1
|
Guo X, Ren H, Sun P, Ding E, Fang J, Fang K, Ma X, Li C, Li C, Xu Y, Cao K, Lin EZ, Guo P, Pollitt KJG, Tong S, Tang S, Shi X. Personal exposure to airborne organic pollutants and lung function changes among healthy older adults. ENVIRONMENTAL RESEARCH 2024; 258:119411. [PMID: 38876423 DOI: 10.1016/j.envres.2024.119411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 06/07/2024] [Accepted: 06/10/2024] [Indexed: 06/16/2024]
Abstract
Epidemiological evidence on the impact of airborne organic pollutants on lung function among the elderly is limited, and their underlying biological mechanisms remain largely unexplored. Herein, a longitudinal panel study was conducted in Jinan, Shandong Province, China, involving 76 healthy older adults monitored over a span of five months repetitively. We systematically evaluated personal exposure to a diverse range of airborne organic pollutants using a wearable passive sampler and their effects on lung function. Participants' pulmonary function indicators were assessed, complemented by comprehensive multi-omics analyses of blood and urine samples. Leveraging the power of interaction analysis, causal inference test (CIT), and integrative pathway analysis (IPA), we explored intricate relationships between specific organic pollutants, biomolecules, and lung function deterioration, elucidating the biological mechanisms underpinning the adverse impacts of these pollutants. We observed that bis (2-chloro-1-methylethyl) ether (BCIE) was significantly associated with negative changes in the forced vital capacity (FVC), with glycerolipids mitigating this adverse effect. Additionally, 31 canonical pathways [e.g., high mobility group box 1 (HMGB1) signaling, phosphatidylinositol 3-kinase (PI3K)/AKT pathway, epithelial mesenchymal transition, and heme and nicotinamide adenine dinucleotide (NAD) biosynthesis] were identified as potential mechanisms. These findings may hold significant implications for developing effective strategies to prevent and mitigate respiratory health risks arising from exposure to such airborne pollutants. However, due to certain limitations of the study, our results should be interpreted with caution.
Collapse
Affiliation(s)
- Xiaojie Guo
- China CDC Key Laboratory of Environment and Population Health, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing 100021, China; School of Public Health, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Huimin Ren
- China CDC Key Laboratory of Environment and Population Health, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing 100021, China; School of Public Health, China Medical University, Shenyang, Liaoning 110001, China
| | - Peijie Sun
- China CDC Key Laboratory of Environment and Population Health, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing 100021, China; School of Public Health, China Medical University, Shenyang, Liaoning 110001, China
| | - Enmin Ding
- China CDC Key Laboratory of Environment and Population Health, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing 100021, China; School of Population Medicine and Public Health, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Jianlong Fang
- China CDC Key Laboratory of Environment and Population Health, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing 100021, China
| | - Ke Fang
- China CDC Key Laboratory of Environment and Population Health, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing 100021, China
| | - Xiao Ma
- China CDC Key Laboratory of Environment and Population Health, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing 100021, China; School of Public Health, Shandong University, Jinan, Shandong 250100, China
| | - Chenfeng Li
- China CDC Key Laboratory of Environment and Population Health, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing 100021, China; School of Public Health, Anhui Medical University, Hefei, Anhui 230032, China
| | - Chenlong Li
- China CDC Key Laboratory of Environment and Population Health, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing 100021, China; School of Public Health, Shandong University, Jinan, Shandong 250100, China
| | - Yibo Xu
- China CDC Key Laboratory of Environment and Population Health, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing 100021, China; School of Public Health, China Medical University, Shenyang, Liaoning 110001, China
| | - Kangning Cao
- China CDC Key Laboratory of Environment and Population Health, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing 100021, China; School of Public Health, Anhui Medical University, Hefei, Anhui 230032, China
| | - Elizabeth Z Lin
- Department of Environmental Health Sciences, Yale School of Public Health, New Haven, CT 06510, USA
| | - Pengfei Guo
- Department of Environmental Health Sciences, Yale School of Public Health, New Haven, CT 06510, USA
| | - Krystal J Godri Pollitt
- Department of Environmental Health Sciences, Yale School of Public Health, New Haven, CT 06510, USA
| | - Shilu Tong
- China CDC Key Laboratory of Environment and Population Health, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing 100021, China; School of Public Health and Social Work, Queensland University of Technology, Brisbane 4001, Australia
| | - Song Tang
- China CDC Key Laboratory of Environment and Population Health, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing 100021, China; Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China; National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing 102206, China.
| | - Xiaoming Shi
- China CDC Key Laboratory of Environment and Population Health, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing 100021, China; Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China; National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing 102206, China.
| |
Collapse
|
2
|
Patel VK, Kokkinis S, De Rubis G, Hansbro PM, Paudel KR, Dua K. Curcumin liposomes attenuate the expression of cigarette smoke extract-induced inflammatory markers IL-8 and IL-24 in vitro. EXCLI JOURNAL 2024; 23:904-907. [PMID: 39165583 PMCID: PMC11333702 DOI: 10.17179/excli2024-7467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 06/11/2024] [Indexed: 08/22/2024]
Affiliation(s)
- Vyoma K. Patel
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Sydney, NSW 2007, Australia
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, Australia
- Faculty of Health and Medicine, School of Clinical Medicine, University of New South Wales, NSW 2052, Australia
| | - Sofia Kokkinis
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Sydney, NSW 2007, Australia
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, Australia
- Pharmako Biotechnologies, Frenchs Forest, NSW 2086, Australia
| | - Gabriele De Rubis
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Sydney, NSW 2007, Australia
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, Australia
| | - Philip Michael Hansbro
- Centre for Inflammation, Centenary Institute and University of Technology Sydney, Faculty of Science, School of Life Sciences, Sydney 2007, Australia
| | - Keshav Raj Paudel
- Centre for Inflammation, Centenary Institute and University of Technology Sydney, Faculty of Science, School of Life Sciences, Sydney 2007, Australia
| | - Kamal Dua
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Sydney, NSW 2007, Australia
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, Australia
| |
Collapse
|
3
|
Banzato R, Pinheiro-Menegasso NM, Novelli FPRS, Olivo CR, Taguchi L, de Oliveira Santos S, Fukuzaki S, Teodoro WPR, Lopes FDTQS, Tibério IFLC, de Toledo-Arruda AC, Prado MAM, Prado VF, Prado CM. Alpha-7 Nicotinic Receptor Agonist Protects Mice Against Pulmonary Emphysema Induced by Elastase. Inflammation 2024; 47:958-974. [PMID: 38227123 DOI: 10.1007/s10753-023-01953-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 12/14/2023] [Accepted: 12/15/2023] [Indexed: 01/17/2024]
Abstract
Pulmonary emphysema is a primary component of chronic obstructive pulmonary disease (COPD), a life-threatening disorder characterized by lung inflammation and restricted airflow, primarily resulting from the destruction of small airways and alveolar walls. Cumulative evidence suggests that nicotinic receptors, especially the α7 subtype (α7nAChR), is required for anti-inflammatory cholinergic responses. We postulated that the stimulation of α7nAChR could offer therapeutic benefits in the context of pulmonary emphysema. To investigate this, we assessed the potential protective effects of PNU-282987, a selective α7nAChR agonist, using an experimental emphysema model. Male mice (C57BL/6) were submitted to a nasal instillation of porcine pancreatic elastase (PPE) (50 µl, 0.667 IU) to induce emphysema. Treatment with PNU-282987 (2.0 mg/kg, ip) was performed pre and post-emphysema induction by measuring anti-inflammatory effects (inflammatory cells, cytokines) as well as anti-remodeling and anti-oxidant effects. Elastase-induced emphysema led to an increase in the number of α7nAChR-positive cells in the lungs. Notably, both groups treated with PNU-282987 (prior to and following emphysema induction) exhibited a significant decrease in the number of α7nAChR-positive cells. Furthermore, both groups treated with PNU-282987 demonstrated decreased levels of macrophages, IL-6, IL-1β, collagen, and elastic fiber deposition. Additionally, both groups exhibited reduced STAT3 phosphorylation and lower levels of SOCS3. Of particular note, in the post-treated group, PNU-282987 successfully attenuated alveolar enlargement, decreased IL-17 and TNF-α levels, and reduced the recruitment of polymorphonuclear cells to the lung parenchyma. Significantly, it is worth noting that MLA, an antagonist of α7nAChR, counteracted the protective effects of PNU-282987 in relation to certain crucial inflammatory parameters. In summary, these findings unequivocally demonstrate the protective abilities of α7nAChR against elastase-induced emphysema, strongly supporting α7nAChR as a pivotal therapeutic target for ameliorating pulmonary emphysema.
Collapse
Affiliation(s)
- Rosana Banzato
- Department of Internal Medicine, School of Medicine, Universidade de São Paulo, São Paulo, Brazil
| | - Nathalia M Pinheiro-Menegasso
- Department of Biosciences, Instituto de Saúde e Sociedade, Universidade Federal de São Paulo, Rua Silva Jardim 136 sala 312, Santos, SP, 11015-020, Brazil
| | | | - Clarice R Olivo
- Department of Internal Medicine, School of Medicine, Universidade de São Paulo, São Paulo, Brazil
| | - Laura Taguchi
- Department of Biosciences, Instituto de Saúde e Sociedade, Universidade Federal de São Paulo, Rua Silva Jardim 136 sala 312, Santos, SP, 11015-020, Brazil
| | - Stheffany de Oliveira Santos
- Department of Biosciences, Instituto de Saúde e Sociedade, Universidade Federal de São Paulo, Rua Silva Jardim 136 sala 312, Santos, SP, 11015-020, Brazil
| | - Silvia Fukuzaki
- Department of Internal Medicine, School of Medicine, Universidade de São Paulo, São Paulo, Brazil
| | - Walcy Paganelli Rosolia Teodoro
- Rheumatology Division of the Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo, FMUSP, São Paulo, Brazil
| | - Fernanda D T Q S Lopes
- Department of Internal Medicine, School of Medicine, Universidade de São Paulo, São Paulo, Brazil
| | - Iolanda F L C Tibério
- Department of Internal Medicine, School of Medicine, Universidade de São Paulo, São Paulo, Brazil
| | | | - Marco Antônio M Prado
- Department of Physiology & Pharmacology, University of Western Ontario, London, Canada
- Department of Anatomy & Cell Biology, University of Western Ontario, London, Canada
| | - Vânia F Prado
- Department of Physiology & Pharmacology, University of Western Ontario, London, Canada
- Department of Anatomy & Cell Biology, University of Western Ontario, London, Canada
| | - Carla M Prado
- Department of Internal Medicine, School of Medicine, Universidade de São Paulo, São Paulo, Brazil.
- Department of Biosciences, Instituto de Saúde e Sociedade, Universidade Federal de São Paulo, Rua Silva Jardim 136 sala 312, Santos, SP, 11015-020, Brazil.
| |
Collapse
|
4
|
Fang H, Dong T, Li S, Zhang Y, Han Z, Liu M, Dong W, Hong Z, Fu M, Zhang H. A Bibliometric Analysis of Comorbidity of COPD and Lung Cancer: Research Status and Future Directions. Int J Chron Obstruct Pulmon Dis 2023; 18:3049-3065. [PMID: 38149238 PMCID: PMC10750778 DOI: 10.2147/copd.s425735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Accepted: 12/15/2023] [Indexed: 12/28/2023] Open
Abstract
Objective Although studies on the association between COPD and lung cancer are of great significance, no bibliometric analysis has been conducted in the field of their comorbidity. This bibliometric analysis explores the current situation and frontier trends in the field of COPD and lung cancer comorbidity, and to lay a new direction for subsequent research. Methods Articles in the field of COPD and cancer comorbidity were retrieved from Web of Science Core Collections (WoSCC) from 2004 to 2023, and analyzed by VOSviewer, CiteSpace, Biblimatrix and WPS Office. Results In total, 3330 publications were included. The USA was the leading country with the most publications and great influence. The University of Groningen was the most productive institution. Edwin Kepner Silverman was the most influential scholar in this field. PLOS One was found to be the most prolific journal. Mechanisms and risk factors were of vital importance in this research field. Environmental pollution and pulmonary fibrosis may be future research prospects. Conclusion This bibliometric analysis provided new guidance for the development of the field of COPD and lung cancer comorbidity by visualizing current research hotspots, and predicting possible hot research directions in the future.
Collapse
Affiliation(s)
- Hanyu Fang
- Graduate School, Beijing University of Chinese Medicine, Beijing, People’s Republic of China, 100029
- Department of Traditional Chinese Medicine for Pulmonary Diseases, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, 100029, People’s Republic of China
| | - Tairan Dong
- Graduate School, Beijing University of Chinese Medicine, Beijing, People’s Republic of China, 100029
| | - Shanlin Li
- Graduate School, Beijing University of Chinese Medicine, Beijing, People’s Republic of China, 100029
| | - Yihan Zhang
- Graduate School, Beijing University of Chinese Medicine, Beijing, People’s Republic of China, 100029
| | - Zhuojun Han
- Graduate School, Beijing University of Chinese Medicine, Beijing, People’s Republic of China, 100029
| | - Mingfei Liu
- Graduate School, Beijing University of Chinese Medicine, Beijing, People’s Republic of China, 100029
- Department of Traditional Chinese Medicine for Pulmonary Diseases, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, 100029, People’s Republic of China
| | - Wenjun Dong
- Graduate School, Beijing University of Chinese Medicine, Beijing, People’s Republic of China, 100029
- Department of Traditional Chinese Medicine for Pulmonary Diseases, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, 100029, People’s Republic of China
| | - Zheng Hong
- Graduate School, Beijing University of Chinese Medicine, Beijing, People’s Republic of China, 100029
- Department of Traditional Chinese Medicine for Pulmonary Diseases, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, 100029, People’s Republic of China
| | - Min Fu
- Department of Infectious Diseases, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, 100029, People’s Republic of China
| | - Hongchun Zhang
- Graduate School, Beijing University of Chinese Medicine, Beijing, People’s Republic of China, 100029
- Department of Traditional Chinese Medicine for Pulmonary Diseases, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, 100029, People’s Republic of China
| |
Collapse
|
5
|
Liu YF, Tang MM, Sun J, Li JF, Jiang YL, Zhao H, Fu L. Arsenic exposure and lung function decline in chronic obstructive pulmonary disease patients: The mediating influence of systematic inflammation and oxidative stress. Food Chem Toxicol 2023; 181:114044. [PMID: 37777081 DOI: 10.1016/j.fct.2023.114044] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2023] [Revised: 09/03/2023] [Accepted: 09/17/2023] [Indexed: 10/02/2023]
Abstract
Lung tissue is one of the target sites of arsenic (As). The goal of this investigation was to assess the associations of blood As concentration with pulmonary function indicators in patients with chronic obstructive pulmonary disease (COPD), as well as the roles of systemic inflammation and oxidative stress in this relationship. All 791 COPD patients were selected. Blood As concentration, and tumour necrosis factor-α (TNF-α) and 8-iso-prostaglandin-F2α (8-iso-PGF2α) were detected in the serum of COPD cases. Blood As was robustly related to pulmonary function parameters in an inverse dose-dependent manner. Multivariate linear regression analyses verified that a 1-unit increase of blood As was linked to declines of 0.263 L in FVC, 0.288 L in FEV1, 3.454 in FEV1/FVC%, and 0.538 in predicted FEV1%, respectively. The potential for pulmonary function decline gradually increased across the elevated tertiles of blood As. Nonsmokers were susceptible to As-induced pulmonary function reduction. Blood As was positively linked to the levels of TNF-α and 8-iso-PGF2α. Increased TNF-α and 8-iso-PGF2α partially mediated As-induced the reductions in FEV1 and FVC among COPD patients. As exposure is intensely linked to pulmonary function reduction. Systematic inflammation and oxidative stress partially mediate such associations in COPD patients.
Collapse
Affiliation(s)
- Yun-Feng Liu
- Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230032, China; Institute of Respiratory Diseases, Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230601, China
| | - Min-Min Tang
- Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230032, China; Institute of Respiratory Diseases, Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230601, China
| | - Jing Sun
- Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230032, China; Institute of Respiratory Diseases, Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230601, China
| | - Jia-Fei Li
- Department of Respiratory and Critical Care Medicine, The First People's Hospital of Chuzhou, Chuzhou, Anhui, 239001, China
| | - Ya-Lin Jiang
- Department of Respiratory and Critical Care Medicine, Bozhou People's Hospital, Bozhou, Anhui, 236800, China
| | - Hui Zhao
- Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230032, China; Institute of Respiratory Diseases, Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230601, China.
| | - Lin Fu
- Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230032, China; Institute of Respiratory Diseases, Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230601, China.
| |
Collapse
|
6
|
Pereira CC, Pedroso CF, Batista SRR, Guimarães RA. Prevalence and factors associated with multimorbidity in adults in Brazil, according to sex: a population-based cross-sectional survey. Front Public Health 2023; 11:1193428. [PMID: 37342274 PMCID: PMC10278573 DOI: 10.3389/fpubh.2023.1193428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Accepted: 05/12/2023] [Indexed: 06/22/2023] Open
Abstract
Introduction Multimorbidity, defined as the coexistence of two or more chronic diseases in the same individual, represents a significant health challenge. However, there is limited evidence on its prevalence and associated factors in developing countries, such as Brazil, especially stratified by sex. Thus, this study aims to estimate the prevalence and analyze the factors associated with multimorbidity in Brazilian adults according to sex. Methods Cross-sectional population-based household survey carried out with Brazilian adults aged 18 years or older. The sampling strategy consisted of a three-stage conglomerate plan. The three stages were performed through simple random sampling. Data were collected through individual interviews. Multimorbidity was classified based on a list of 14 self-reported chronic diseases/conditions. Poisson regression analysis was performed to estimate the magnitude of the association between sociodemographic and lifestyle factors with the prevalence of multimorbidity stratified by sex. Results A total of 88,531 individuals were included. In absolute terms, the prevalence of multimorbidity was 29.4%. The frequency in men and women was 22.7 and 35.4%, respectively. Overall, multimorbidity was more prevalent among women, the older people, residents of the South and Southeast regions, urban area residents, former smokers, current smokers, physically inactive, overweight, and obese adults. Individuals with complete high school/incomplete higher education had a lower prevalence of multimorbidity than those with higher educational level. The associations between education and multimorbidity differed between sexes. In men, multimorbidity was inversely associated with the strata of complete middle school/incomplete high school and complete high school/incomplete higher education, while in women, the association between these variables was not observed. Physical inactivity was positively associated with a higher prevalence of multimorbidity only in men. An inverse association was verified between the recommended fruit and vegetable consumption and multimorbidity for the total sample and both sexes. Conclusion One in four adults had multimorbidity. Prevalence increased with increasing age, among women, and was associated with some lifestyles. Multimorbidity was significantly associated with educational level and physical inactivity only in men. The results suggest the need to adopt integrated strategies to reduce the magnitude of multimorbidity, specific by gender, including actions for health promotion, disease prevention, health surveillance and comprehensive health care in Brazil.
Collapse
Affiliation(s)
| | | | - Sandro Rogério Rodrigues Batista
- Department of Internal Medicine, School of Medicine, Federal University of Goiás, Goiânia, Brazil
- Federal District Health Department, Brasília, Brazil
| | - Rafael Alves Guimarães
- Institute of Tropical Pathology and Public Health, Federal University of Goiás, Goiânia, Brazil
- Faculty of Nursing, Federal University of Goiás, Goiânia, Brazil
| |
Collapse
|
7
|
Cho SB, Kim IK, Kang HS, Lee SH, Yeo CD. S100A8/A9-RAGE pathway and chronic airway inflammation in smoke-induced lung carcinogenesis. Mol Cell Toxicol 2023. [DOI: 10.1007/s13273-023-00339-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/17/2023]
|
8
|
Lung microRNAs Expression in Lung Cancer and COPD: A Preliminary Study. Biomedicines 2023; 11:biomedicines11030736. [PMID: 36979715 PMCID: PMC10045129 DOI: 10.3390/biomedicines11030736] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 02/21/2023] [Accepted: 02/24/2023] [Indexed: 03/05/2023] Open
Abstract
Non-small cell lung cancer (NSCLC) is one of the deadliest diseases worldwide and represents an impending burden on the healthcare system. Despite increasing attention, the mechanisms underlying tumorigenesis in cancer-related diseases such as COPD remain unclear, making novel biomarkers necessary to improve lung cancer early diagnosis. MicroRNAs (miRNAs) are short non-coding RNA that interfere with several pathways and can act as oncogenes or tumor suppressors. This study aimed to compare miRNA lung expression between subjects with NSCLC and COPD and healthy controls to obtain the miRNA expression profile by analyzing shared pathways. Lung specimens were collected from a prospective cohort of 21 sex-matched subjects to determine the tissue miRNA expression of hsa-miR-34a-5p, 33a-5p, 149-3p, 197-3p, 199-5p, and 320a-3p by RT-PCR. In addition, an in silico prediction of miRNA target genes linked to cancer was performed. We found a specific trend for has-miR-149-3p, 197-3p, and 34a-5p in NSCLC, suggesting their possible role as an index of the tumor microenvironment. Moreover, we identified novel miRNA targets, such as the Cyclin-Dependent Kinase (CDK) family, linked to carcinogenesis by in silico analysis. In conclusion. this study identified lung miRNA signatures related to the tumorigenic microenvironment, suggesting their possible role in improving the evaluation of lung cancer onset.
Collapse
|
9
|
Yuan L, Guo T, Hu C, Yang W, Tang X, Cheng H, Xiang Y, Qu X, Liu H, Qin X, Qin L, Liu C. Clinical characteristics and gene mutation profiles of chronic obstructive pulmonary disease in non-small cell lung cancer. Front Oncol 2022; 12:946881. [PMID: 36267961 PMCID: PMC9576924 DOI: 10.3389/fonc.2022.946881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 06/27/2022] [Indexed: 12/03/2022] Open
Abstract
Purpose The coexistence of chronic obstructive pulmonary disease (COPD) often leads to a worse prognosis in patients with non-small cell lung cancer (NSCLC). Meanwhile, approaches targeting specific genetic alterations have been shown to significantly improve the diagnosis and treatment outcomes of patients with NSCLC. Herein, we sought to evaluate the impact of COPD on the clinical manifestations and gene mutation profiles of NSCLC patients with both circulating tumor (ctDNA) and tumor DNA (tDNA). Materials and methods The influence of COPD on clinical features was observed in 285 NSCLC cohorts suffering from NSCLC alone, NSCLC coexisting with COPD, or NSCLC coexisting with prodromal changes in COPD (with emphysema, bullae, or chronic bronchitis). The gene mutation profiles of specific 168 NSCLC-related genes were further analyzed in the NSCLC sub-cohorts with formalin-fixed and paraffin-embedded tumor DNA (FFPE tDNA) samples and plasma circulating tumor DNA (PLA ctDNA) samples. Moreover, mutation concordance was assessed in tDNA and paired ctDNA of 110 NSCLC patients. Results Relative to patients with NSCLC alone, patients with NSCLC coexisting with COPD and prodromal changes presented with worse lung functions, more clinical symptoms, signs and comorbidities, and inconsistent gene mutation profiles. In addition, patients in the latter two groups exhibited a higher average frequency of gene mutation. Lastly, mutation concordance between tDNA and ctDNA samples was significantly reduced in NSCLC patients coexisting with COPD. Conclusions Collectively, our findings revealed that coexistence of COPD leads to worse clinical manifestations and altered gene mutation profiles in patients with NSCLC. Additionally, for NSCLC patients with COPD, the use of ctDNA instead of tDNA may not be the most efficient approach to identifying gene mutations.
Collapse
Affiliation(s)
- Lin Yuan
- Department of Respiratory Medicine, National Key Clinical Specialty, Branch of National Clinical Research Center for Respiratory Disease, Xiangya Hospital, Central South University, Changsha, China
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, China
- Basic and Clinical Research Laboratory of Major Respiratory Diseases, Central South University, Changsha, China
| | - Ting Guo
- Department of Respiratory Medicine, The First Hospital of Changsha, Changsha, China
| | - Chengping Hu
- Department of Respiratory Medicine, National Key Clinical Specialty, Branch of National Clinical Research Center for Respiratory Disease, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, China
| | - Wei Yang
- Department of Respiratory Medicine, National Key Clinical Specialty, Branch of National Clinical Research Center for Respiratory Disease, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, China
| | - Xiaoli Tang
- Pulmonary and Critical Care Medicine, Huaihua Tumor Hospital, Huaihua, China
| | - Hao Cheng
- Department of Radiotherapy, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
- Department of Nasopharyngeal Carcinoma, The First People’s Hospital of Chenzhou, Chenzhou, China
| | - Yang Xiang
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, China
| | - Xiangping Qu
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, China
| | - Huijun Liu
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, China
| | - Xiaoqun Qin
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, China
| | - Ling Qin
- Department of Respiratory Medicine, National Key Clinical Specialty, Branch of National Clinical Research Center for Respiratory Disease, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, China
- *Correspondence: Ling Qin, ; Chi Liu,
| | - Chi Liu
- Department of Respiratory Medicine, National Key Clinical Specialty, Branch of National Clinical Research Center for Respiratory Disease, Xiangya Hospital, Central South University, Changsha, China
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, China
- Basic and Clinical Research Laboratory of Major Respiratory Diseases, Central South University, Changsha, China
- *Correspondence: Ling Qin, ; Chi Liu,
| |
Collapse
|
10
|
Fathinavid A, Mousavian Z, Najafi A, Nematzadeh S, Salimi M, Masoudi-Nejad A. Identifying common signatures and potential therapeutic biomarkers in COPD and lung cancer using miRNA-mRNA co-expression networks. INFORMATICS IN MEDICINE UNLOCKED 2022. [DOI: 10.1016/j.imu.2022.101115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
|
11
|
Fathinavid A, Ghobadi MZ, Najafi A, Masoudi-Nejad A. Identification of common microRNA between COPD and non-small cell lung cancer through pathway enrichment analysis. BMC Genom Data 2021; 22:41. [PMID: 34635059 PMCID: PMC8507163 DOI: 10.1186/s12863-021-00986-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Accepted: 08/20/2021] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Different factors have been introduced which influence the pathogenesis of chronic obstructive pulmonary disease (COPD) and non-small cell lung cancer (NSCLC). COPD as an independent factor is involved in the development of lung cancer. Moreover, there are certain resemblances between NSCLC and COPD, such as growth factors, activation of intracellular pathways, as well as epigenetic factors. One of the best approaches to understand the possible shared pathogenesis routes between COPD and NSCLC is to study the biological pathways that are activated. MicroRNAs (miRNAs) are critical biomolecules that implicate the regulation of several biological and cellular processes. As such, the main goal of this study was to use a systems biology approach to discover common dysregulated miRNAs between COPD and NSCLC, one that targets most genes within common enriched pathways. RESULTS To reconstruct the miRNA-pathways for each disease, we used the microarray miRNA expression data. Then, we employed "miRNA set enrichment analysis" (MiRSEA) to identify the most significant joint miRNAs between COPD and NSCLC based on the enrichment scores. Overall, our study revealed the involvement of the targets of miRNAs (such as has-miR-15b, hsa-miR-106a, has-miR-17, has-miR-103, and has-miR-107) in the most important common biological pathways. CONCLUSIONS According to the promising results of the pathway analysis, the identified miRNAs can be utilized as the new potential signatures for therapy through understanding the molecular mechanisms of both diseases.
Collapse
Affiliation(s)
- Amirhossein Fathinavid
- Laboratory of Systems Biology and Bioinformatics (LBB), Department of Bioinformatics, Kish International Campus, University of Tehran, Kish Island, Iran
| | - Mohadeseh Zarei Ghobadi
- Laboratory of Systems Biology and Bioinformatics (LBB), Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran
| | - Ali Najafi
- Molecular Biology Research Center, System Biology and Poisoning Institute, Tehran, Iran
| | - Ali Masoudi-Nejad
- Laboratory of Systems Biology and Bioinformatics (LBB), Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran.
| |
Collapse
|
12
|
Zhou TJ, Liu JF, Wang P, Hu AN, Chen LL, Zan JF. Identification of Targets and Active Components of Yiqi SanJie Formula Against Lung Neoplasms Based on Network Pharmacology Analysis and Molecular Docking. Nat Prod Commun 2021. [DOI: 10.1177/1934578x21997677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/09/2022] Open
Abstract
Yiqi Sanjie formula (YQSJF) is mainly applied clinically for the treatment of lung neoplasms. The purpose of this study was to explore the pharmacodynamics of the active components of YQSJF and the mechanism of therapeutic effects in the treatment of lung neoplasm diseases based on network pharmacology. The network of component-target, target-pathway, and pathway-disease of YQSJF was constructed by using Cytoscape software. According to the screening result, 37 key components, 57 important targets, and 866 candidate pathways were obtained. The enrichment analysis results indicated that YQSJF might play a therapeutic role in lung cancer by regulating several signaling pathways, such as the PI3K-AKT, non-small cell lung cancer, small cell lung cancer, and apoptosis pathways. There were 53 intersection genes between YQSJF and the lung cancer gene, 52 common genes, and 11 key targets, including CASP8, CASP9, AR, ESR1, PTGS2, NOS3, PGR, TGFB1, PPARG, RELA, and NOS2, screened by using Protein-Protein Interaction (PPI) analysis. These could be the potential therapeutic targets of YQSJF against lung cancer. Enrichment analysis of the intersection gene pathways revealed 10 major functional pathways, including the VEGF, apoptosis, and IL-17 signaling pathways. The molecular docking results showed the potential regulating activity of kaempferol against AR, pelargonidin against PGR, and baicalein against both PTGS2 and AR. In conclusion, combinational network pharmacology analysis results indicated that YQSJF might present its efficacy of alleviating lung neoplasm symptoms through multiple targets in a synergetic way.
Collapse
Affiliation(s)
- Tian-jiao Zhou
- Pharmacy School, Hubei University of Chinese Medicine, Wuhan, China
| | - Jun-feng Liu
- Ministry of Education Key Laboratory of Chinese Medicine Resource and Compound Prescription, Hubei University of Chinese Medicine, Wuhan, China
| | - Ping Wang
- Institute of Geriatrics, Hubei University of Chinese Medicine, Wuhan, China
| | - An-na Hu
- Ministry of Education Key Laboratory of Chinese Medicine Resource and Compound Prescription, Hubei University of Chinese Medicine, Wuhan, China
| | - Lin-lin Chen
- Ministry of Education Key Laboratory of Chinese Medicine Resource and Compound Prescription, Hubei University of Chinese Medicine, Wuhan, China
| | - Jun-feng Zan
- Pharmacy School, Hubei University of Chinese Medicine, Wuhan, China
- Ministry of Education Key Laboratory of Chinese Medicine Resource and Compound Prescription, Hubei University of Chinese Medicine, Wuhan, China
| |
Collapse
|
13
|
Dos Santos Arcas C, Lin-Wang HT, Umeda IIK, de Sousa MG, Utiyama DMO, de Padua Mansur A, Macchione M, Hirata MH, Nakagawa NK. Smoking load reduction is insufficient to downregulate miR-301b, a lung cancer promoter. Sci Rep 2020; 10:21112. [PMID: 33273694 PMCID: PMC7713348 DOI: 10.1038/s41598-020-78242-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Accepted: 11/20/2020] [Indexed: 12/23/2022] Open
Abstract
Several circulating miRNAs identified in the plasma of smokers have been implicated as promoters of nasopharyngeal and lung carcinoma. To investigate the plasma profile of miRNAs in subjects who reduces the number of smoked cigarettes and who quit after six months. We accompanied 28 individuals enrolled in a Smoking Cessation Program over 6 months. At Baseline, clinical characteristics, co-morbidities, and smoking history were similar among subjects. After 6 months, two groups were defined: who successfully quitted smoking (named "quitters", n = 18, mean age 57 years, 11 male) and who reduced the number of cigarettes smoked (20-90%) but failed to quit smoking (named "smokers", n = 10, mean age 52 years, 3 male). No significant clinical changes were observed between groups at baseline and after a 6-month period, however, quitters showed significant downregulations in seven miRNAs at baseline: miR-17 (- 2.90-fold, p = 0.029), miR-20a (- 3.80-fold, p = 0.021); miR-20b (- 4.71-fold, p = 0.027); miR-30a (- 3.95-fold, p = 0.024); miR-93 (- 3.63-fold, p = 0.022); miR-125a (- 1.70-fold, p = 0.038); and miR-195 (- 5.37-fold, p = 0.002), and after a 6-month period in 6 miRNAs: miR-17 (- 5.30-fold, p = 0.012), miR-20a (- 2.04-fold, p = 0.017), miR-20b (- 5.44-fold, p = 0.017), miR-93 (- 4.00-fold, p = 0.041), miR-101 (- 4.82-fold, p = 0.047) and miR-125b (- 3.65-fold, p = 0.025). Using time comparisons, only quitters had significant downregulation in miR-301b (- 2.29-fold, p = 0.038) after 6-month. Reductions in the number of smoked cigarettes was insufficient to change the plasma profile of miRNA after 6 months. Only quitting smoking (100% reduction) significantly downregulated miR-301b related to hypoxic conditions, promotion of cell proliferation, decreases in apoptosis, cancer development, and progression as increases in radiotherapy and chemotherapy resistance.
Collapse
Affiliation(s)
- Camila Dos Santos Arcas
- Department of Physiotherapy, LIM-54, Faculdade de Medicina da Universidade de São Paulo, Av. Dr. Arnaldo, 455 room 1150, São Paulo, São Paulo, 01246-930, Brazil
| | - Hui Tzu Lin-Wang
- Dante Pazzanese Institute of Cardiology São Paulo State, São Paulo, Brazil
| | - Iracema Ioco Kikuchi Umeda
- Department of Physiotherapy, LIM-54, Faculdade de Medicina da Universidade de São Paulo, Av. Dr. Arnaldo, 455 room 1150, São Paulo, São Paulo, 01246-930, Brazil
- Dante Pazzanese Institute of Cardiology São Paulo State, São Paulo, Brazil
| | | | | | - Antonio de Padua Mansur
- Department of Cardiopneumology, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Mariangela Macchione
- Department of Pathology, LIM05, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Mario Hiroyuki Hirata
- Department of Clinical and Toxicological Analysis, School of Pharmaceutical Sciences, Universidade de São Paulo, São Paulo, Brazil
| | - Naomi Kondo Nakagawa
- Department of Physiotherapy, LIM-54, Faculdade de Medicina da Universidade de São Paulo, Av. Dr. Arnaldo, 455 room 1150, São Paulo, São Paulo, 01246-930, Brazil.
| |
Collapse
|
14
|
Heo JW, Kang HS, Park CK, Kim SK, Kim JS, Kim JW, Kim SJ, Lee SH, Yeo CD. Regional emphysema score is associated with tumor location and poor prognosis in completely resected NSCLC patients. BMC Pulm Med 2020; 20:242. [PMID: 32917179 PMCID: PMC7488536 DOI: 10.1186/s12890-020-01268-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Accepted: 08/19/2020] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Lung cancer is a frequent comorbidity of chronic obstructive pulmonary disease (COPD). However, the local risk of developing lung cancer related to regional emphysema distribution and clinical outcome has not been investigated. Our aim was to evaluate the impact of regional emphysema score (RES) on tumor location and prognosis in non-small cell lung cancer (NSCLC) patients. METHODS We enrolled 457 patients who underwent curative surgery for NSCLC at seven hospitals at The Catholic University of Korea from 2014 to 2018. Emphysema was visually assessed for each lobe, with the lingula as a separate lobe. Semi-quantitative emphysema scoring was classified as follows: 0 = none, 0.5 = 1 to 10%, 1 = 11 to 25%, 2 = 26 to 50%, 3 = 51 to 75%, and 4 = 76 to 100%. An RES was given to each of the six lung zone: the upper, middle, and lower lobes in the right and left lungs. RESULTS There were 145 patients in the high RES (≥ 3) group and 312 in the low RES (< 3) group. The mean RES in each lobe with cancer was significantly higher than that in other lobes without cancer (0.51 vs. 0.37, P < 0.001). This group showed significantly shorter disease-free survival (P < 0.001), in addition, presence of COPD, low diffusing capacity of the lung for carbon monoxide (< 80), smoking status, and poor differentiation were more frequent in this group. Also, cancer in a lobe with a higher RES (odds ratio (OR) = 1.56; 95% confidence interval (CI:1.01-2.42; P = 0.04), pathologic stage ≥ III (OR = 2.23; 95% CI: 1.28-3.89; P < 0.001), and poor differentiation (OR = 1.99; 95% CI: 1.22-3.21; P < 0.001) were independent factors for tumor recurrence. CONCLUSIONS The regional severity of emphysema by visual qualification was associated with the location of lung cancer, and was an independently poor prognostic factor for tumor recurrence in completely resected NSCLC patients.
Collapse
Affiliation(s)
- Jung Won Heo
- Division of Pulmonary, Critical Care and Allergy, Department of Internal Medicine, Eunpyeong St. Mary's Hospital, College of Medicine, The Catholic University of Korea, 1021, Tongil-ro, Eunpyeong-gu, Seoul, 03312, Republic of Korea
| | - Hye Seon Kang
- Division of Pulmonary, Critical Care and Allergy, Department of Internal Medicine, Bucheon St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Chan Kwon Park
- Division of Pulmonary, Critical Care and Allergy, Department of Internal Medicine, Yeouido St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Sung Kyoung Kim
- Division of Pulmonary, Critical Care and Allergy, Department of Internal Medicine, St. Vincent's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Ju Sang Kim
- Division of Pulmonary, Critical Care and Allergy, Department of Internal Medicine, Incheon St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Jin Woo Kim
- Division of Pulmonary, Critical Care and Allergy, Department of Internal Medicine, Uijeongbu St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Seung Joon Kim
- Division of Pulmonary, Critical Care and Allergy, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Sang Haak Lee
- Division of Pulmonary, Critical Care and Allergy, Department of Internal Medicine, Eunpyeong St. Mary's Hospital, College of Medicine, The Catholic University of Korea, 1021, Tongil-ro, Eunpyeong-gu, Seoul, 03312, Republic of Korea
| | - Chang Dong Yeo
- Division of Pulmonary, Critical Care and Allergy, Department of Internal Medicine, Eunpyeong St. Mary's Hospital, College of Medicine, The Catholic University of Korea, 1021, Tongil-ro, Eunpyeong-gu, Seoul, 03312, Republic of Korea.
| |
Collapse
|
15
|
Knight DA, Grainge CL, Stick SM, Kicic A, Schuliga M. Epithelial Mesenchymal Transition in Respiratory Disease: Fact or Fiction. Chest 2020; 157:1591-1596. [PMID: 31952949 DOI: 10.1016/j.chest.2019.12.014] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2019] [Revised: 11/20/2019] [Accepted: 12/13/2019] [Indexed: 12/20/2022] Open
Affiliation(s)
- Darryl A Knight
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, NSW, Australia; Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, University of Newcastle, Callaghan, NSW, Australia; Department of Anesthesiology, Pharmacology and Therapeutics, University of British Columbia, Vancouver, BC, Canada; Australian Respiratory Epithelium Consortium, Perth, WA, Australia.
| | - Christopher L Grainge
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, University of Newcastle, Callaghan, NSW, Australia; School of Medicine and Public Health, University of Newcastle, Callaghan, NSW, Australia; Department of Respiratory and Sleep Medicine, John Hunter Hospital, Newcastle, NSW, Australia; Australian Respiratory Epithelium Consortium, Perth, WA, Australia
| | - Stephen M Stick
- Telethon Kids Institute, Subiaco, WA, Australia; Australian Respiratory Epithelium Consortium, Perth, WA, Australia
| | - Anthony Kicic
- Telethon Kids Institute, Subiaco, WA, Australia; Australian Respiratory Epithelium Consortium, Perth, WA, Australia
| | - Michael Schuliga
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, NSW, Australia; Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, University of Newcastle, Callaghan, NSW, Australia; Australian Respiratory Epithelium Consortium, Perth, WA, Australia
| |
Collapse
|
16
|
Parris BA, O'Farrell HE, Fong KM, Yang IA. Chronic obstructive pulmonary disease (COPD) and lung cancer: common pathways for pathogenesis. J Thorac Dis 2019; 11:S2155-S2172. [PMID: 31737343 DOI: 10.21037/jtd.2019.10.54] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Chronic obstructive pulmonary disease (COPD) and lung cancer comprise the leading causes of lung disease-related mortality worldwide. Exposure to tobacco smoke is a mutual aetiology underlying the two diseases, accounting for almost 90% of cases. There is accumulating evidence supporting the role of immune dysfunction, the lung microbiome, extracellular vesicles and underlying genetic susceptibility in the development of COPD and lung cancer. Further, epigenetic factors, involving DNA methylation and microRNA expression, have been implicated in both diseases. Chronic inflammation is a key feature of COPD and could be a potential driver of lung cancer development. Using next generation technologies, further studies investigating the genomics, epigenetics and gene-environment interaction in key molecular pathways will continue to elucidate the pathogenic mechanisms underlying the development of COPD and lung cancer, and contribute to the development of novel diagnostic and prognostic tools for early intervention and personalised therapeutic strategies.
Collapse
Affiliation(s)
- Brielle A Parris
- UQ Thoracic Research Centre, The Prince Charles Hospital, University of Queensland, Brisbane, Australia
| | - Hannah E O'Farrell
- UQ Thoracic Research Centre, The Prince Charles Hospital, University of Queensland, Brisbane, Australia
| | - Kwun M Fong
- UQ Thoracic Research Centre, The Prince Charles Hospital, University of Queensland, Brisbane, Australia.,Department of Thoracic Medicine, The Prince Charles Hospital, Metro North Hospital and Health Service, Brisbane, Australia
| | - Ian A Yang
- UQ Thoracic Research Centre, The Prince Charles Hospital, University of Queensland, Brisbane, Australia.,Department of Thoracic Medicine, The Prince Charles Hospital, Metro North Hospital and Health Service, Brisbane, Australia
| |
Collapse
|
17
|
Advances in Molecular Mechanisms and Immunotherapy Involving the Immune Cell-Promoted Epithelial-to-Mesenchymal Transition in Lung Cancer. JOURNAL OF ONCOLOGY 2019; 2019:7475364. [PMID: 31531020 PMCID: PMC6721259 DOI: 10.1155/2019/7475364] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Revised: 07/24/2019] [Accepted: 08/04/2019] [Indexed: 12/16/2022]
Abstract
Immunotherapy has offered a new opportunity for the treatment of many malignancies. In patients with lung cancer, immune checkpoint inhibitors have significantly improved survival. However, little is known about predictive factors or primary and acquired resistance mechanisms. Epithelial-to-mesenchymal transition (EMT) is a complex of phenotypic changes involved in carcinogenesis and resistance to cancer treatments. Specifically, immune cells in the tumor microenvironment can promote EMT, and mesenchymal phenotype acquisition negatively regulates the anticancer immune response. EMT is associated with higher expression of PD-L1 and other immune checkpoints. In this review, we focused on the role of EMT in the interplay between tumor cells and the immune system, with particular emphasis on lung cancer. On the basis of our findings, we hypothesize that the effects of EMT on immune cells could be overcome in this disease by a new combination of immune checkpoint inhibitors.
Collapse
|
18
|
Media AS, Persson M, Tajhizi N, Weinreich UM. Chronic obstructive pulmonary disease and comorbidities' influence on mortality in non-small cell lung cancer patients. Acta Oncol 2019; 58:1102-1106. [PMID: 31092081 DOI: 10.1080/0284186x.2019.1612942] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Background: In Denmark, lung cancer is the most common cause of cancer-related death and chronic obstructive pulmonary disease (COPD) is the most common comorbidity in patients with non-small cell lung cancer (NSCLC). The aim of this study was to investigate the influence of COPD and other common comorbidities on NSCLC mortality. Methods: Patients (n = 534) diagnosed with NSCLC at Aalborg University Hospital from 2008-2010 were included retrospectively in this study. Patient records were assessed and the population was dichotomized in COPD and non-COPD subgroups. Comorbidities i.e., ischemic heart disease, hypertension, diabetes mellitus, apoplexia, former malignancy, interstitial lung disease and psychiatric comorbidity were registered and a comorbidity count were calculated. Survival was assessed with log-rank test and uni- and multivariate regression analysis were performed for COPD-status and comorbidity count adjusting for age, gender, BMI, smoking exposure, cancer stage, method of treatment and eastern cooperative cancer group (ECOG) performance status score. Results: Of 534 NSCLC patients included, 470 were divided into COPD and non-COPD subgroups, 70% with COPD (329/470) and 30% without COPD (141/470). Only 32.5% of the patients in the COPD-group had previously diagnosed COPD. Log-rank test did not show statistically significant difference in survival between the COPD and non-COPD groups (p = .215). Multivariate Cox regression analysis did not show statistically significant association between overall 5-year mortality and the presence of COPD (HR-adj = 0.808, 95% CI = 0.612; 1.068) or other comorbidities (HR-adj = 1.101, 95% CI = 0.979; 1.237) when adjusted for age, BMI, gender, smoking exposure, ECOG performance status, treatment and TNM-stage. Conclusion: Our findings do not suggest that COPD nor other common comorbidities are significantly associated with higher mortality in NSCLC patients.
Collapse
Affiliation(s)
- Ara Shwan Media
- Department of Cardiothoracic and Vascular Surgery, Aarhus University Hospital, Skejby, Denmark
- Department of Respiratory Diseases, Aalborg University Hospital, Aalborg, Denmark
| | - Martin Persson
- Solljungahälsan Health Center (General Practice), Örkelljunga, Sweden
| | - Navid Tajhizi
- Department of Urology, Örebro University Hospital, Örebro, Sweden
| | - Ulla Møller Weinreich
- Department of Respiratory Diseases, Aalborg University Hospital, Aalborg, Denmark
- Respiratory Research Center, Aalborg University Hospital, Aalborg, Denmark
- The Clinical Institute, Aalborg University, Aalborg, Denmark
| |
Collapse
|
19
|
Engle ML, Monk JN, Jania CM, Martin JR, Gomez JC, Dang H, Parker JS, Doerschuk CM. Dynamic changes in lung responses after single and repeated exposures to cigarette smoke in mice. PLoS One 2019; 14:e0212866. [PMID: 30818335 PMCID: PMC6395068 DOI: 10.1371/journal.pone.0212866] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Accepted: 02/11/2019] [Indexed: 12/18/2022] Open
Abstract
Cigarette smoke is well recognized to cause injury to the airways and the alveolar walls over time. This injury usually requires many years of exposure, suggesting that the lungs may rapidly develop responses that initially protect it from this repetitive injury. Our studies tested the hypotheses that smoke induces an inflammatory response and changes in mRNA profiles that are dependent on sex and the health status of the lung, and that the response of the lungs to smoke differs after 1 day compared to 5 days of exposure. Male and female wildtype (WT) and Scnn1b-transgenic (βENaC) mice, which have chronic bronchitis and emphysematous changes due to dehydrated mucus, were exposed to cigarette smoke or sham air conditions for 1 or 5 days. The inflammatory response and gene expression profiles were analyzed in lung tissue. Overall, the inflammatory response to cigarette smoke was mild, and changes in mediators were more numerous after 1 than 5 days. βENaC mice had more airspace leukocytes than WT mice, and smoke exposure resulted in additional significant alterations. Many genes and gene sets responded similarly at 1 and 5 days: genes involved in oxidative stress responses were upregulated while immune response genes were downregulated. However, certain genes and biological processes were regulated differently after 1 compared to 5 days. Extracellular matrix biology genes and gene sets were upregulated after 1 day but downregulated by 5 days of smoke compared to sham exposure. There was no difference in the transcriptional response to smoke between WT and βENaC mice or between male and female mice at either 1 or 5 days. Taken together, these studies suggest that the lungs rapidly alter gene expression after only one exposure to cigarette smoke, with few additional changes after four additional days of repeated exposure. These changes may contribute to preventing lung damage.
Collapse
Affiliation(s)
- Michelle L. Engle
- Marsico Lung Institute, University of North Carolina, Chapel Hill, NC, United States of America
- Curriculum in Genetics and Molecular Biology, University of North Carolina, Chapel Hill, NC, United States of America
| | - Justine N. Monk
- Marsico Lung Institute, University of North Carolina, Chapel Hill, NC, United States of America
- Pathobiology and Translational Science Graduate Program, University of North Carolina, Chapel Hill, NC, United States of America
| | - Corey M. Jania
- Marsico Lung Institute, University of North Carolina, Chapel Hill, NC, United States of America
- Division of Pulmonary Diseases and Critical Care Medicine, University of North Carolina, Chapel Hill, NC, United States of America
- Department of Medicine, University of North Carolina, Chapel Hill, NC, United States of America
| | - Jessica R. Martin
- Marsico Lung Institute, University of North Carolina, Chapel Hill, NC, United States of America
| | - John C. Gomez
- Marsico Lung Institute, University of North Carolina, Chapel Hill, NC, United States of America
| | - Hong Dang
- Marsico Lung Institute, University of North Carolina, Chapel Hill, NC, United States of America
| | - Joel S. Parker
- Department of Genetics, University of North Carolina, Chapel Hill, NC, United States of America
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, United States of America
| | - Claire M. Doerschuk
- Marsico Lung Institute, University of North Carolina, Chapel Hill, NC, United States of America
- Division of Pulmonary Diseases and Critical Care Medicine, University of North Carolina, Chapel Hill, NC, United States of America
- Department of Medicine, University of North Carolina, Chapel Hill, NC, United States of America
| |
Collapse
|
20
|
Lee CH, Shim HE, Song L, Moon HG, Lee K, Yang JE, Song HY, Choi YJ, Choi DS, Jeon J. Efficient and stable radiolabeling of polycyclic aromatic hydrocarbon assemblies: in vivo imaging of diesel exhaust particulates in mice. Chem Commun (Camb) 2019; 55:447-450. [DOI: 10.1039/c8cc08304e] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
As a robust radioanalytical method for tracking carbonaceous particulates in vivo, polycyclic aromatic hydrocarbons from diesel exhaust were labeled with a radioactive-iodine-tagged pyrene analogue.
Collapse
Affiliation(s)
- Chang Heon Lee
- Advanced Radiation Technology Institute
- Korea Atomic Energy Research Institute
- Jeongeup 56212
- Republic of Korea
| | - Ha Eun Shim
- Advanced Radiation Technology Institute
- Korea Atomic Energy Research Institute
- Jeongeup 56212
- Republic of Korea
| | - Lee Song
- Advanced Radiation Technology Institute
- Korea Atomic Energy Research Institute
- Jeongeup 56212
- Republic of Korea
| | - Hi Gyu Moon
- Jeonbuk Department of Inhalation Research
- Korea Institute of Toxicology
- Jeongeup 56212
- Republic of Korea
| | - Kyuhong Lee
- Jeonbuk Department of Inhalation Research
- Korea Institute of Toxicology
- Jeongeup 56212
- Republic of Korea
| | - Jung Eun Yang
- World Institute of Kimchi
- Gwangju 61755
- Republic of Korea
| | - Ha Yeon Song
- Advanced Radiation Technology Institute
- Korea Atomic Energy Research Institute
- Jeongeup 56212
- Republic of Korea
| | - Yong Jun Choi
- School of Environmental Engineering
- University of Seoul
- Seoul 02504
- Republic of Korea
| | - Dae Seong Choi
- Advanced Radiation Technology Institute
- Korea Atomic Energy Research Institute
- Jeongeup 56212
- Republic of Korea
| | - Jongho Jeon
- Advanced Radiation Technology Institute
- Korea Atomic Energy Research Institute
- Jeongeup 56212
- Republic of Korea
- Radiation Biotechnology and Applied Radioisotope Science
| |
Collapse
|
21
|
Brzóska K, Bartłomiejczyk T, Sochanowicz B, Cymerman M, Grudny J, Kołakowski J, Kruszewski M, Śliwiński P, Roszkowski-Śliż K, Kapka-Skrzypczak L. Carcinogenesis-related changes in iron metabolism in chronic obstructive pulmonary disease subjects with lung cancer. Oncol Lett 2018; 16:6831-6837. [PMID: 30405827 DOI: 10.3892/ol.2018.9459] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Accepted: 08/10/2018] [Indexed: 12/17/2022] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is often accompanied by lung cancer. In our previous work, it was observed that matrix metalloproteinase-3 and haptoglobin (HP) polymorphisms were potential markers of enhanced susceptibility to lung cancer development among male COPD subjects. Here, results are reported on blood serum levels of several proteins involved in iron metabolism, inflammation and the oxidative stress response compared between the same groups of subjects. The blood serum levels of tumor necrosis factor α (TNFα), transferrin, hepcidin, ferritin, soluble transferrin receptor and 8-oxo-2'-deoxyguanosine were compared, as well as total iron-binding capacity (TIBC) and ceruloplasmin ferroxidase activity in two groups of subjects: Male COPD patients (54 subjects) and male COPD patients diagnosed with lung cancer (53 subjects). Statistically significant differences were identified between the two groups in transferrin and TNFα levels, as well as in TIBC; all three parameters were lower in the group consisting of COPD patients diagnosed with lung cancer (P<0.01). It was also revealed that HP genotype 1/2 was concomitant with low transferrin blood level in subjects with COPD; this apparent dependence was absent in the COPD + cancer subjects. The results indicate a role of iron metabolism in the susceptibility to lung cancer in COPD-affected subjects. They also emphasize the importance of individual capacity for an effective response to oxidative stress during the pathogenic process as HP is a plasma protein that binds free hemoglobin and its polymorphism results in proteins with altered hemoglobin-binding capacity and different antioxidant and iron-recycling functions.
Collapse
Affiliation(s)
- Kamil Brzóska
- Institute of Nuclear Chemistry and Technology, Centre for Radiobiology and Biological Dosimetry, 03-195 Warsaw, Poland
| | - Teresa Bartłomiejczyk
- Institute of Nuclear Chemistry and Technology, Centre for Radiobiology and Biological Dosimetry, 03-195 Warsaw, Poland
| | - Barbara Sochanowicz
- Institute of Nuclear Chemistry and Technology, Centre for Radiobiology and Biological Dosimetry, 03-195 Warsaw, Poland
| | - Magdalena Cymerman
- Institute of Nuclear Chemistry and Technology, Centre for Radiobiology and Biological Dosimetry, 03-195 Warsaw, Poland
| | - Jacek Grudny
- Institute of Tuberculosis and Lung Diseases, Third Department of Lung Diseases, 01-138 Warsaw, Poland
| | - Jacek Kołakowski
- Institute of Tuberculosis and Lung Diseases, Department of Diagnosis and Treatment of Respiratory Insufficiency, 01-138 Warsaw, Poland
| | - Marcin Kruszewski
- Institute of Nuclear Chemistry and Technology, Centre for Radiobiology and Biological Dosimetry, 03-195 Warsaw, Poland.,Institute of Rural Health, Department of Molecular Biology and Translational Research, 20-090 Lublin, Poland.,University of Information Technology and Management, Faculty of Medicine, Department of Medical Biology and Translational Research, 35-225 Rzeszów, Poland
| | - Paweł Śliwiński
- Institute of Tuberculosis and Lung Diseases, Department of Diagnosis and Treatment of Respiratory Insufficiency, 01-138 Warsaw, Poland
| | - Kazimierz Roszkowski-Śliż
- Institute of Tuberculosis and Lung Diseases, Third Department of Lung Diseases, 01-138 Warsaw, Poland
| | - Lucyna Kapka-Skrzypczak
- Institute of Rural Health, Department of Molecular Biology and Translational Research, 20-090 Lublin, Poland.,University of Information Technology and Management, Faculty of Medicine, Department of Medical Biology and Translational Research, 35-225 Rzeszów, Poland
| |
Collapse
|
22
|
Jiang B, Guan Y, Shen HJ, Zhang LH, Jiang JX, Dong XW, Shen HH, Xie QM. Akt/PKB signaling regulates cigarette smoke-induced pulmonary epithelial-mesenchymal transition. Lung Cancer 2018; 122:44-53. [DOI: 10.1016/j.lungcan.2018.05.019] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Revised: 05/19/2018] [Accepted: 05/21/2018] [Indexed: 01/31/2023]
|
23
|
Bronte G, Bravaccini S, Bronte E, Burgio MA, Rolfo C, Delmonte A, Crinò L. Epithelial-to-mesenchymal transition in the context of epidermal growth factor receptor inhibition in non-small-cell lung cancer. Biol Rev Camb Philos Soc 2018; 93:1735-1746. [PMID: 29671943 DOI: 10.1111/brv.12416] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Revised: 03/20/2018] [Accepted: 03/23/2018] [Indexed: 12/12/2022]
Abstract
The identification of oncogenic driver mutations in non-small-cell lung cancer (NSCLC) has led to the development of targeted drugs. Tyrosine kinase inhibitors (TKIs) directed against the epidermal growth factor receptor (EGFR) target lung tumours bearing EGFR-activating mutations. This new therapeutic strategy has greatly improved tumour response rates. However, drug resistance invariably occurs during TKI-based treatment. Epithelial-to-mesenchymal transition (EMT) is one of the resistance mechanisms identified in EGFR-mutated NSCLC treated with TKIs. In this review we gather together the most important findings on this phenomenon in relation to cancer stem cells and cancer epigenetics. We also outline the correlation between the effects of stromal factors from the microenvironment, the transcription factors activated, the epigenetic changes in chromatin, and the evolution of cellular behaviour. Notably, EMT has already been shown to be the link between benign lung diseases such as chronic obstructive pulmonary disease and lung carcinogenesis. The various mechanisms of acquired resistance to EGFR-TKIs are also briefly described to provide background information on EMT. Our extensive review of the scientific literature serves to highlight the cellular and molecular events that lead to the onset of EMT in NSCLC cells treated with EGFR-TKIs. Finally, we put forward a hypothesis to explain why, in some cases, EMT rather than other known mechanisms is involved in resistance to TKIs.
Collapse
Affiliation(s)
- Giuseppe Bronte
- Department of Medical Oncology, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Via P. Maroncelli 40, 47014, Meldola, Italy
| | - Sara Bravaccini
- Biosciences Laboratory, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Enrico Bronte
- Department of Surgical, Oncological and Oral Sciences, University of Palermo, Palermo, Italy
| | - Marco Angelo Burgio
- Department of Medical Oncology, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Via P. Maroncelli 40, 47014, Meldola, Italy
| | - Christian Rolfo
- Phase I Early Clinical Trials Unit, Department of Oncology, Universitair Ziekenhuis Antwerpen, Edegem, Belgium
| | - Angelo Delmonte
- Department of Medical Oncology, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Via P. Maroncelli 40, 47014, Meldola, Italy
| | - Lucio Crinò
- Department of Medical Oncology, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Via P. Maroncelli 40, 47014, Meldola, Italy
| |
Collapse
|
24
|
Sharma M, Levenson C, Browning JC, Becker EM, Clements I, Castella P, Cox ME. East Indian Sandalwood Oil Is a Phosphodiesterase Inhibitor: A New Therapeutic Option in the Treatment of Inflammatory Skin Disease. Front Pharmacol 2018; 9:200. [PMID: 29593534 PMCID: PMC5854648 DOI: 10.3389/fphar.2018.00200] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2017] [Accepted: 02/22/2018] [Indexed: 01/01/2023] Open
Abstract
Cyclic adenosine monophosphate phosphodiesterases (PDEs) regulate pro-inflammatory cytokine production. One isoform, PDE4, is overactive in chronic relapsing inflammatory skin diseases: psoriasis and eczema/atopic dermatitis, and in several cancers. East Indian sandalwood oil (EISO) has significant anti-inflammatory properties. Here, we report that 75% of pediatric eczema/atopic dermatitis patients treated with topical EISO formulations achieved a >50% reduction in their Eczema Area and Severity Index score. EISO treatment of a psoriasis model reduced PDE4 expression and reversed histopathology. EISO directly inhibited PDE enzymatic activity in vitro. In lipopolysaccharide-stimulated human dermal fibroblast, BEAS-2B, A549, and THP-1 cells, EISO suppressed total cellular PDE activity, PDE4, and 7 transcript levels, nuclear factor kappa B (NF-κB) activation, and pro-inflammatory cytokines/chemokine production. These results suggest that EISO anti-inflammatory activity is mediated through suppressing PDE activity, thus facilitating cAMP-regulated inhibition of NF-κB and indicate EISO as an attractive natural therapeutic for chronic and acute inflammatory disorders.
Collapse
Affiliation(s)
- Manju Sharma
- The Vancouver Prostate Centre, Vancouver Coastal Health Research Institute, Vancouver, BC, Canada
| | - Corey Levenson
- Santalis Pharmaceuticals, Inc., San Antonio, TX, United States
| | - John C Browning
- Texas Dermatology and Laser Specialists, San Antonio, TX, United States
| | - Emily M Becker
- Texas Dermatology and Laser Specialists, San Antonio, TX, United States
| | - Ian Clements
- Santalis Pharmaceuticals, Inc., San Antonio, TX, United States
| | - Paul Castella
- Santalis Pharmaceuticals, Inc., San Antonio, TX, United States
| | - Michael E Cox
- The Vancouver Prostate Centre, Vancouver Coastal Health Research Institute, Vancouver, BC, Canada.,Department of Urologic Sciences, The University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
25
|
Liu JC, Yang TY, Hsu YP, Hao WR, Kao PF, Sung LC, Chen CC, Wu SY. Statins dose-dependently exert a chemopreventive effect against lung cancer in COPD patients: a population-based cohort study. Oncotarget 2018; 7:59618-59629. [PMID: 27517752 PMCID: PMC5312335 DOI: 10.18632/oncotarget.11162] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2016] [Accepted: 07/09/2016] [Indexed: 02/03/2023] Open
Abstract
Purpose Chronic obstructive pulmonary disease (COPD) is associated with increased lung cancer risk. We evaluated the association of statin use with lung cancer risk in COPD patients and identified which statins possess the highest chemopreventive potential. Results After adjustment for age, sex, CCI, diabetes, hypertension, dyslipidemia, urbanization level, and monthly income according to propensity scores, lung cancer risk in the statin users was lower than that in the statin nonusers (adjusted hazard ratio [aHR] = 0.37). Of the individual statins, lovastatin and fluvastatin did not reduce lung cancer risk significantly. By contrast, lung cancer risk in patients using rosuvastatin, simvastatin, atorvastatin, and pravastatin was significantly lower than that in statin nonusers (aHRs = 0.41, 0.44, 0.52, and 0.58, respectively). Statins dose-dependently reduced lung cancer risk in all subgroups and the main model with additional covariates (nonstatin drug use). MATERIALS AND METHODS The study cohort comprised all patients diagnosed with COPD at health care facilities in Taiwan (n = 116,017) between January 1, 2001 and December 31, 2012. Our final study cohort comprised 43,802 COPD patients: 10,086 used statins, whereas 33,716 did not. Patients were followed up to assess lung cancer risk or protective factors. In addition, we also considered demographic characteristics, namely age, sex, comorbidities (diabetes, hypertension, dyslipidemia, and Charlson comorbidity index [CCI]), urbanization level, monthly income, and nonstatin drug use. The index date of statin use was the COPD confirmation date. To examine the dose–response relationship, we categorized statin use into four groups in each cohort: < 28, 28–90, 91–365, and > 365 cumulative defined daily doses (cDDDs). Patients receiving < 28 cDDDs were defined as nonstatin users. Conclusions Statins dose-dependently exert a significant chemopreventive effect against lung cancer in COPD patients. Rosuvastatin, simvastatin, and atorvastatin exhibited the highest chemopreventive potential.
Collapse
Affiliation(s)
- Ju-Chi Liu
- Division of Cardiovascular Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan.,Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Tsung-Yeh Yang
- Division of Cardiovascular Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
| | - Yi-Ping Hsu
- Division of Cardiovascular Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
| | - Wen-Rui Hao
- Division of Cardiovascular Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
| | - Pai-Feng Kao
- Division of Cardiovascular Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan.,Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Li-Chin Sung
- Division of Cardiovascular Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan.,Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Chun-Chao Chen
- Division of Cardiovascular Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
| | - Szu-Yuan Wu
- Institute of Toxicology, College of Medicine, National Taiwan University, Taipei, Taiwan.,Department of Radiation Oncology, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan.,Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Department of Biotechnology, Hungkuang University, Taichung, Taiwan
| |
Collapse
|
26
|
β-catenin, Twist and Snail: Transcriptional regulation of EMT in smokers and COPD, and relation to airflow obstruction. Sci Rep 2017; 7:10832. [PMID: 28883453 PMCID: PMC5589881 DOI: 10.1038/s41598-017-11375-x] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Accepted: 08/23/2017] [Indexed: 12/20/2022] Open
Abstract
COPD is characterised by poorly reversible airflow obstruction usually due to cigarette smoking. The transcription factor clusters of β-catenin/Snail1/Twist has been implicated in the process of epithelial mesenchymal transition (EMT), an intermediate between smoking and airway fibrosis, and indeed lung cancer. We have investigated expression of these transcription factors and their "cellular localization" in bronchoscopic airway biopsies from patients with COPD, and in smoking and non-smoking controls. An immune-histochemical study compared cellular protein expression of β-catenin, Snail1 and Twist, in these subject groups in 3 large airways compartment: epithelium (basal region), reticular basement membrane (Rbm) and underlying lamina propria (LP). β-catenin and Snail1 expression was generally high in all subjects throughout the airway wall with marked cytoplasmic to nuclear shift in COPD (P < 0.01). Twist expression was generalised in the epithelium in normal but become more basal and nuclear with smoking (P < 0.05). In addition, β-catenin and Snail1 expression, and to lesser extent of Twist, was related to airflow obstruction and to expression of a canonical EMT biomarker (S100A4). The β-catenin-Snail1-Twist transcription factor cluster is up-regulated and nuclear translocated in smokers and COPD, and their expression is closely related to both EMT activity and airway obstruction.
Collapse
|
27
|
Wang T, Liu Y, Zou JF, Cheng ZS. Interleukin-17 induces human alveolar epithelial to mesenchymal cell transition via the TGF-β1 mediated Smad2/3 and ERK1/2 activation. PLoS One 2017; 12:e0183972. [PMID: 28873461 PMCID: PMC5584923 DOI: 10.1371/journal.pone.0183972] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Accepted: 08/15/2017] [Indexed: 12/31/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a chronic and usually progressive lung disease and the epithelial-mesenchymal transition (EMT) may play an important role in the pathogenesis of pulmonary fibrosis. IL-17 is a proinflammatory cytokine which promotes EMT profiles in lung inflammatory diseases. In this study, we investigated the effect of IL-17 on EMT in alveolar epithelial cell line A549 and the role of TGFβ1-Smad and ERK signaling pathways in the process. Morphological observation on the cells was performed under inverted microscope. The mRNA and protein expressions of E-cad and α-SMA were detected by quantitative RT-PCR and western blotting. The mRNA and protein expressions of TGF-β1 were analyzed via quantitative RT-PCR and ELISA. Expressions of Smad2/3, p-Smad2/3, ERK1/2, p-ERK1/2 and p-JNK were examined by western blotting. The results indicated that IL-17 can induce A549 cells to undergo morphological changes and phenotypic markers changes, such as down-regulated E-cad expression and up-regulated α-SMA expression. Additionally, IL-17 enhanced TGF-β1 expression and stimulated Smad2/3 and ERK1/2 phosphorylation in A549 cells. However, there were no significant differences in the expression of phosphorylated JNK in A549 cells with or without IL-17 treatment. SB431542 or U0126 treated cells showed inhibited morphological changes and phenotypic markers expression, such as up-regulated E-cad expression and down-regulated α-SMA expression. In summary, our results suggest that IL-17 can induce A549 alveolar epithelial cells to undergo EMT via the TGF-β1 mediated Smad2/3 and ERK1/2 activation.
Collapse
Affiliation(s)
- Ting Wang
- Department of Respiratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Yuan Liu
- Department of Respiratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Jing-Feng Zou
- Department of Respiratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Zhen-Shun Cheng
- Department of Respiratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
- * E-mail:
| |
Collapse
|
28
|
Spyratos D, Papadaki E, Lampaki S, Kontakiotis T. Chronic obstructive pulmonary disease in patients with lung cancer: prevalence, impact and management challenges. LUNG CANCER-TARGETS AND THERAPY 2017; 8:101-107. [PMID: 28860884 PMCID: PMC5558876 DOI: 10.2147/lctt.s117178] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Chronic obstructive pulmonary disease (COPD) and lung cancer share a common etiological factor (cigarette smoking) and usually coexist in everyday clinical practice. The prevalence of COPD among newly diagnosed patients with lung cancer sometimes exceeds 50%. COPD is an independent risk factor (2-4 times higher than non-COPD subjects) for lung cancer development. The presence of emphysema in addition to other factors (e.g., smoking history, age) could be incorporated into risk scores in order to define the most appropriate target group for lung cancer screening using low-dose computed tomography. Clinical management of patients with coexistence of COPD and lung cancer requires a multidisciplinary oncology board that includes a pulmonologist. Detailed evaluation (lung function tests, cardiopulmonary exercise test) and management (inhaled drugs, smoking cessation, pulmonary rehabilitation) of COPD should be taken into account for lung cancer treatment (surgical approach, radiotherapy).
Collapse
Affiliation(s)
- Dionisios Spyratos
- Pulmonary Department, Lung Cancer Oncology Unit, Aristotle University of Thessaloniki, G. Papanicolaou Hospital, Thessaloniki, Greece
| | - Eleni Papadaki
- Pulmonary Department, Lung Cancer Oncology Unit, Aristotle University of Thessaloniki, G. Papanicolaou Hospital, Thessaloniki, Greece
| | - Sofia Lampaki
- Pulmonary Department, Lung Cancer Oncology Unit, Aristotle University of Thessaloniki, G. Papanicolaou Hospital, Thessaloniki, Greece
| | - Theodoros Kontakiotis
- Pulmonary Department, Lung Cancer Oncology Unit, Aristotle University of Thessaloniki, G. Papanicolaou Hospital, Thessaloniki, Greece
| |
Collapse
|
29
|
Shields PG, Berman M, Brasky TM, Freudenheim JL, Mathe E, McElroy JP, Song MA, Wewers MD. A Review of Pulmonary Toxicity of Electronic Cigarettes in the Context of Smoking: A Focus on Inflammation. Cancer Epidemiol Biomarkers Prev 2017; 26:1175-1191. [PMID: 28642230 PMCID: PMC5614602 DOI: 10.1158/1055-9965.epi-17-0358] [Citation(s) in RCA: 81] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2017] [Revised: 05/22/2017] [Accepted: 05/24/2017] [Indexed: 12/30/2022] Open
Abstract
The use of electronic cigarettes (e-cigs) is increasing rapidly, but their effects on lung toxicity are largely unknown. Smoking is a well-established cause of lung cancer and respiratory disease, in part through inflammation. It is plausible that e-cig use might affect similar inflammatory pathways. E-cigs are used by some smokers as an aid for quitting or smoking reduction, and by never smokers (e.g., adolescents and young adults). The relative effects for impacting disease risk may differ for these groups. Cell culture and experimental animal data indicate that e-cigs have the potential for inducing inflammation, albeit much less than smoking. Human studies show that e-cig use in smokers is associated with substantial reductions in blood or urinary biomarkers of tobacco toxicants when completely switching and somewhat for dual use. However, the extent to which these biomarkers are surrogates for potential lung toxicity remains unclear. The FDA now has regulatory authority over e-cigs and can regulate product and e-liquid design features, such as nicotine content and delivery, voltage, e-liquid formulations, and flavors. All of these factors may impact pulmonary toxicity. This review summarizes current data on pulmonary inflammation related to both smoking and e-cig use, with a focus on human lung biomarkers. Cancer Epidemiol Biomarkers Prev; 26(8); 1175-91. ©2017 AACR.
Collapse
Affiliation(s)
- Peter G Shields
- Comprehensive Cancer Center, The Ohio State University and James Cancer Hospital, and College of Medicine, Columbus, Ohio.
| | - Micah Berman
- Comprehensive Cancer Center, The Ohio State University and James Cancer Hospital, and College of Public Health, Ohio
| | - Theodore M Brasky
- Comprehensive Cancer Center, The Ohio State University and James Cancer Hospital, and College of Medicine, Columbus, Ohio
| | - Jo L Freudenheim
- Department of Epidemiology and Environmental Health, School of Public Health and Health Professions, University at Buffalo, Buffalo, New York
| | - Ewy Mathe
- Department of Biomedical Informatics, The Ohio State University, Columbus, Ohio
| | - Joseph P McElroy
- Center for Biostatistics, Department of Biomedical Informatics, The Ohio State University, Columbus, Ohio
| | - Min-Ae Song
- Comprehensive Cancer Center, The Ohio State University and James Cancer Hospital, and College of Medicine, Columbus, Ohio
| | - Mark D Wewers
- Department of Internal Medicine, The Ohio State University, Columbus, Ohio
| |
Collapse
|
30
|
Jolly MK, Ward C, Eapen MS, Myers S, Hallgren O, Levine H, Sohal SS. Epithelial-mesenchymal transition, a spectrum of states: Role in lung development, homeostasis, and disease. Dev Dyn 2017. [DOI: 10.1002/dvdy.24541] [Citation(s) in RCA: 137] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Affiliation(s)
- Mohit Kumar Jolly
- Center for Theoretical Biological Physics; Rice University; Houston Texas
| | - Chris Ward
- Institute of Cellular Medicine; Newcastle University; Newcastle upon Tyne United Kingdom
| | - Mathew Suji Eapen
- School of Health Sciences; Faculty of Health, University of Tasmania, Launceston, University of Tasmania; Hobart Tasmania Australia
- NHMRC Centre of Research Excellence for Chronic Respiratory Disease; University of Tasmania; Hobart Tasmania Australia
| | - Stephen Myers
- School of Health Sciences; Faculty of Health, University of Tasmania, Launceston, University of Tasmania; Hobart Tasmania Australia
| | - Oskar Hallgren
- Department of Experimental Medical Sciences; Department of Respiratory Medicine and Allergology, Lund University; Sweden
| | - Herbert Levine
- Center for Theoretical Biological Physics; Rice University; Houston Texas
| | - Sukhwinder Singh Sohal
- School of Health Sciences; Faculty of Health, University of Tasmania, Launceston, University of Tasmania; Hobart Tasmania Australia
- NHMRC Centre of Research Excellence for Chronic Respiratory Disease; University of Tasmania; Hobart Tasmania Australia
| |
Collapse
|
31
|
Oke O, Azzopardi D, Corke S, Hewitt K, Carr T, Cockcroft N, Foss-Smith G, Taylor M, Lowe F. Assessment of AcuteIn VitroHuman Cellular Responses to Smoke Extracts from a Reduced Toxicant Prototype Cigarette. ACTA ACUST UNITED AC 2017. [DOI: 10.1089/aivt.2016.0038] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Oluwatobiloba Oke
- Research and Development, British American Tobacco, Southampton, United Kingdom
| | - David Azzopardi
- Research and Development, British American Tobacco, Southampton, United Kingdom
| | - Sarah Corke
- Research and Development, British American Tobacco, Southampton, United Kingdom
| | - Katherine Hewitt
- Research and Development, British American Tobacco, Southampton, United Kingdom
| | - Tony Carr
- Research and Development, British American Tobacco, Southampton, United Kingdom
| | - Natalia Cockcroft
- Research and Development, British American Tobacco, Southampton, United Kingdom
| | - Geoff Foss-Smith
- Research and Development, British American Tobacco, Southampton, United Kingdom
| | - Mark Taylor
- Research and Development, British American Tobacco, Southampton, United Kingdom
| | - Frazer Lowe
- Research and Development, British American Tobacco, Southampton, United Kingdom
| |
Collapse
|
32
|
Integrative analysis of genomic sequencing data reveals higher prevalence of LRP1B mutations in lung adenocarcinoma patients with COPD. Sci Rep 2017; 7:2121. [PMID: 28522810 PMCID: PMC5437030 DOI: 10.1038/s41598-017-02405-9] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Accepted: 04/25/2017] [Indexed: 12/31/2022] Open
Abstract
Both chronic Obstruction Pulmonary Disease (COPD) and lung cancer are leading causes of death globally. Although COPD and lung cancer coexist frequently, it is unknown whether lung cancer patients with COPD harbor distinct genomic characteristics compared to those without COPD. In this study, we retrospectively analyzed genomic sequencing data from 272 patients with lung adenocarcinoma (LUAD) and compared the genetic alterations in LUAD patients with and without COPD. Integrative analysis of whole-genome and exome sequencing data revealed that COPD and non-COPD groups showed high concordance in mutational burden and spectra. Notably, we also found that EGFR mutations were more prevalent in LUAD patients without COPD, whereas mutated LRP1B was more frequently observed in LUAD patients with COPD. In addition, multi-variable analysis with logistic regression demonstrated that mutation of LRP1B was a predictive marker for the presence of COPD in the patients with LUAD. Our analysis demonstrated for the first time the high concordance in genomic alterations between the tumors from LUAD patients with and without COPD. We also identified higher prevalence of LRP1B among the LUAD patients with COPD, which might help understand the underlying mechanisms which link COPD and lung cancer.
Collapse
|
33
|
Mahmood MQ, Reid D, Ward C, Muller HK, Knight DA, Sohal SS, Walters EH. Transforming growth factor (TGF) β 1 and Smad signalling pathways: A likely key to EMT-associated COPD pathogenesis. Respirology 2016; 22:133-140. [PMID: 27614607 DOI: 10.1111/resp.12882] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2016] [Revised: 06/06/2016] [Accepted: 06/22/2016] [Indexed: 12/18/2022]
Abstract
BACKGROUND AND OBJECTIVE COPD is characterized by poorly reversible airflow obstruction usually due to cigarette smoking. Transforming growth factor (TGF)-β1 has been implicated in the pathogenesis of COPD, and in particular a process called epithelial mesenchymal transition (EMT), which may well be an intermediatory between smoking and both airway fibrosis and lung cancer. The downstream classical or 'canonical' TGF-β1 pathway is via the phosphorylated (p) Smad transcription factor system. METHODS We have investigated TGF-β1 expression and its 'pSmad fingerprint' in bronchoscopic airway biopsies from patients with COPD, and in smoking and non-smoking controls. A cross-sectional immunohistochemical study compared TGF-β1 and pSmad 2, 3 (excitatory) and 7 (inhibitory) expression in cells and blood vessels of three compartments of large airways: epithelium (especially the basal region), reticular basement membrane (Rbm) and underlying lamina propria (LP). RESULTS TGF-β1 expression was generally higher in COPD subjects throughout the airway wall (P < 0.01), while pSmad 2/3 expression was associated with smoking especially in current smoking COPD (P < 0.05). Expression of inhibitory pSmad 7 was also prominently reduced in patients with COPD in contrast to smokers and controls (P < 0.01). In addition, pSmad, but not TGF-β1 expression, was related to airflow obstruction and a canonical EMT biomarker (S100 A4) expression. CONCLUSION Activation of the Smad pathway in the airways is linked to EMT activity and loss of lung function. The disconnection between TGF-β1 and pSmad in terms of relationships to EMT activity and lung function suggests that factors other than or in addition to TGF-β1 are driving the process.
Collapse
Affiliation(s)
- Malik Q Mahmood
- NHMRC Centre of Research Excellence for Chronic Respiratory Disease, School of Medicine, University of Tasmania, Hobart, Tasmania, Australia
| | - David Reid
- Queensland Institute of Medical Research, Iron Metabolism Laboratory, Brisbane, Queensland, Australia
| | - Chris Ward
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, UK
| | - Hans K Muller
- NHMRC Centre of Research Excellence for Chronic Respiratory Disease, School of Medicine, University of Tasmania, Hobart, Tasmania, Australia
| | - Darryl A Knight
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Newcastle, New South Wales, Australia
| | - Sukhwinder S Sohal
- NHMRC Centre of Research Excellence for Chronic Respiratory Disease, School of Medicine, University of Tasmania, Hobart, Tasmania, Australia.,Faculty of Health, School of Health Sciences, University of Tasmania, Launceston, Tasmania, Australia
| | - Eugene H Walters
- NHMRC Centre of Research Excellence for Chronic Respiratory Disease, School of Medicine, University of Tasmania, Hobart, Tasmania, Australia
| |
Collapse
|
34
|
Heijink IH, de Bruin HG, Dennebos R, Jonker MR, Noordhoek JA, Brandsma CA, van den Berge M, Postma DS. Cigarette smoke-induced epithelial expression of WNT-5B: implications for COPD. Eur Respir J 2016; 48:504-15. [PMID: 27126693 DOI: 10.1183/13993003.01541-2015] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Accepted: 03/10/2016] [Indexed: 02/04/2023]
Abstract
Wingless/integrase-1 (WNT) signalling is associated with lung inflammation and repair, but its role in chronic obstructive pulmonary disease (COPD) pathogenesis is unclear. We investigated whether cigarette smoke-induced dysregulation of WNT-5B contributes to airway remodelling in COPD.We analysed WNT-5B protein expression in the lung tissue of COPD patients and (non)smoking controls, and investigated the effects of cigarette smoke exposure on WNT-5B expression in COPD and control-derived primary bronchial epithelial cells (PBECs). Additionally, we studied downstream effects of WNT-5B on remodelling related genes fibronectin, matrix metalloproteinase (MMP)-2, MMP-9 and SnaiI in BEAS-2B and air-liquid interface (ALI)-cultured PBECs.We observed that airway epithelial WNT-5B expression is significantly higher in lung tissue from COPD patients than controls. Cigarette smoke extract significantly increased mRNA expression of WNT-5B in COPD, but not control-derived PBECs. Exogenously added WNT-5B augmented the expression of remodelling related genes in BEAS-2B cells, which was mediated by transforming growth factor (TGF)-β/Smad3 signalling. In addition, WNT-5B upregulated the expression of these genes in ALI-cultured PBECs, particularly PBECs from COPD patients.Together, our results provide evidence that exaggerated WNT-5B expression upon cigarette smoke exposure in the bronchial epithelium of COPD patients leads to TGF-β/Smad3-dependent expression of genes related to airway remodelling.
Collapse
Affiliation(s)
- Irene H Heijink
- University of Groningen, University Medical Center Groningen, Dept of Pathology and Medical Biology, Groningen, The Netherlands University of Groningen, University Medical Center Groningen, Dept of Pulmonology, Groningen, The Netherlands University of Groningen, University Medical Center Groningen GRIAC Research Institute, Groningen, The Netherlands
| | - Harold G de Bruin
- University of Groningen, University Medical Center Groningen, Dept of Pathology and Medical Biology, Groningen, The Netherlands
| | - Robin Dennebos
- University of Groningen, University Medical Center Groningen, Dept of Pathology and Medical Biology, Groningen, The Netherlands
| | - Marnix R Jonker
- University of Groningen, University Medical Center Groningen, Dept of Pathology and Medical Biology, Groningen, The Netherlands
| | - Jacobien A Noordhoek
- University of Groningen, University Medical Center Groningen, Dept of Pathology and Medical Biology, Groningen, The Netherlands
| | - 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 GRIAC Research Institute, Groningen, The Netherlands
| | - Maarten van den Berge
- University of Groningen, University Medical Center Groningen, Dept of Pulmonology, Groningen, The Netherlands University of Groningen, University Medical Center Groningen GRIAC Research Institute, Groningen, The Netherlands
| | - Dirkje S Postma
- University of Groningen, University Medical Center Groningen, Dept of Pulmonology, Groningen, The Netherlands University of Groningen, University Medical Center Groningen GRIAC Research Institute, Groningen, The Netherlands
| |
Collapse
|
35
|
Steiner S, Bisig C, Petri-Fink A, Rothen-Rutishauser B. Diesel exhaust: current knowledge of adverse effects and underlying cellular mechanisms. Arch Toxicol 2016; 90:1541-53. [PMID: 27165416 PMCID: PMC4894930 DOI: 10.1007/s00204-016-1736-5] [Citation(s) in RCA: 156] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2016] [Accepted: 04/28/2016] [Indexed: 12/03/2022]
Abstract
Diesel engine emissions are among the most prevalent anthropogenic pollutants worldwide, and with the growing popularity of diesel-fueled engines in the private transportation sector, they are becoming increasingly widespread in densely populated urban regions. However, a large number of toxicological studies clearly show that diesel engine emissions profoundly affect human health. Thus the interest in the molecular and cellular mechanisms underlying these effects is large, especially concerning the nature of the components of diesel exhaust responsible for the effects and how they could be eliminated from the exhaust. This review describes the fundamental properties of diesel exhaust as well as the human respiratory tract and concludes that adverse health effects of diesel exhaust not only emerge from its chemical composition, but also from the interplay between its physical properties, the physiological and cellular properties, and function of the human respiratory tract. Furthermore, the primary molecular and cellular mechanisms triggered by diesel exhaust exposure, as well as the fundamentals of the methods for toxicological testing of diesel exhaust toxicity, are described. The key aspects of adverse effects induced by diesel exhaust exposure described herein will be important for regulators to support or ban certain technologies or to legitimate incentives for the development of promising new technologies such as catalytic diesel particle filters.
Collapse
Affiliation(s)
- Sandro Steiner
- Adolphe Merkle Institute, University of Fribourg, Chemin des Verdiers 4, 1700, Fribourg, Switzerland
| | - Christoph Bisig
- Adolphe Merkle Institute, University of Fribourg, Chemin des Verdiers 4, 1700, Fribourg, Switzerland
| | - Alke Petri-Fink
- Adolphe Merkle Institute, University of Fribourg, Chemin des Verdiers 4, 1700, Fribourg, Switzerland
| | | |
Collapse
|
36
|
Sohal SS. Chronic Obstructive Pulmonary Disease (COPD) and Lung Cancer: Epithelial Mesenchymal Transition (EMT), the Missing Link? EBioMedicine 2015; 2:1578-9. [PMID: 26870773 PMCID: PMC4740323 DOI: 10.1016/j.ebiom.2015.10.016] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Accepted: 10/15/2015] [Indexed: 01/24/2023] Open
|
37
|
Mahmood MQ, Sohal SS, Shukla SD, Ward C, Hardikar A, Noor WD, Muller HK, Knight DA, Walters EH. Epithelial mesenchymal transition in smokers: large versus small airways and relation to airflow obstruction. Int J Chron Obstruct Pulmon Dis 2015; 10:1515-24. [PMID: 26346976 PMCID: PMC4531032 DOI: 10.2147/copd.s81032] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Background Small airway fibrosis is the main contributor in airflow obstruction in chronic obstructive pulmonary disease. Epithelial mesenchymal transition (EMT) has been implicated in this process, and in large airways, is associated with angiogenesis, ie, Type-3, which is classically promalignant. Objective In this study we have investigated whether EMT biomarkers are expressed in small airways compared to large airways in subjects with chronic airflow limitation (CAL) and what type of EMT is present on the basis of vascularity. Methods We evaluated epithelial activation, reticular basement membrane fragmentation (core structural EMT marker) and EMT-related mesenchymal biomarkers in small and large airways from resected lung tissue from 18 lung cancer patients with CAL and 9 normal controls. Tissues were immunostained for epidermal growth factor receptor (EGFR; epithelial activation marker), vimentin (mesenchymal marker), and S100A4 (fibroblast epitope). Type-IV collagen was stained to demonstrate vessels. Results There was increased expression of EMT-related markers in CAL small airways compared to controls: EGFR (P<0.001), vimentin (P<0.001), S100A4 (P<0.001), and fragmentation (P<0.001), but this was less than that in large airways. Notably, there was no hypervascularity in small airway reticular basement membrane as in large airways. Epithelial activation and S100A4 expression were related to airflow obstruction. Conclusion EMT is active in small airways, but less so than in large airways in CAL, and may be relevant to the key pathologies of chronic obstructive pulmonary disease, small airway fibrosis, and airway cancers.
Collapse
Affiliation(s)
- Malik Quasir Mahmood
- NHMRC Centre of Research Excellence for Chronic Respiratory Disease and Lung Ageing, School of Medicine, University of Tasmania, Hobart, TAS, Australia
| | - Sukhwinder Singh Sohal
- NHMRC Centre of Research Excellence for Chronic Respiratory Disease and Lung Ageing, School of Medicine, University of Tasmania, Hobart, TAS, Australia ; School of Health Sciences, Faculty of Health, University of Tasmania, Launceston, TAS, Australia
| | - Shakti Dhar Shukla
- NHMRC Centre of Research Excellence for Chronic Respiratory Disease and Lung Ageing, School of Medicine, University of Tasmania, Hobart, TAS, Australia
| | - Chris Ward
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, Tyne and Wear, UK
| | | | - Wan Danial Noor
- NHMRC Centre of Research Excellence for Chronic Respiratory Disease and Lung Ageing, School of Medicine, University of Tasmania, Hobart, TAS, Australia
| | - Hans Konrad Muller
- NHMRC Centre of Research Excellence for Chronic Respiratory Disease and Lung Ageing, School of Medicine, University of Tasmania, Hobart, TAS, Australia
| | - Darryl A Knight
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, NSW, Australia
| | - Eugene Haydn Walters
- NHMRC Centre of Research Excellence for Chronic Respiratory Disease and Lung Ageing, School of Medicine, University of Tasmania, Hobart, TAS, Australia
| |
Collapse
|
38
|
Mikhed Y, Görlach A, Knaus UG, Daiber A. Redox regulation of genome stability by effects on gene expression, epigenetic pathways and DNA damage/repair. Redox Biol 2015; 5:275-289. [PMID: 26079210 PMCID: PMC4475862 DOI: 10.1016/j.redox.2015.05.008] [Citation(s) in RCA: 116] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2015] [Revised: 05/28/2015] [Accepted: 05/29/2015] [Indexed: 02/07/2023] Open
Abstract
Reactive oxygen and nitrogen species (e.g. H2O2, nitric oxide) confer redox regulation of essential cellular signaling pathways such as cell differentiation, proliferation, migration and apoptosis. In addition, classical regulation of gene expression or activity, including gene transcription to RNA followed by translation to the protein level, by transcription factors (e.g. NF-κB, HIF-1α) and mRNA binding proteins (e.g. GAPDH, HuR) is subject to redox regulation. This review will give an update of recent discoveries in this field, and specifically highlight the impact of reactive oxygen and nitrogen species on DNA repair systems that contribute to genomic stability. Emphasis will be placed on the emerging role of redox mechanisms regulating epigenetic pathways (e.g. miRNA, DNA methylation and histone modifications). By providing clinical correlations we discuss how oxidative stress can impact on gene regulation/activity and vise versa, how epigenetic processes, other gene regulatory mechanisms and DNA repair can influence the cellular redox state and contribute or prevent development or progression of disease.
Collapse
Affiliation(s)
- Yuliya Mikhed
- 2nd Medical Clinic, Department of Cardiology, Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Agnes Görlach
- German Heart Center Munich at the Technical University Munich, DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany
| | - Ulla G Knaus
- Conway Institute, School of Medicine, University College Dublin, Dublin, Ireland
| | - Andreas Daiber
- 2nd Medical Clinic, Department of Cardiology, Medical Center of the Johannes Gutenberg University, Mainz, Germany.
| |
Collapse
|
39
|
Yang IA, Holloway JW, Fong KM. Genetic susceptibility to lung cancer and co-morbidities. J Thorac Dis 2014; 5 Suppl 5:S454-62. [PMID: 24163739 DOI: 10.3978/j.issn.2072-1439.2013.08.06] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2013] [Accepted: 08/02/2013] [Indexed: 12/18/2022]
Abstract
Lung cancer is a leading cause of cancer death and disease burden in many countries. Understanding of the biological pathways involved in lung cancer aetiology is required to identify key biomolecules that could be of significant clinical value, either as predictive, prognostic or diagnostic markers, or as targets for the development of novel therapies to treat this disease, in addition to smoking avoidance strategies. Genome-wide association studies (GWAS) have enabled significant progress in the past 5 years in investigating genetic susceptibility to lung cancer. Large scale, multi-cohort GWAS of mainly Caucasian, smoking, populations have identified strong associations for lung cancer mapped to chromosomal regions 15q [nicotinic acetylcholine receptor (nAChR) subunits: CHRNA3, CHRNA5], 5p (TERT-CLPTM1L locus) and 6p (BAT3-MSH5). Some studies in Asian populations of smokers have found similar risk loci, whereas GWAS in never smoking Asian females have identified associations in other chromosomal regions, e.g., 3q (TP63), that are distinct from smoking-related lung cancer risk loci. GWAS of smoking behaviour have identified risk loci for smoking quantity at 15q (similar genes to lung cancer susceptibility: CHRNA3, CHRNA5) and 19q (CYP2A6). Other genes have been mapped for smoking initiation and smoking cessation. In chronic obstructive pulmonary disease (COPD), which is a known risk factor for lung cancer, GWAS in large cohorts have also found CHRNA3 and CHRNA5 single nucleotide polymorphisms (SNPs) mapping at 15q as risk loci, as well as other regions at 4q31 (HHIP), 4q24 (FAM13A) and 5q (HTR4). The overlap in risk loci between lung cancer, smoking behaviour and COPD may be due to the effects of nicotine addiction; however, more work needs to be undertaken to explore the potential direct effects of nicotine and its metabolites in gene-environment interaction in these phenotypes. Goals of future genetic susceptibility studies of lung cancer should focus on refining the strongest risk loci in a wide range of populations with lung cancer, and integrating other clinical and biomarker information, in order to achieve the aim of personalised therapy for lung cancer.
Collapse
Affiliation(s)
- Ian A Yang
- Department of Thoracic Medicine, The Prince Charles Hospital, Brisbane, Australia; ; UQ Thoracic Research Centre, The University of Queensland, Brisbane, Australia
| | | | | |
Collapse
|
40
|
Shen HJ, Sun YH, Zhang SJ, Jiang JX, Dong XW, Jia YL, Shen J, Guan Y, Zhang LH, Li FF, Lin XX, Wu XM, Xie QM, Yan XF. Cigarette smoke-induced alveolar epithelial-mesenchymal transition is mediated by Rac1 activation. BIOCHIMICA ET BIOPHYSICA ACTA 2014; 1840:1838-49. [PMID: 24508121 DOI: 10.1016/j.bbagen.2014.01.033] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/08/2013] [Revised: 01/13/2014] [Accepted: 01/28/2014] [Indexed: 10/25/2022]
Abstract
BACKGROUND Epithelial-mesenchymal transition (EMT) is the major pathophysiological process in lung fibrosis observed in chronic obstructive pulmonary disease (COPD) and lung cancer. Smoking is a risk factor for developing EMT, yet the mechanism remains largely unknown. In this study, we investigated the role of Rac1 in cigarette smoke (CS) induced EMT. METHODS EMT was induced in mice and pulmonary epithelial cells by exposure of CS and cigarette smoke extract (CSE) respectively. RESULTS Treatment of pulmonary epithelial cells with CSE elevated Rac1 expression associated with increased TGF-β1 release. Blocking TGF-β pathway restrained CSE-induced changes in EMT-related markers. Pharmacological inhibition or knockdown of Rac1 decreased the CSE exposure induced TGF-β1 release and ameliorated CSE-induced EMT. In CS-exposed mice, pharmacological inhibition of Rac1 reduced TGF-β1 release and prevented aberrations in expression of EMT markers, suggesting that Rac1 is a critical signaling molecule for induction of CS-stimulated EMT. Furthermore, Rac1 inhibition or knockdown abrogated CSE-induced Smad2 and Akt (PKB, protein kinase B) activation in pulmonary epithelial cells. Inhibition of Smad2, PI3K (phosphatidylinositol 3-kinase) or Akt suppressed CSE-induced changes in epithelial and mesenchymal marker expression. CONCLUSIONS AND GENERAL SIGNIFICANCE Altogether, these data suggest that CS initiates EMT through Rac1/Smad2 and Rac1/PI3K/Akt signaling pathway. Our data provide new insights into the fundamental basis of EMT and suggest a possible new course of therapy for COPD and lung cancer.
Collapse
Affiliation(s)
- Hui-juan Shen
- The Second Affiliated Hospital, Medical College of Zhejiang University, Hangzhou 310009, China; Zhejiang Respiratory Drugs Research Laboratory of State Food and Drug Administration of China, Medical College of Zhejiang University, Hangzhou 310058, China
| | - Yan-hong Sun
- Zhejiang Respiratory Drugs Research Laboratory of State Food and Drug Administration of China, Medical College of Zhejiang University, Hangzhou 310058, China
| | - Shui-juan Zhang
- Pharmacy College of Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Jun-xia Jiang
- The Second Affiliated Hospital, Medical College of Zhejiang University, Hangzhou 310009, China; Zhejiang Respiratory Drugs Research Laboratory of State Food and Drug Administration of China, Medical College of Zhejiang University, Hangzhou 310058, China
| | - Xin-wei Dong
- Zhejiang Respiratory Drugs Research Laboratory of State Food and Drug Administration of China, Medical College of Zhejiang University, Hangzhou 310058, China
| | - Yong-liang Jia
- Zhejiang Respiratory Drugs Research Laboratory of State Food and Drug Administration of China, Medical College of Zhejiang University, Hangzhou 310058, China
| | - Jian Shen
- Zhejiang Respiratory Drugs Research Laboratory of State Food and Drug Administration of China, Medical College of Zhejiang University, Hangzhou 310058, China
| | - Yan Guan
- Zhejiang Respiratory Drugs Research Laboratory of State Food and Drug Administration of China, Medical College of Zhejiang University, Hangzhou 310058, China
| | - Lin-hui Zhang
- Zhejiang Respiratory Drugs Research Laboratory of State Food and Drug Administration of China, Medical College of Zhejiang University, Hangzhou 310058, China
| | - Fen-fen Li
- Zhejiang Respiratory Drugs Research Laboratory of State Food and Drug Administration of China, Medical College of Zhejiang University, Hangzhou 310058, China
| | - Xi-xi Lin
- Zhejiang Respiratory Drugs Research Laboratory of State Food and Drug Administration of China, Medical College of Zhejiang University, Hangzhou 310058, China
| | - Xi-mei Wu
- Zhejiang Respiratory Drugs Research Laboratory of State Food and Drug Administration of China, Medical College of Zhejiang University, Hangzhou 310058, China
| | - Qiang-min Xie
- Zhejiang Respiratory Drugs Research Laboratory of State Food and Drug Administration of China, Medical College of Zhejiang University, Hangzhou 310058, China; Laboratory Animal Center of Zhejiang University, Hangzhou 310058, China.
| | - Xiao-feng Yan
- The Second Affiliated Hospital, Medical College of Zhejiang University, Hangzhou 310009, China.
| |
Collapse
|
41
|
Sohal SS, Soltani A, Reid D, Ward C, Wills KE, Muller HK, Walters EH. A randomized controlled trial of inhaled corticosteroids (ICS) on markers of epithelial-mesenchymal transition (EMT) in large airway samples in COPD: an exploratory proof of concept study. Int J Chron Obstruct Pulmon Dis 2014; 9:533-42. [PMID: 24920891 PMCID: PMC4043431 DOI: 10.2147/copd.s63911] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Background We recently reported that epithelial–mesenchymal transition (EMT) is active in the airways in chronic obstructive pulmonary disease (COPD), suggesting presence of an active profibrotic and promalignant stroma. With no data available on potential treatment effects, we undertook a blinded analysis of inhaled corticosteroids (ICS) effects versus placebo on EMT markers in previously obtained endobronchial biopsies in COPD patients, as a “proof of concept” study. Methods Assessment of the effects of inhaled fluticasone propionate (FP; 500 μg twice daily for 6 months) versus placebo in 34 COPD patients (23 on fluticasone propionate and eleven on placebo). The end points were epidermal growth factor receptor (EGFR; marker of epithelial activation) and the biomarkers of EMT: reticular basement membrane (Rbm) fragmentation (“hallmark” structural marker), matrix metalloproteinase-9 (MMP-9) cell expression, and S100A4 expression in basal epithelial and Rbm cells (mesenchymal transition markers). Results Epithelial activation, “clefts/fragmentation” in the Rbm, and changes in the other biomarkers all regressed on ICS, at or close to conventional levels of statistical significance. From these data, we have been able to nominate primary and secondary end points and develop power calculations that would be applicable to a definitive prospective study. Conclusion Although only a pilot “proof of concept” study, this trial provided strong suggestive support for an anti-EMT effect of ICS in COPD airways. A larger and fully powered prospective study is now indicated as this issue is likely to be extremely important. Such studies may clarify the links between ICS use and better clinical outcomes and protection against lung cancer in COPD.
Collapse
Affiliation(s)
- Sukhwinder Singh Sohal
- National Health and Medical Research Council Centre of Research Excellence for Chronic Respiratory Disease, School of Medicine, University of Tasmania, Hobart, Tasmania, Australia
| | - Amir Soltani
- National Health and Medical Research Council Centre of Research Excellence for Chronic Respiratory Disease, School of Medicine, University of Tasmania, Hobart, Tasmania, Australia
| | - David Reid
- National Health and Medical Research Council Centre of Research Excellence for Chronic Respiratory Disease, School of Medicine, University of Tasmania, Hobart, Tasmania, Australia ; Iron Metabolism Laboratory, Queensland Institute of Medical Research, Brisbane, Queensland, Australia
| | - Chris Ward
- National Health and Medical Research Council Centre of Research Excellence for Chronic Respiratory Disease, School of Medicine, University of Tasmania, Hobart, Tasmania, Australia ; Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, Tyne and Wear, UK
| | - Karen E Wills
- National Health and Medical Research Council Centre of Research Excellence for Chronic Respiratory Disease, School of Medicine, University of Tasmania, Hobart, Tasmania, Australia ; Department of Biostatistics, Menzies Research Institute Tasmania, University of Tasmania, Hobart, Tasmania, Australia
| | - H Konrad Muller
- National Health and Medical Research Council Centre of Research Excellence for Chronic Respiratory Disease, School of Medicine, University of Tasmania, Hobart, Tasmania, Australia
| | - Eugene Haydn Walters
- National Health and Medical Research Council Centre of Research Excellence for Chronic Respiratory Disease, School of Medicine, University of Tasmania, Hobart, Tasmania, Australia
| |
Collapse
|
42
|
Pathological changes in the COPD lung mesenchyme--novel lessons learned from in vitro and in vivo studies. Pulm Pharmacol Ther 2014; 29:121-8. [PMID: 24747433 DOI: 10.1016/j.pupt.2014.04.004] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2013] [Revised: 04/01/2014] [Accepted: 04/08/2014] [Indexed: 12/11/2022]
Abstract
Chronic obstructive pulmonary disease (COPD) is currently the fourth leading cause of death worldwide and, in contrast to the trend for cardiovascular diseases, mortality rates still continue to climb. This increase is in part due to an aging population, being expanded by the "Baby boomer" generation who grew up when smoking rates were at their peak and by people in developing countries living longer. Sadly, there has been a disheartening lack of new therapeutic approaches to counteract the progressive decline in lung function associated with the disease that leads to disability and death. COPD is characterized by irreversible chronic airflow limitation that is caused by emphysematous destruction of lung elastic tissue and/or obstruction in the small airways due to occlusion of their lumen by inflammatory mucus exudates, narrowing and obliteration. These lesions are mainly produced by the response of the tissue to the repetitive inhalational injury inflicted by noxious gases, including cigarette smoke, which involves interaction between infiltrating inflammatory immune cells, resident cells (e.g. epithelial cells and fibroblasts) and the extra cellular matrix. This interaction leads to tissue destruction and airway remodeling with changes in elastin and collagen, such that the epithelial-mesenchymal trophic unit is dysregulated in both the disease pathologies. This review focuses on: 1--novel inflammatory and remodeling factors that are altered in COPD; 2--in vitro and in vivo models to understand the mechanism whereby the extra cellular matrix environment in altered in COPD; and 3--COPD in the context of wound-repair tissue responses, with a focus on the regulation of mesenchymal cell fate and phenotype.
Collapse
|
43
|
Gong WY, Wu JF, Liu BJ, Zhang HY, Cao YX, Sun J, Lv YB, Wu X, Dong JC. Flavonoid components in Scutellaria baicalensis inhibit nicotine-induced proliferation, metastasis and lung cancer-associated inflammation in vitro. Int J Oncol 2014; 44:1561-70. [PMID: 24604573 DOI: 10.3892/ijo.2014.2320] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2013] [Accepted: 02/12/2014] [Indexed: 11/06/2022] Open
Abstract
The objective of the present study was to investigate the therapeutic efficacy of flavonoid components in Scutellaria baicalensis on proliferation, metastasis and lung cancer-associated inflammation during nicotine induction in the A549 and H1299 lung cancer cell lines. After experimental period, augmentation of proliferation was observed, accompanied by marked decrease in apoptotic cells in nicotine-induced lung cancer cells; additionally, nicotine-exposed cells exhibited increased invasive and migratory abilities based on invasion and wound-healing assay. Flavones in Scutellaria, baicalin, baicalein and wogonin significantly counteracted the above deleterious changes. Moreover, assessment of tumor apoptotic and metastatic factors on mRNA levels by quantitative PCR and protein levels by western blotting revealed that these phytochemical treatments effectively negated nicotine-induced upregulated expression of bcl-2, bcl-2/bax ratio, caspase-3, matrix metalloproteinase (MMP)-2 and MMP-9 as well as downregulated expression of bax. Further analysis of inflammatory markers such as tumor necrosis factor (TNF)-α and interleukin (IL)-6 in cell culture supernatant and mRNA and protein expression of nuclear transcription factor-kappaB (NF-κB) and I kappa B-alpha (IκB-α) was carried out to substantiate the anti-inflammatory effect of flavones in Scutellaria in nicotine-exposed lung cancer cells. The therapeutic effects observed in the present study are attributed to the potent potential against proliferation, metastasis and inflammatory microenvironment by flavonoid components in Scutellaria in nicotine-induced lung cancer cells.
Collapse
Affiliation(s)
- Wei-Yi Gong
- Institute of Integrative Medicine, Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai 200041, P.R. China
| | - Jin-Feng Wu
- Institute of Integrative Medicine, Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai 200041, P.R. China
| | - Bao-Jun Liu
- Institute of Integrative Medicine, Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai 200041, P.R. China
| | - Hong-Ying Zhang
- Institute of Integrative Medicine, Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai 200041, P.R. China
| | - Yu-Xue Cao
- Institute of Integrative Medicine, Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai 200041, P.R. China
| | - Jing Sun
- Institute of Integrative Medicine, Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai 200041, P.R. China
| | - Yu-Bao Lv
- Institute of Integrative Medicine, Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai 200041, P.R. China
| | - Xiao Wu
- Institute of Integrative Medicine, Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai 200041, P.R. China
| | - Jing-Cheng Dong
- Institute of Integrative Medicine, Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai 200041, P.R. China
| |
Collapse
|
44
|
Tiep B, Carter R, Zachariah F, Williams AC, Horak D, Barnett M, Dunham R. Oxygen for end-of-life lung cancer care: managing dyspnea and hypoxemia. Expert Rev Respir Med 2014; 7:479-90. [DOI: 10.1586/17476348.2013.816565] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
45
|
Young RP, Hopkins RJ. Update on the potential role of statins in chronic obstructive pulmonary disease and its co-morbidities. Expert Rev Respir Med 2014; 7:533-44. [DOI: 10.1586/17476348.2013.838018] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
|
46
|
Christenson SA, Brandsma CA, Campbell JD, Knight DA, Pechkovsky DV, Hogg JC, Timens W, Postma DS, Lenburg M, Spira A. miR-638 regulates gene expression networks associated with emphysematous lung destruction. Genome Med 2013; 5:114. [PMID: 24380442 PMCID: PMC3971345 DOI: 10.1186/gm519] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2013] [Accepted: 12/20/2013] [Indexed: 12/21/2022] Open
Abstract
Background Chronic obstructive pulmonary disease (COPD) is a heterogeneous disease characterized by varying degrees of emphysematous lung destruction and small airway disease, each with distinct effects on clinical outcomes. There is little known about how microRNAs contribute specifically to the emphysema phenotype. We examined how genome-wide microRNA expression is altered with regional emphysema severity and how these microRNAs regulate disease-associated gene expression networks. Methods We profiled microRNAs in different regions of the lung with varying degrees of emphysema from 6 smokers with COPD and 2 controls (8 regions × 8 lungs = 64 samples). Regional emphysema severity was quantified by mean linear intercept. Whole genome microRNA and gene expression data were integrated in the same samples to build co-expression networks. Candidate microRNAs were perturbed in human lung fibroblasts in order to validate these networks. Results The expression levels of 63 microRNAs (P < 0.05) were altered with regional emphysema. A subset, including miR-638, miR-30c, and miR-181d, had expression levels that were associated with those of their predicted mRNA targets. Genes correlated with these microRNAs were enriched in pathways associated with emphysema pathophysiology (for example, oxidative stress and accelerated aging). Inhibition of miR-638 expression in lung fibroblasts led to modulation of these same emphysema-related pathways. Gene targets of miR-638 in these pathways were amongst those negatively correlated with miR-638 expression in emphysema. Conclusions Our findings demonstrate that microRNAs are altered with regional emphysema severity and modulate disease-associated gene expression networks. Furthermore, miR-638 may regulate gene expression pathways related to the oxidative stress response and aging in emphysematous lung tissue and lung fibroblasts.
Collapse
Affiliation(s)
- Stephanie A Christenson
- Division of Computational Biomedicine, Department of Medicine, Boston University School of Medicine, 72 East Concord Street Boston, MA 02118, USA ; Department of Pulmonary and Critical Care Medicine, University of California, San Francisco, 513 Parnassus Ave, San Francisco, CA 94143, USA
| | - Corry-Anke Brandsma
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 Groningen, Netherlands ; University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD (GRIAC), Hanzeplein 1, 9713 Groningen, Netherlands
| | - Joshua D Campbell
- Division of Computational Biomedicine, Department of Medicine, Boston University School of Medicine, 72 East Concord Street Boston, MA 02118, USA ; Bioinformatics Program, Boston University, 44 Cummington Street Boston, MA 02215, USA
| | - Darryl A Knight
- UBC James Hogg Research Centre, Institute for Heart and Lung Health, St Paul's Hospital and Department of Pathology and Laboratory Medicine, University of British Columbia, 1081 Burrard St Vancouver, BC V6Z 1Y6, Canada ; School of Biomedical Sciences and Pharmacy, University of Newcastle, University Drive Callaghan, New South Wales 2308, Australia
| | - Dmitri V Pechkovsky
- UBC James Hogg Research Centre, Institute for Heart and Lung Health, St Paul's Hospital and Department of Pathology and Laboratory Medicine, University of British Columbia, 1081 Burrard St Vancouver, BC V6Z 1Y6, Canada ; Respiratory Division, Department of Medicine, University of British Columbia, The Jack Bell Research Center, 2660 Oak Street Vancouver, BC V6H 3Z6, Canada
| | - James C Hogg
- UBC James Hogg Research Centre, Institute for Heart and Lung Health, St Paul's Hospital and Department of Pathology and Laboratory Medicine, University of British Columbia, 1081 Burrard St Vancouver, BC V6Z 1Y6, Canada
| | - Wim Timens
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 Groningen, Netherlands ; University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD (GRIAC), Hanzeplein 1, 9713 Groningen, Netherlands
| | - Dirkje S Postma
- University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD (GRIAC), Hanzeplein 1, 9713 Groningen, Netherlands ; Department of Pulmonary Diseases, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 Groningen, Netherlands
| | - Marc Lenburg
- Division of Computational Biomedicine, Department of Medicine, Boston University School of Medicine, 72 East Concord Street Boston, MA 02118, USA ; Bioinformatics Program, Boston University, 44 Cummington Street Boston, MA 02215, USA
| | - Avrum Spira
- Division of Computational Biomedicine, Department of Medicine, Boston University School of Medicine, 72 East Concord Street Boston, MA 02118, USA ; Bioinformatics Program, Boston University, 44 Cummington Street Boston, MA 02215, USA
| |
Collapse
|
47
|
Martinez CH, Kim V, Chen Y, Kazerooni EA, Murray S, Criner GJ, Curtis JL, Regan EA, Wan E, Hersh CP, Silverman EK, Crapo JD, Martinez FJ, Han MK. The clinical impact of non-obstructive chronic bronchitis in current and former smokers. Respir Med 2013; 108:491-9. [PMID: 24280543 DOI: 10.1016/j.rmed.2013.11.003] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2013] [Revised: 11/05/2013] [Accepted: 11/07/2013] [Indexed: 01/11/2023]
Abstract
BACKGROUND As the clinical significance of chronic bronchitis among smokers without airflow obstruction is unclear, we sought to determine morbidity associated with this disorder. METHODS We examined subjects from the COPDGene study and compared those with FEV1/FVC ≥ 0.70, no diagnosis of asthma and chronic bronchitis as defined as a history of cough and phlegm production for ≥ 3 months/year for ≥ 2 years (NCB) to non-obstructed subjects without chronic bronchitis (CB-). Multivariate analysis was used to determine factors associated with and impact of NCB. RESULTS We identified 597 NCB and 4283 CB- subjects. NCB participants were younger (55.4 vs. 57.2 years, p < 0.001) with greater tobacco exposure (42.9 vs. 37.8 pack-years, p < 0.001) and more often current smokers; more frequently reported occupational exposure to fumes (52.8% vs. 42.2%, p < 0.001), dust for ≥ 1 year (55.3% vs. 42.0%, p < 0.001) and were less likely to be currently working. NCB subjects demonstrated worse quality-of-life (SGRQ 35.6 vs. 15.1, p < 0.001) and exercise capacity (walk distance 415 vs. 449 m, p < 0.001) and more frequently reported respiratory "flare-ups" requiring treatment with antibiotics or steroids (0.30 vs. 0.10 annual events/subject, p < 0.001) prior to enrollment and during follow-up (0.34 vs. 0.16 annual events/subject, p < 0.001). In multivariate analysis, current smoking, GERD, sleep apnea and occupational exposures were significantly associated with NCB. CONCLUSIONS While longitudinal data will be needed to determine whether NCB progresses to COPD, NCB patients have poorer quality-of-life, exercise capacity and frequent respiratory events. Beyond smoking cessation interventions, further research is warranted to determine the benefit of other therapeutics in this population. Clinical Trials Registration # NCT00608764 (http://clinicaltrials.gov/show/NCT00608764). Link to study protocol: http://www.copdgene.org/sites/default/files/COPDGeneProtocol-5-0_06-19-2009.pdf.
Collapse
Affiliation(s)
- Carlos H Martinez
- Pulmonary & Critical Care Division, University of Michigan Health System, Ann Arbor, MI, USA.
| | - Victor Kim
- Division of Pulmonary and Critical Care, Temple University School of Medicine, Philadelphia, PA, USA
| | - Yahong Chen
- Respiratory Department, Peking University Third Hospital, Beijing, China
| | - Ella A Kazerooni
- Department of Radiology, University of Michigan, Ann Arbor, MI, USA
| | - Susan Murray
- School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - Gerard J Criner
- Division of Pulmonary and Critical Care, Temple University School of Medicine, Philadelphia, PA, USA
| | - Jeffrey L Curtis
- Pulmonary & Critical Care Division, University of Michigan Health System, Ann Arbor, MI, USA; Medicine Service, VA Healthcare System, Ann Arbor, MI, USA
| | - Elizabeth A Regan
- Department of Medicine, National Jewish Medical and Research Center, Denver, CO, USA
| | - Emily Wan
- Channing Division of Network Medicine and Pulmonary and Critical Care Division, Brigham and Women's Hospital, Boston, MA, USA
| | - Craig P Hersh
- Channing Division of Network Medicine and Pulmonary and Critical Care Division, Brigham and Women's Hospital, Boston, MA, USA
| | - Edwin K Silverman
- Channing Division of Network Medicine and Pulmonary and Critical Care Division, Brigham and Women's Hospital, Boston, MA, USA
| | - James D Crapo
- Department of Medicine, National Jewish Medical and Research Center, Denver, CO, USA
| | - Fernando J Martinez
- Pulmonary & Critical Care Division, University of Michigan Health System, Ann Arbor, MI, USA
| | - Meilan K Han
- Pulmonary & Critical Care Division, University of Michigan Health System, Ann Arbor, MI, USA
| | | |
Collapse
|
48
|
Goh F, Shaw JG, Savarimuthu Francis SM, Vaughan A, Morrison L, Relan V, Marshall HM, Dent AG, O'Hare PE, Hsiao A, Bowman RV, Fong KM, Yang IA. Personalizing and targeting therapy for COPD: the role of molecular and clinical biomarkers. Expert Rev Respir Med 2013; 7:593-605. [PMID: 24160750 DOI: 10.1586/17476348.2013.842468] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Chronic obstructive pulmonary disease (COPD) is a heterogeneous disease characterized by persistent airflow limitation. It is the third leading cause of death worldwide, and there are currently no curative strategies for this disease. Many factors contribute to COPD susceptibility, progression and exacerbations. These include cigarette smoking, environmental and occupational pollutants, respiratory infections and comorbidities. As the clinical phenotypes of COPD are so variable, it has been difficult to devise an individualized treatment plan for patients with this complex chronic disease. This review will highlight how potential clinical, inflammatory, genomic and epigenomic biomarkers for COPD could be used to personalize treatment, leading to improved disease management and prevention for our patients.
Collapse
Affiliation(s)
- Felicia Goh
- Department of Thoracic Medicine, The Prince Charles Hospital, Brisbane, Australia
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Biomarkers in Exhaled Breath Condensate and Serum of Chronic Obstructive Pulmonary Disease and Non-Small-Cell Lung Cancer. Int J Chronic Dis 2013; 2013:578613. [PMID: 26464846 PMCID: PMC4590922 DOI: 10.1155/2013/578613] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2013] [Accepted: 07/08/2013] [Indexed: 01/17/2023] Open
Abstract
Chronic obstructive pulmonary disease (COPD) and lung cancer are leading causes of deaths worldwide which are associated with chronic inflammation and oxidative stress. Lung cancer, in particular, has a very high mortality rate due to the characteristically late diagnosis. As such, identification of novel biomarkers which allow for early diagnosis of these diseases could improve outcome and survival rate. Markers of oxidative stress in exhaled breath condensate (EBC) are examples of potential diagnostic markers for both COPD and non-small-cell lung cancer (NSCLC). They may even be useful in monitoring treatment response. In the serum, S100A8, S100A9, and S100A12 of the S100 proteins are proinflammatory markers. They have been indicated in several inflammatory diseases and cancers including secondary metastasis into the lung. It is highly likely that they not only have the potential to be diagnostic biomarkers for NSCLC but also prognostic indicators and therapeutic targets.
Collapse
|
50
|
Westra JW, Schlage WK, Hengstermann A, Gebel S, Mathis C, Thomson T, Wong B, Hoang V, Veljkovic E, Peck M, Lichtner RB, Weisensee D, Talikka M, Deehan R, Hoeng J, Peitsch MC. A modular cell-type focused inflammatory process network model for non-diseased pulmonary tissue. Bioinform Biol Insights 2013; 7:167-92. [PMID: 23843693 PMCID: PMC3700945 DOI: 10.4137/bbi.s11509] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Exposure to environmental stressors such as cigarette smoke (CS) elicits a variety of biological responses in humans, including the induction of inflammatory responses. These responses are especially pronounced in the lung, where pulmonary cells sit at the interface between the body’s internal and external environments. We combined a literature survey with a computational analysis of multiple transcriptomic data sets to construct a computable causal network model (the Inflammatory Process Network (IPN)) of the main pulmonary inflammatory processes. The IPN model predicted decreased epithelial cell barrier defenses and increased mucus hypersecretion in human bronchial epithelial cells, and an attenuated pro-inflammatory (M1) profile in alveolar macrophages following exposure to CS, consistent with prior results. The IPN provides a comprehensive framework of experimentally supported pathways related to CS-induced pulmonary inflammation. The IPN is freely available to the scientific community as a resource with broad applicability to study the pathogenesis of pulmonary disease.
Collapse
|