236331 DOI: 10.1007/s00005-016-0405-y] [Citation(s) in RCA: 87] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Accepted: 04/14/2016] [Indexed: 12/25/2022]
Abstract
The nuclear factor (NF)-κB family of transcription factors are ubiquitous and pleiotropic molecules that regulate the expression of more than 150 genes involved in a broad range of processes including inflammation, immunity, cell proliferation, differentiation, and survival. The chronic activation or dysregulation of NF-κB signaling is the central cause of pathogenesis in many disease conditions and, therefore, NF-κB is a major focus of therapeutic intervention. Because of this, understanding the relationship between NF-κB and the induction of various downstream signaling molecules is imperative. In this review, we provide an updated synopsis of the role of NF-κB in DNA repair and in various ailments including cardiovascular diseases, HIV infection, asthma, herpes simplex virus infection, chronic obstructive pulmonary disease, and cancer. Furthermore, we also discuss the specific targets for selective inhibitors and future therapeutic strategies.
Collapse
Affiliation(s)
- Arvind Panday
- Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, 70803, USA.,Department of Biological Sciences, Louisiana State University, Baton Rouge, LA, 70803, USA
| | - Maria Eugenia Inda
- Departamento de Microbiología, CONICET, Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional Rosario, Suipacha 531, Santa Fe, Argentina
| | - Prathyusha Bagam
- Laboratory of Pulmonary Immunotoxicology, Environmental Toxicology PhD Program, 207 Health Research Center, Southern University and A&M College, Baton Rouge, LA, 70813, USA
| | - Malaya K Sahoo
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, 94304, USA
| | - Diana Osorio
- Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, 70803, USA
| | - Sanjay Batra
- Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, 70803, USA. .,Laboratory of Pulmonary Immunotoxicology, Environmental Toxicology PhD Program, 207 Health Research Center, Southern University and A&M College, Baton Rouge, LA, 70813, USA.
| |
Collapse
|
85
|
Zeng H, Shi Z, Kong X, Chen Y, Zhang H, Peng H, Luo H, Chen P. Involvement of B-cell CLL/lymphoma 2 promoter methylation in cigarette smoke extract-induced emphysema. Exp Biol Med (Maywood) 2016; 241:808-16. [PMID: 26924842 DOI: 10.1177/1535370216635759] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Accepted: 02/02/2016] [Indexed: 12/26/2022] Open
Abstract
Abnormal apoptotic events play an important role in the pathogenesis of emphysema. The B-cell CLL/lymphoma 2 (Bcl-2) family proteins are essential and critical regulators of apoptosis. We determined whether the anti-apoptotic Bcl-2 play a role in the cigarette smoke extract (CSE)-induced emphysema. Furthermore, given the involvement of epigenetics in chronic obstructive pulmonary disease, we hypothesized that the deregulation of Bcl-2 might be caused by gene methylation. The emphysema in BALB/C mice was established by intraperitoneally injection of CSE. 5-aza-2'-deoxycytidine (AZA; a demethylation reagent) and phosphate-buffered saline were also administered intraperitoneally as CSE. TUNEL assay was used to assess apoptotic index of pulmonary cells. The methylation status of CpG dinucleotides within the Bcl-2 promoter was observed in all groups by bisulfite sequencing PCR. Pulmonary expression of Bcl-2, Bax, and cytochrome C were measured after four weeks of treatment. The apoptotic index of pulmonary cells in CSE injection group was much higher than control ((25.88 ± 7.55)% vs (6.28 ± 2.96)%). Compared to control mice, decreased expression of Bcl-2 and high methylation of Bcl-2 promoter was observed in CSE injected mice (0.88 ± 0.08 vs 0.49 ± 0.11, (3.82 ± 1.34)% vs (35.68 ± 5.99)%, P < 0.01).CSE treatment induced lung cell apoptosis and decreased lung function. AZA treatment increased Bcl-2 expression with Bcl-2 promoter demethylation. AZA also alleviated the lung cell apoptosis and function failure caused by CSE treatment. The decreased expression of anti-apoptotic Bcl-2 might account for the increased apoptosis in CSE induced-emphysema. Apparently, epigenetic alternation played a role in this deregulation of Bcl-2 expression, and it might support the involvement of epigenetic events in the pathogenesis of emphysema.
Collapse
Affiliation(s)
- Huihui Zeng
- Department of Respiratory Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Zhihui Shi
- Department of Respiratory Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Xianglong Kong
- Department of Respiratory Medicine, The First Hospital of Changsha, Changsha, Hunan 410011, China
| | - Yan Chen
- Department of Respiratory Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Hongliang Zhang
- Department of Emergency Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Hong Peng
- Department of Respiratory Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Hong Luo
- Department of Respiratory Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Ping Chen
- Department of Respiratory Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| |
Collapse
|
86
|
Shivalingappa PC, Hole R, Westphal CV, Vij N. Airway Exposure to E-Cigarette Vapors Impairs Autophagy and Induces Aggresome Formation. Antioxid Redox Signal 2016; 24:186-204. [PMID: 26377848 PMCID: PMC4744882 DOI: 10.1089/ars.2015.6367] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
AIMS Electronic cigarettes (e-cigarettes) are proposed to be a safer alternative to tobacco cigarettes. Hence, we evaluated if e-cigarette vapors (eCV) impair cellular proteostasis similar to cigarette smoke exposure. RESULTS First, we evaluated the impact of eCV exposure (2.5 or 7.5 mg) on Beas2b cells that showed significant increase in accumulation of total polyubiquitinated proteins (Ub, insoluble fractions) with time-dependent decrease in proteasomal activities from 1 h (p < 0.05), 3 h (p < 0.001) to 6 h (p < 0.001) of eCV exposure compared to room air control. We verified that even minimal eCV exposure (1 h) induces valosin-containing protein (VCP; p < 0.001), sequestosome-1/p62 (aberrant autophagy marker; p < 0.05), and aggresome formation (total poly-Ub-accumulation; p < 0.001) using immunoblotting (IB), fluorescence microscopy, and immunoprecipitation (IP). The inhibition of protein synthesis by 6 h of cycloheximide (50 μg/ml) treatment significantly (p < 0.01) alleviates eCV-induced (1 h) aggresome bodies. We also observed that eCV (1 h)-induced protein aggregation can activate oxidative stress, apoptosis (caspase-3/7), and senescence (p < 0.01) compared to room air controls. We verified using an autophagy inducer carbamazepine (20 μM, 6 h) or cysteamine (250 μM; 6 h, antioxidant) that eCV-induced changes in oxidative stress, poly-ub-accumulation, proteasomal activity, autophagy, apoptosis, and/or senescence could be controlled by autophagy induction. We further confirmed the role of acute eCV exposure on autophagy impairment in murine lungs (C57BL/6 and CD1) by IB (Ub, p62, VCP) and IP (VCP, p62), similar to in-vitro experiments. INNOVATION In this study, we report for the first time that eCV exposure induces proteostasis/autophagy impairment leading to oxidative stress, apoptosis, and senescence that can be ameliorated by an autophagy inducer. CONCLUSION eCV-induced autophagy impairment and aggresome formation suggest their potential role in chronic obstructive pulmonary disease-emphysema pathogenesis. Antioxid. Redox Signal. 00, 000-000.
Collapse
Affiliation(s)
| | - Rachel Hole
- 1 College of Medicine, Central Michigan University , Mt Pleasant, Michigan
| | - Colin Van Westphal
- 1 College of Medicine, Central Michigan University , Mt Pleasant, Michigan
| | - Neeraj Vij
- 1 College of Medicine, Central Michigan University , Mt Pleasant, Michigan.,2 Department of Pediatric Respiratory Science, The Johns Hopkins University School of Medicine , Baltimore, Maryland
| |
Collapse
|
87
|
Lactosylceramide-accumulation in lipid-rafts mediate aberrant-autophagy, inflammation and apoptosis in cigarette smoke induced emphysema. Apoptosis 2015; 20:725-39. [PMID: 25638276 DOI: 10.1007/s10495-015-1098-0] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Ceramide-accumulation is known to be involved in the pathogenesis of chronic inflammatory lung diseases including cigarette smoke-induced emphysema (CS-emphysema) but the exact sphingolipid metabolite that initiates emphysema progression remains ambiguous. We evaluated here a novel role for the sphingolipid, lactosylceramide (LacCer), as a potential mechanism for pathogenesis of CS-emphysema. We assessed the expression of LacCer, and LacCer-dependent inflammatory, apoptosis and autophagy responses in lungs of mice exposed to CS, as well as peripheral lung tissues from COPD subjects followed by experimental analysis to verify the role of LacCer in CS-emphysema. We observed significantly elevated LacCer-accumulation in human COPD lungs with increasing severity of emphysema over non-emphysema controls. Moreover, increased expression of defective-autophagy marker, p62, in lung tissues of severe COPD subjects suggest that LacCer induced aberrant-autophagy may contribute to the pathogenesis of CS-emphysema. We verified that CS-extract treatment significantly induces LacCer-accumulation in both bronchial-epithelial cells (BEAS2B) and macrophages (Raw264.7) as a mechanism to initiate aberrant-autophagy (p62-accumulation) and apoptosis that was rescued by pharmacological inhibitor of LacCer-synthase. Further, we corroborated that CS exposure induces LacCer-accumulation in murine lungs that can be controlled by LacCer-synthase inhibitor. We propose LacCer-accumulation as a novel prognosticator of COPD-emphysema severity, and provide evidence on the therapeutic efficacy of LacCer-synthase inhibitor in CS induced COPD-emphysema.
Collapse
|
88
|
Koga T, Suico MA, Shimasaki S, Watanabe E, Kai Y, Koyama K, Omachi K, Morino-Koga S, Sato T, Shuto T, Mori K, Hino S, Nakao M, Kai H. Endoplasmic Reticulum (ER) Stress Induces Sirtuin 1 (SIRT1) Expression via the PI3K-Akt-GSK3β Signaling Pathway and Promotes Hepatocellular Injury. J Biol Chem 2015; 290:30366-74. [PMID: 26499802 DOI: 10.1074/jbc.m115.664169] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2015] [Indexed: 12/31/2022] Open
Abstract
Sirtuin 1 (SIRT1), an NAD(+)-dependent histone deacetylase, plays crucial roles in various biological processes including longevity, stress response, and cell survival. Endoplasmic reticulum (ER) stress is caused by dysfunction of ER homeostasis and exacerbates various diseases including diabetes, fatty liver, and chronic obstructive pulmonary disease. Although several reports have shown that SIRT1 negatively regulates ER stress and ER stress-induced responses in vitro and in vivo, the effect of ER stress on SIRT1 is less explored. In this study, we showed that ER stress induced SIRT1 expression in vitro and in vivo. We further determined the molecular mechanisms of how ER stress induces SIRT1 expression. Surprisingly, the conventional ER stress-activated transcription factors XBP1, ATF4, and ATF6 seem to be dispensable for SIRT1 induction. Based on inhibitor screening experiments with SIRT1 promoter, we found that the PI3K-Akt-GSK3β signaling pathway is required for SIRT1 induction by ER stress. Moreover, we showed that pharmacological inhibition of SIRT1 by EX527 inhibited the ER stress-induced cellular death in vitro and severe hepatocellular injury in vivo, indicating a detrimental role of SIRT1 in ER stress-induced damage responses. Collectively, these data suggest that SIRT1 expression is up-regulated by ER stress and contributes to ER stress-induced cellular damage.
Collapse
Affiliation(s)
- Tomoaki Koga
- From the Department of Molecular Medicine, Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-honmachi, Kumamoto, 862-0973, Japan, Department of Biochemistry, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan
| | - Mary Ann Suico
- From the Department of Molecular Medicine, Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-honmachi, Kumamoto, 862-0973, Japan
| | - Shogo Shimasaki
- From the Department of Molecular Medicine, Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-honmachi, Kumamoto, 862-0973, Japan
| | - Eriko Watanabe
- From the Department of Molecular Medicine, Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-honmachi, Kumamoto, 862-0973, Japan
| | - Yukari Kai
- From the Department of Molecular Medicine, Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-honmachi, Kumamoto, 862-0973, Japan
| | - Kosuke Koyama
- From the Department of Molecular Medicine, Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-honmachi, Kumamoto, 862-0973, Japan
| | - Kohei Omachi
- From the Department of Molecular Medicine, Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-honmachi, Kumamoto, 862-0973, Japan
| | - Saori Morino-Koga
- From the Department of Molecular Medicine, Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-honmachi, Kumamoto, 862-0973, Japan
| | - Takashi Sato
- From the Department of Molecular Medicine, Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-honmachi, Kumamoto, 862-0973, Japan
| | - Tsuyoshi Shuto
- From the Department of Molecular Medicine, Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-honmachi, Kumamoto, 862-0973, Japan
| | - Kazutoshi Mori
- Department of Biophysics, Graduate School of Science, Kyoto University, Oiwake-machi, Kitashirakawa-oiwake, Sakyo-ku, Kyoto, 606-8502, Japan, and
| | - Shinjiro Hino
- Department of Medical Cell Biology, Institute of Molecular Embryology and Genetics, Kumamoto University, 2-2-1 Honjo, Chuo-ku, Kumamoto, 860-0811, Japan
| | - Mitsuyoshi Nakao
- Department of Medical Cell Biology, Institute of Molecular Embryology and Genetics, Kumamoto University, 2-2-1 Honjo, Chuo-ku, Kumamoto, 860-0811, Japan
| | - Hirofumi Kai
- From the Department of Molecular Medicine, Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-honmachi, Kumamoto, 862-0973, Japan,
| |
Collapse
|
89
|
Tran I, Ji C, Ni I, Min T, Tang D, Vij N. Role of Cigarette Smoke-Induced Aggresome Formation in Chronic Obstructive Pulmonary Disease-Emphysema Pathogenesis. Am J Respir Cell Mol Biol 2015; 53:159-73. [PMID: 25490051 DOI: 10.1165/rcmb.2014-0107oc] [Citation(s) in RCA: 91] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Cigarette smoke (CS) exposure is known to induce proteostasis imbalance that can initiate accumulation of ubiquitinated proteins. Therefore, the primary goal of this study was to determine if first- and secondhand CS induces localization of ubiquitinated proteins in perinuclear spaces as aggresome bodies. Furthermore, we sought to determine the mechanism by which smoke-induced aggresome formation contributes to chronic obstructive pulmonary disease (COPD)-emphysema pathogenesis. Hence, Beas2b cells were treated with CS extract (CSE) for in vitro experimental analysis of CS-induced aggresome formation by immunoblotting, microscopy, and reporter assays, whereas chronic CS-exposed murine model and human COPD-emphysema lung tissues were used for validation. In preliminary analysis, we observed a significant (P < 0.01) increase in ubiquitinated protein aggregation in the insoluble protein fraction of CSE-treated Beas2b cells. We verified that CS-induced ubiquitin aggregrates are localized in the perinuclear spaces as aggresome bodies. These CS-induced aggresomes (P < 0.001) colocalize with autophagy protein microtubule-associated protein 1 light chain-3B(+) autophagy bodies, whereas U.S. Food and Drug Administration-approved autophagy-inducing drug (carbamazepine) significantly (P < 0.01) decreases their colocalization and expression, suggesting CS-impaired autophagy. Moreover, CSE treatment significantly increases valosin-containing protein-p62 protein-protein interaction (P < 0.0005) and p62 expression (aberrant autophagy marker; P < 0.0001), verifying CS-impaired autophagy as an aggresome formation mechanism. We also found that inhibiting protein synthesis by cycloheximide does not deplete CS-induced ubiquitinated protein aggregates, suggesting the role of CS-induced protein synthesis in aggresome formation. Next, we used an emphysema murine model to verify that chronic CS significantly (P < 0.0005) induces aggresome formation. Moreover, we observed that autophagy induction by carbamazepine inhibits CS-induced aggresome formation and alveolar space enlargement (P < 0.001), confirming involvement of aggresome bodies in COPD-emphysema pathogenesis. Finally, significantly higher p62 accumulation in smokers and severe COPD-emphysema lungs (Global Initiative for Chronic Obstructive Lung Disease Stage III/IV) as compared with normal nonsmokers (Global Initiative for Chronic Obstructive Lung Disease Stage 0) substantiates the pathogenic role of autophagy impairment in aggresome formation and COPD-emphysema progression. In conclusion, CS-induced aggresome formation is a novel mechanism involved in COPD-emphysema pathogenesis.
Collapse
Affiliation(s)
- Ian Tran
- Departments of 1 Pediatric Respiratory Science and.,2 Biomedical Engineering, the Johns Hopkins University School of Medicine, Baltimore, Maryland; and
| | - Changhoon Ji
- Departments of 1 Pediatric Respiratory Science and.,2 Biomedical Engineering, the Johns Hopkins University School of Medicine, Baltimore, Maryland; and
| | - Inzer Ni
- Departments of 1 Pediatric Respiratory Science and.,2 Biomedical Engineering, the Johns Hopkins University School of Medicine, Baltimore, Maryland; and
| | - Taehong Min
- Departments of 1 Pediatric Respiratory Science and.,2 Biomedical Engineering, the Johns Hopkins University School of Medicine, Baltimore, Maryland; and
| | - Danni Tang
- Departments of 1 Pediatric Respiratory Science and.,2 Biomedical Engineering, the Johns Hopkins University School of Medicine, Baltimore, Maryland; and
| | - Neeraj Vij
- Departments of 1 Pediatric Respiratory Science and.,3 College of Medicine, Central Michigan University, Mount Pleasant, Michigan
| |
Collapse
|
90
|
Caniard A, Ballweg K, Lukas C, Yildirim AÖ, Eickelberg O, Meiners S. Proteasome function is not impaired in healthy aging of the lung. Aging (Albany NY) 2015; 7:776-792. [PMID: 26540298 PMCID: PMC4637206 DOI: 10.18632/aging.100820] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2015] [Accepted: 09/25/2015] [Indexed: 06/05/2023]
Abstract
Aging is the progressive loss of cellular function which inevitably leads to death. Failure of proteostasis including the decrease in proteasome function is one hallmark of aging. In the lung, proteasome activity was shown to be impaired in age-related diseases such as chronic obstructive pulmonary disease. However, little is known on proteasome function during healthy aging. Here, we comprehensively analyzed healthy lung aging and proteasome function in wildtype, proteasome reporter and immunoproteasome knockout mice. Wildtype mice spontaneously developed senile lung emphysema while expression and activity of proteasome complexes and turnover of ubiquitinated substrates was not grossly altered in lungs of aged mice. Immunoproteasome subunits were specifically upregulated in the aged lung and the caspase-like proteasome activity concomitantly decreased. Aged knockout mice for the LMP2 or LMP7 immunoproteasome subunits showed no alteration in proteasome activities but exhibited typical lung aging phenotypes suggesting that immunoproteasome function is dispensable for physiological lung aging in mice. Our results indicate that healthy aging of the lung does not involve impairment of proteasome function. Apparently, the reserve capacity of the proteostasis systems in the lung is sufficient to avoid severe proteostasis imbalance during healthy aging.
Collapse
Affiliation(s)
- Anne Caniard
- Comprehensive Pneumology Center (CPC), University Hospital Ludwig‐Maximilians University, Helmholtz Zentrum München, Munich, Member of the German Center for Lung Research (DZL), Germany
| | - Korbinian Ballweg
- Comprehensive Pneumology Center (CPC), University Hospital Ludwig‐Maximilians University, Helmholtz Zentrum München, Munich, Member of the German Center for Lung Research (DZL), Germany
| | - Christina Lukas
- Comprehensive Pneumology Center (CPC), University Hospital Ludwig‐Maximilians University, Helmholtz Zentrum München, Munich, Member of the German Center for Lung Research (DZL), Germany
| | - Ali Ö. Yildirim
- Comprehensive Pneumology Center (CPC), University Hospital Ludwig‐Maximilians University, Helmholtz Zentrum München, Munich, Member of the German Center for Lung Research (DZL), Germany
| | - Oliver Eickelberg
- Comprehensive Pneumology Center (CPC), University Hospital Ludwig‐Maximilians University, Helmholtz Zentrum München, Munich, Member of the German Center for Lung Research (DZL), Germany
| | - Silke Meiners
- Comprehensive Pneumology Center (CPC), University Hospital Ludwig‐Maximilians University, Helmholtz Zentrum München, Munich, Member of the German Center for Lung Research (DZL), Germany
| |
Collapse
|
91
|
Matsuoka M, Komoike Y. Experimental Evidence Shows Salubrinal, an eIF2α Dephosphorylation Inhibitor, Reduces Xenotoxicant-Induced Cellular Damage. Int J Mol Sci 2015; 16:16275-87. [PMID: 26193263 PMCID: PMC4519949 DOI: 10.3390/ijms160716275] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Revised: 06/30/2015] [Accepted: 07/10/2015] [Indexed: 12/21/2022] Open
Abstract
Accumulating evidence indicates that endoplasmic reticulum (ER) stress and the subsequent unfolded protein response (UPR) are involved in the pathogenesis of not only the protein misfolding disorders such as certain neurodegenerative and metabolic diseases, but also in the cytotoxicity of environmental pollutants, industrial chemicals, and drugs. Thus, the modulation of ER stress signaling pathways is an important issue for protection against cellular damage induced by xenotoxicants. The substance salubrinal has been shown to prevent dephosphorylation of the eukaryotic translation initiation factor 2 alpha (eIF2α). The phosphorylation of eIF2α appears to be cytoprotective during ER stress, because inhibition of the translation initiation activity of eIF2α reduces global protein synthesis. In addition, the expression of activating transcription factor 4 (ATF4), a transcription factor that induces the expression of UPR target genes, is up-regulated through alternative translation. This review shows that salubrinal can protect cells from the damage induced by a wide range of xenotoxicants, including environmental pollutants and drugs. The canonical and other possible mechanisms of cytoprotection by salubrinal from xenotoxicant-induced ER stress are also discussed.
Collapse
Affiliation(s)
- Masato Matsuoka
- Department of Hygiene and Public Health I, Tokyo Women's Medical University, Tokyo 162-8666, Japan.
| | - Yuta Komoike
- Department of Hygiene and Public Health I, Tokyo Women's Medical University, Tokyo 162-8666, Japan.
| |
Collapse
|
92
|
Cui Y, Niu M, Zhang X, Zhong Z, Wang J, Pang D. High expression of valosin-containing protein predicts poor prognosis in patients with breast carcinoma. Tumour Biol 2015; 36:9919-27. [PMID: 26168958 DOI: 10.1007/s13277-015-3748-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2015] [Accepted: 07/02/2015] [Indexed: 02/03/2023] Open
Abstract
Valosin-containing protein (VCP) is one of the AAA-ATPase superfamily members. The correlation between elevated expression of VCP and progression, prognosis, and the metastatic potential has been identified in various tumor types. However, the clinical impact of VCP in breast carcinoma has not been investigated. In the current study, the expression of VCP in 421 breast tumors and adjacent normal breast tissues was examined to investigate the correlation between VCP expression and clinicopathological features in patients with breast carcinoma. We found that the expression of VCP correlated with the TNM stage, Ki67 labeling, and lymph node metastasis (LNM). The expression of VCP was increased significantly in the cytoplasm of cancer cells compared to normal mammary epithelial cells, which was associated with decreased overall survival rates of patients with breast carcinoma (P < 0.001). In conclusion, this study demonstrates significant correlation between the cytoplasmic expression of VCP and adverse prognosis in breast carcinoma, suggesting that VCP may serve as a prognostic biomarker in breast carcinoma.
Collapse
Affiliation(s)
- Yan Cui
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Haping Road No.150, Nangang District, Harbin, 150081, Heilongjiang Province, China
| | - Ming Niu
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Haping Road No.150, Nangang District, Harbin, 150081, Heilongjiang Province, China
| | - Xianyu Zhang
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Haping Road No.150, Nangang District, Harbin, 150081, Heilongjiang Province, China
| | - Zhenbin Zhong
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Haping Road No.150, Nangang District, Harbin, 150081, Heilongjiang Province, China
| | - Ji Wang
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Haping Road No.150, Nangang District, Harbin, 150081, Heilongjiang Province, China
| | - Da Pang
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Haping Road No.150, Nangang District, Harbin, 150081, Heilongjiang Province, China.
| |
Collapse
|
93
|
Yamada Y, Tomaru U, Ishizu A, Ito T, Kiuchi T, Ono A, Miyajima S, Nagai K, Higashi T, Matsuno Y, Dosaka-Akita H, Nishimura M, Miwa S, Kasahara M. Decreased proteasomal function accelerates cigarette smoke-induced pulmonary emphysema in mice. J Transl Med 2015; 95:625-34. [PMID: 25915723 DOI: 10.1038/labinvest.2015.43] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2014] [Revised: 12/28/2014] [Accepted: 01/23/2015] [Indexed: 12/20/2022] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a disease common in elderly people, characterized by progressive destruction of lung parenchyma and chronic inflammation of the airways. The pathogenesis of COPD remains unclear, but recent studies suggest that oxidative stress-induced apoptosis in alveolar cells contributes to emphysematous lung destruction. The proteasome is a multicatalytic enzyme complex that plays a critical role in proteostasis by rapidly destroying misfolded and modified proteins generated by oxidative and other stresses. Proteasome activity decreases with aging in many organs including lungs, and an age-related decline in proteasomal function has been implicated in various age-related pathologies. However, the role of the proteasome system in the pathogenesis of COPD has not been investigated. Recently, we have established a transgenic (Tg) mouse model with decreased proteasomal chymotrypsin-like activity, showing age-related phenotypes. Using this model, we demonstrate here that decreased proteasomal function accelerates cigarette smoke (CS)-induced pulmonary emphysema. CS-exposed Tg mice showed remarkable airspace enlargement and increased foci of inflammation compared with wild-type controls. Importantly, apoptotic cells were found in the alveolar walls of the affected lungs. Impaired proteasomal activity also enhanced apoptosis in cigarette smoke extract (CSE)-exposed fibroblastic cells derived from mice and humans in vitro. Notably, aggresome formation and prominent nuclear translocation of apoptosis-inducing factor were observed in CSE-exposed fibroblastic cells isolated from Tg mice. Collective evidence suggests that CS exposure and impaired proteasomal activity coordinately enhance apoptotic cell death in the alveolar walls that may be involved in the development and progression of emphysema in susceptible individuals such as the elderly.
Collapse
Affiliation(s)
- Yosuke Yamada
- 1] Department of Pathology, Hokkaido University Graduate School of Medicine, Sapporo, Japan [2] Department of Surgical Pathology, Hokkaido University Hospital, Sapporo, Japan
| | - Utano Tomaru
- Department of Pathology, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Akihiro Ishizu
- Faculty of Health Sciences, Hokkaido University, Sapporo, Japan
| | - Tomoki Ito
- Department of Pathology, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Takayuki Kiuchi
- Department of Pathology, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Ayako Ono
- Department of Pathology, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Syota Miyajima
- Department of Pathology, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Katsura Nagai
- First Department of Medicine, Hokkaido University School of Medicine, Sapporo, Japan
| | - Tsunehito Higashi
- Department of Cellular Pharmacology, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Yoshihiro Matsuno
- Department of Surgical Pathology, Hokkaido University Hospital, Sapporo, Japan
| | - Hirotoshi Dosaka-Akita
- Department of Medical Oncology, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Masaharu Nishimura
- First Department of Medicine, Hokkaido University School of Medicine, Sapporo, Japan
| | - Soichi Miwa
- Department of Cellular Pharmacology, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Masanori Kasahara
- Department of Pathology, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| |
Collapse
|
94
|
Keller IE, Vosyka O, Takenaka S, Kloß A, Dahlmann B, Willems LI, Verdoes M, Overkleeft HS, Marcos E, Adnot S, Hauck SM, Ruppert C, Günther A, Herold S, Ohno S, Adler H, Eickelberg O, Meiners S. Regulation of immunoproteasome function in the lung. Sci Rep 2015; 5:10230. [PMID: 25989070 PMCID: PMC4437306 DOI: 10.1038/srep10230] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2014] [Accepted: 04/07/2015] [Indexed: 12/24/2022] Open
Abstract
Impaired immune function contributes to the development of chronic obstructive pulmonary disease (COPD). Disease progression is further exacerbated by pathogen infections due to impaired immune responses. Elimination of infected cells is achieved by cytotoxic CD8+ T cells that are activated by MHC I-mediated presentation of pathogen-derived antigenic peptides. The immunoproteasome, a specialized form of the proteasome, improves generation of antigenic peptides for MHC I presentation thereby facilitating anti-viral immune responses. However, immunoproteasome function in the lung has not been investigated in detail yet. In this study, we comprehensively characterized the function of immunoproteasomes in the human and murine lung. Parenchymal cells of the lung express low constitutive levels of immunoproteasomes, while they are highly and specifically expressed in alveolar macrophages. Immunoproteasome expression is not altered in whole lung tissue of COPD patients. Novel activity-based probes and native gel analysis revealed that immunoproteasome activities are specifically and rapidly induced by IFNγ treatment in respiratory cells in vitro and by virus infection of the lung in mice. Our results suggest that the lung is potentially capable of mounting an immunoproteasome-mediated efficient adaptive immune response to intracellular infections.
Collapse
Affiliation(s)
- Ilona E Keller
- Comprehensive Pneumology Center (CPC), University Hospital, Ludwig-Maximilians University, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Oliver Vosyka
- 1] Comprehensive Pneumology Center (CPC), University Hospital, Ludwig-Maximilians University, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany [2] Research Unit Protein Science, Helmholtz Zentrum München, Munich, Germany
| | - Shinji Takenaka
- Comprehensive Pneumology Center (CPC), University Hospital, Ludwig-Maximilians University, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Alexander Kloß
- Institute of Biochemistry, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Burkhardt Dahlmann
- Institute of Biochemistry, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Lianne I Willems
- Department of Bio-organic Synthesis, Leiden University, Leiden, The Netherlands
| | - Martijn Verdoes
- Department of Bio-organic Synthesis, Leiden University, Leiden, The Netherlands
| | - Hermen S Overkleeft
- Department of Bio-organic Synthesis, Leiden University, Leiden, The Netherlands
| | - Elisabeth Marcos
- INSERM U955, Département de Physiologie, Université Paris-Est Créteil (UPEC), Créteil, France
| | - Serge Adnot
- INSERM U955, Département de Physiologie, Université Paris-Est Créteil (UPEC), Créteil, France
| | - Stefanie M Hauck
- Research Unit Protein Science, Helmholtz Zentrum München, Munich, Germany
| | - Clemens Ruppert
- Department of Internal Medicine, Justus-Liebig-University Giessen, Universities Giessen &Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Giessen, Germany
| | - Andreas Günther
- 1] Department of Internal Medicine, Justus-Liebig-University Giessen, Universities Giessen &Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Giessen, Germany [2] Agaplesion Pneumologische Klinik Waldhof-Elgershausen, Greifenstein, Germany
| | - Susanne Herold
- Department of Internal Medicine II, Section of Infectious Diseases, Justus- Liebig-University, Universities Giessen &Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Giessen, Germany
| | - Shinji Ohno
- Research Unit Gene Vectors, Helmholtz Zentrum München, Munich, Germany
| | - Heiko Adler
- Research Unit Gene Vectors, Helmholtz Zentrum München, Munich, Germany
| | - Oliver Eickelberg
- Comprehensive Pneumology Center (CPC), University Hospital, Ludwig-Maximilians University, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Silke Meiners
- Comprehensive Pneumology Center (CPC), University Hospital, Ludwig-Maximilians University, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
| |
Collapse
|
95
|
Ni I, Ji C, Vij N. Second-hand cigarette smoke impairs bacterial phagocytosis in macrophages by modulating CFTR dependent lipid-rafts. PLoS One 2015; 10:e0121200. [PMID: 25794013 PMCID: PMC4368805 DOI: 10.1371/journal.pone.0121200] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2014] [Accepted: 01/27/2015] [Indexed: 01/08/2023] Open
Abstract
Introduction First/Second-hand cigarette-smoke (FHS/SHS) exposure weakens immune defenses inducing chronic obstructive pulmonary disease (COPD) but the underlying mechanisms are not fully understood. Hence, we evaluated if SHS induced changes in membrane/lipid-raft (m-/r)-CFTR (cystic fibrosis transmembrane conductance regulator) expression/activity is a potential mechanism for impaired bacterial phagocytosis in COPD. Methods RAW264.7 murine macrophages were exposed to freshly prepared CS-extract (CSE) containing culture media and/or Pseudomonas-aeruginosa-PA01-GFP for phagocytosis (fluorescence-microscopy), bacterial survival (colony-forming-units-CFU), and immunoblotting assays. The CFTR-expression/activity and lipid-rafts were modulated by transient-transfection or inhibitors/inducers. Next, mice were exposed to acute/sub-chronic-SHS or room-air (5-days/3-weeks) and infected with PA01-GFP, followed by quantification of bacterial survival by CFU-assay. Results We investigated the effect of CSE treatment on RAW264.7 cells infected by PA01-GFP and observed that CSE treatment significantly (p<0.01) inhibits PA01-GFP phagocytosis as compared to the controls. We also verified this in murine model, exposed to acute/sub-chronic-SHS and found significant (p<0.05, p<0.02) increase in bacterial survival in the SHS-exposed lungs as compared to the room-air controls. Next, we examined the effect of impaired CFTR ion-channel-activity on PA01-GFP infection of RAW264.7 cells using CFTR172-inhibitor and found no significant change in phagocytosis. We also similarly evaluated the effect of a CFTR corrector-potentiator compound, VRT-532, and observed no significant rescue of CSE impaired PA01-GFP phagocytosis although it significantly (p<0.05) decreases CSE induced bacterial survival. Moreover, induction of CFTR expression in macrophages significantly (p<0.03) improves CSE impaired PA01-GFP phagocytosis as compared to the control. Next, we verified the link between m-/r-CFTR expression and phagocytosis using methyl-β-cyclodextran (CD), as it is known to deplete CFTR from membrane lipid-rafts. We observed that CD treatment significantly (p<0.01) inhibits bacterial phagocytosis in RAW264.7 cells and adding CSE further impairs phagocytosis suggesting synergistic effect on CFTR dependent lipid-rafts. Conclusion Our data suggest that SHS impairs bacterial phagocytosis by modulating CFTR dependent lipid-rafts.
Collapse
Affiliation(s)
- Inzer Ni
- Department of Pediatric Respiratory Science, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Department of Biomedical Engineering, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Changhoon Ji
- Department of Pediatric Respiratory Science, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Department of Biomedical Engineering, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Neeraj Vij
- Department of Pediatric Respiratory Science, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Department of Foundational Sciences, College of Medicine, Central Michigan University, Mount Pleasant, Michigan, United States of America
- * E-mail:
| |
Collapse
|
96
|
Identification and characterization of PERK activators by phenotypic screening and their effects on NRF2 activation. PLoS One 2015; 10:e0119738. [PMID: 25780921 PMCID: PMC4363567 DOI: 10.1371/journal.pone.0119738] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Accepted: 01/16/2015] [Indexed: 01/22/2023] Open
Abstract
Endoplasmic reticulum stress plays a critical role to restore the homeostasis of protein production in eukaryotic cells. This vital process is hence involved in many types of diseases including COPD. PERK, one branch in the ER stress signaling pathways, has been reported to activate NRF2 signaling pathway, a known protective response to COPD. Based on this scientific rationale, we aimed to identify PERK activators as a mechanism to achieve NRF2 activation. In this report, we describe a phenotypic screening assay to identify PERK activators. This assay measures phosphorylation of GFP-tagged eIF2α upon PERK activation via a cell-based LanthaScreen technology. To obtain a robust assay with sufficient signal to background and low variation, multiple parameters were optimized including GFP-tagged eIF2α BacMam concentration, cell density and serum concentration. The assay was validated by a tool compound, Thapsigargin, which induces phosphorylation of eIF2α. In our assay, this compound showed maximal signal window of approximately 2.5-fold with a pEC50 of 8.0, consistent with literature reports. To identify novel PERK activators through phosphorylation of eIF2α, a focused set of 8,400 compounds was screened in this assay at 10 µM. A number of hits were identified and validated. The molecular mechanisms for several selected hits were further characterized in terms of PERK activation and effects on PERK downstream components. Specificity of these compounds in activating PERK was demonstrated with a PERK specific inhibitor and in PERK knockout mouse embryonic fibroblast (MEF) cells. In addition, these hits showed NRF2-dependent anti-oxidant gene induction. In summary, our phenotypic screening assay is demonstrated to be able to identify PERK specific activators. The identified PERK activators could potentially be used as chemical probes to further investigate this pathway as well as the link between PERK activation and NRF2 pathway activation.
Collapse
|
97
|
Huang C, Wang JJ, Ma JH, Jin C, Yu Q, Zhang SX. Activation of the UPR protects against cigarette smoke-induced RPE apoptosis through up-regulation of Nrf2. J Biol Chem 2015; 290:5367-80. [PMID: 25568320 PMCID: PMC4342454 DOI: 10.1074/jbc.m114.603738] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2014] [Revised: 12/22/2014] [Indexed: 11/06/2022] Open
Abstract
Recent studies have revealed a role of endoplasmic reticulum (ER) stress-induced unfolded protein response (UPR) in the regulation of RPE cell activity and survival. Herein, we examined the mechanisms by which the UPR modulates apoptotic signaling in human RPE cells challenged with cigarette smoking extract (CSE). Our results show that CSE exposure induced a dose- and time-dependent increase in ER stress markers, enhanced reactive oxygen species (ROS), mitochondrial fragmentation, and apoptosis of RPE cells. These changes were prevented by the anti-oxidant NAC or chemical chaperone TMAO, suggesting a close interaction between oxidative and ER stress in CSE-induced apoptosis. To decipher the role of the UPR, overexpression or down-regulation of XBP1 and CHOP genes was manipulated by adenovirus or siRNA. Overexpressing XBP1 protected against CSE-induced apoptosis by reducing CHOP, p-p38, and caspase-3 activation. In contrast, XBP1 knockdown sensitized the cells to CSE-induced apoptosis, which is likely through a CHOP-independent pathway. Surprisingly, knockdown of CHOP reduced p-eIF2α and Nrf2 resulting in a marked increase in caspase-3 activation and apoptosis. Furthermore, Nrf2 inhibition increased ER stress and exacerbated cell apoptosis, while Nrf2 overexpression reduced CHOP and protected RPE cells. Our data suggest that although CHOP may function as a pro-apoptotic gene during ER stress, it is also required for Nrf2 up-regulation and RPE cell survival. In addition, enhancing Nrf2 and XBP1 activity may help reduce oxidative and ER stress and protect RPE cells from cigarette smoke-induced damage.
Collapse
Affiliation(s)
- Chuangxin Huang
- From the Department of Ophthalmology/Ross Eye Institute, School of Medicine and Biomedical Sciences, University at Buffalo, The State University of New York, Buffalo, New York 14215, SUNY Eye Institute, The State University of New York, Buffalo, New York 14215, State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, 510060 China, and
| | - Joshua J Wang
- From the Department of Ophthalmology/Ross Eye Institute, School of Medicine and Biomedical Sciences, University at Buffalo, The State University of New York, Buffalo, New York 14215, SUNY Eye Institute, The State University of New York, Buffalo, New York 14215
| | - Jacey H Ma
- From the Department of Ophthalmology/Ross Eye Institute, School of Medicine and Biomedical Sciences, University at Buffalo, The State University of New York, Buffalo, New York 14215, SUNY Eye Institute, The State University of New York, Buffalo, New York 14215, State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, 510060 China, and
| | - Chenjin Jin
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, 510060 China, and
| | - Qiang Yu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, 510060 China, and
| | - Sarah X Zhang
- From the Department of Ophthalmology/Ross Eye Institute, School of Medicine and Biomedical Sciences, University at Buffalo, The State University of New York, Buffalo, New York 14215, SUNY Eye Institute, The State University of New York, Buffalo, New York 14215, Department of Biochemistry, School of Medicine and Biomedical Sciences, University at Buffalo, The State University of New York, Buffalo, New York 14215
| |
Collapse
|
98
|
Meiners S, Keller IE, Semren N, Caniard A. Regulation of the proteasome: evaluating the lung proteasome as a new therapeutic target. Antioxid Redox Signal 2014; 21:2364-82. [PMID: 24437504 DOI: 10.1089/ars.2013.5798] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
SIGNIFICANCE Lung diseases are on the second rank worldwide with respect to morbidity and mortality. For most respiratory diseases, no effective therapies exist. Whereas the proteasome has been successfully evaluated as a novel target for therapeutic interventions in cancer, neurodegenerative, and cardiac disorders, there is a profound lack of knowledge on the regulation of proteasome activity in chronic and acute lung diseases. RECENT ADVANCES There are various means of how the amount of active proteasome complexes in the cell can be regulated such as transcriptional regulation of proteasomal subunit expression, association with different regulators, assembly and half-life of proteasomes and regulatory complexes, as well as post-translational modifications. It also becomes increasingly evident that proteasome activity is fine-tuned and depends on the state of the cell. We propose here that 20S proteasomes and their regulators can be regarded as dynamic building blocks, which assemble or disassemble in response to cellular needs. The composition of proteasome complexes in a cell may vary depending on tissue, cell type and compartment, stage of development, or pathological context. CRITICAL ISSUES AND FUTURE DIRECTIONS Dissecting the expression and regulation of the various catalytic forms of 20S proteasomes, such as constitutive, immuno-, and mixed proteasomes, together with their associated regulatory complexes will not only greatly enhance our understanding of proteasome function in lung pathogenesis but will also pave the way to develop new classes of drugs that inhibit or activate proteasome function in a defined setting for treatment of lung diseases.
Collapse
Affiliation(s)
- Silke Meiners
- Comprehensive Pneumology Center (CPC), University Hospital , Ludwig-Maximilians University, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
| | | | | | | |
Collapse
|
99
|
Stepaniants S, Wang IM, Boie Y, Mortimer J, Kennedy B, Elliott M, Hayashi S, Luo H, Wong J, Loy L, Coulter S, Roberts CJ, Hogg JC, Sin DD, O'Neill G, Crackower M, Morris M, Paré PD, Obeidat M. Genes related to emphysema are enriched for ubiquitination pathways. BMC Pulm Med 2014; 14:187. [PMID: 25432663 PMCID: PMC4280711 DOI: 10.1186/1471-2466-14-187] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2014] [Accepted: 11/19/2014] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND Increased small airway resistance and decreased lung elasticity contribute to the airflow limitation in chronic obstructive pulmonary disease (COPD). The lesion that corresponds to loss of lung elasticity is emphysema; the small airway obstruction is due to inflammatory narrowing and obliteration. Despite their convergence in altered physiology, different mechanisms contribute to these processes. The relationships between gene expression and these specific phenotypes may be more revealing than comparison with lung function. METHODS We measured the ratio of alveolar surface area to lung volume (SA/V) in lung tissue from 43 smokers. Two samples from 21 subjects, in which SA/V differed by >49 cm2/mL were profiled to select genes whose expression correlated with SA/V. Significant genes were tested for replication in the 22 remaining subjects. RESULTS The level of expression of 181 transcripts was related to SA/V ( p < 0.05). When these genes were tested in the 22 remaining subjects as a replication, thirty of the 181 genes remained significantly associated with SA/V (P < 0.05) and the direction of association was the same in 164/181. Pathway and network analysis revealed enrichment of genes involved in protein ubiquitination, and western blotting showed altered expression of genes involved in protein ubiquitination in obstructed individuals. CONCLUSION This study implicates modified protein ubiquitination and degradation as a potentially important pathway in the pathogenesis of emphysema.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Peter D Paré
- University of British Columbia Centre for Heart and Lung Innovation, St Paul's Hospital, 1081 Burrard St, Vancouver V6Z 1Y6, BC, Canada.
| | | |
Collapse
|
100
|
Balch WE, Sznajder JI, Budinger S, Finley D, Laposky AD, Cuervo AM, Benjamin IJ, Barreiro E, Morimoto RI, Postow L, Weissman AM, Gail D, Banks-Schlegel S, Croxton T, Gan W. Malfolded protein structure and proteostasis in lung diseases. Am J Respir Crit Care Med 2014; 189:96-103. [PMID: 24033344 DOI: 10.1164/rccm.201306-1164ws] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Recent discoveries indicate that disorders of protein folding and degradation play a particularly important role in the development of lung diseases and their associated complications. The overarching purpose of the National Heart, Lung, and Blood Institute workshop on "Malformed Protein Structure and Proteostasis in Lung Diseases" was to identify mechanistic and clinical research opportunities indicated by these recent discoveries in proteostasis science that will advance our molecular understanding of lung pathobiology and facilitate the development of new diagnostic and therapeutic strategies for the prevention and treatment of lung disease. The workshop's discussion focused on identifying gaps in scientific knowledge with respect to proteostasis and lung disease, discussing new research advances and opportunities in protein folding science, and highlighting novel technologies with potential therapeutic applications for diagnosis and treatment.
Collapse
Affiliation(s)
- William E Balch
- 1 Department of Cell Biology and Chemical Physiology, The Scripps Research Institute, La Jolla, California
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|