1
|
Sinha I, Bitzer Z, Barnett S, Reinhart L, Umstead TM, Chroneos ZC, Lanza M, Sun D, Zhu J, Richie JP, Sinha R. Short-Term and Long-Term Effects of Electronic Cigarettes on Mouse Lungs Following Nose-Only Exposures. Chem Res Toxicol 2025. [PMID: 40401807 DOI: 10.1021/acs.chemrestox.4c00525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/23/2025]
Abstract
Health effects of electronic cigarettes (ECs) remain unknown, despite their popularity. We have determined that ECs produce highly reactive free radicals that could potentially cause damage in exposed tissues, mainly lungs. Goal for this study was to investigate the short- and long-term effects of ECs in mouse lungs. We focused on evaluating lung functions, oxidative stress related markers, and lung injury following nose-only exposures in male and female mice after 4- and 12-week periods. The EC exposure was modeled in vivo using nose-only exposures to C57BL/6 mice. For all studies, E-liquid (60:40; PG:VG) aerosols were compared to sham (compressed air) and to very low non-nicotine cigarette smoke (CS) controls in both sexes. Oxidative stress biomarkers (GSH, 8-Isoprostane, REDD1, and pGSK3β) and their selected downstream (RPS6) as well as upstream (AKT) target proteins in addition to pH2AX were measured by Western blot analysis. Lung function in mice was assessed by flexiVent and the injury scores were calculated following lung histology. Changes in cytology were also observed in cytospins from bronchoalveolar lavage (BALF). The lung injury (LI) score following 12-week exposures was significantly higher with EC and CS in female mice. Higher cell counts in BALF were mainly observed in CS exposed males and females at 4 and 12 weeks. 8-Isoprostane levels were significantly higher in EC and CS exposed males at 12 weeks. pGSK3β/GSK3β was low in males and higher in female mice at 4 weeks, and this difference was more pronounced at 12 weeks in CS exposed mice. Some mice exposed to EC and CS also showed DNA damage, as measured by pH2AX/H2AX expression. Based on the LI score, ECs were placed in between compressed air and CS. Our results showed the differentially expressed inflammation and oxidative stress/damage-related pathways from in vivo exposures to EC aerosols vs CS that could be an effective strategy for identifying EC relevant biomarkers of exposure and potential harm.
Collapse
Affiliation(s)
- Indu Sinha
- Department of Molecular and Precision Medicine, Penn State College of Medicine, Hershey, Pennsylvania 17033, United States
| | - Zachary Bitzer
- Department of Public Health Sciences, Penn State College of Medicine, Hershey, Pennsylvania 17033, United States
| | - Stephanie Barnett
- Department of Public Health Sciences, Penn State College of Medicine, Hershey, Pennsylvania 17033, United States
| | - Lisa Reinhart
- Department of Public Health Sciences, Penn State College of Medicine, Hershey, Pennsylvania 17033, United States
| | - Todd M Umstead
- Department of Pediatrics, Penn State College of Medicine, Hershey, Pennsylvania 17033, United States
| | - Zissis C Chroneos
- Department of Pediatrics, Penn State College of Medicine, Hershey, Pennsylvania 17033, United States
| | - Matthew Lanza
- Department of Comparative Medicine, Penn State College of Medicine, Hershey, Pennsylvania 17033, United States
| | - Dongxiao Sun
- Mass Spectrometry Core Facility (small molecule), Department of Pharmacology, Penn State College of Medicine, Hershey, Pennsylvania 17033, United States
| | - Junjia Zhu
- Department of Public Health Sciences, Penn State College of Medicine, Hershey, Pennsylvania 17033, United States
| | - John P Richie
- Department of Public Health Sciences, Penn State College of Medicine, Hershey, Pennsylvania 17033, United States
| | - Raghu Sinha
- Department of Molecular and Precision Medicine, Penn State College of Medicine, Hershey, Pennsylvania 17033, United States
| |
Collapse
|
2
|
Steinberg AW, Ozga JE, Tang Z, Stanton CA, Sargent JD, Paulin LM. Gender, tobacco and chronic obstructive pulmonary disease: analysis of the 2020 National Health Interview Survey. BMJ Open Respir Res 2025; 12:e002462. [PMID: 40340913 PMCID: PMC12067816 DOI: 10.1136/bmjresp-2024-002462] [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: 03/29/2024] [Accepted: 02/19/2025] [Indexed: 05/10/2025] Open
Abstract
RATIONALE Recent studies describe an increasing prevalence of chronic obstructive pulmonary disease (COPD) and higher COPD exacerbation rates among women compared with men despite lower average cigarette use, which has raised the question of whether women are more susceptible to the effects of tobacco smoke. We examined associations between gender, cigarette smoking and COPD in a national dataset. METHODS We used cross-sectional data for US respondents aged ≥40 years from the 2020 National Health Interview Survey (NHIS). Weighted multivariable logistic regressions assessed the relationship between gender and respondent-reported physician-diagnosed COPD, adjusting for tobacco use and sociodemographic covariates. Additional analyses were performed to determine if the relationship between cigarette smoking and COPD was modified by gender. RESULTS Women had a higher COPD prevalence (7.8%) than men (6.5%) despite lower cigarette smoke exposure. Women were less likely to have ever smoked, and among respondents who had smoked, women had a lower average pack-year history compared with men. In multivariable regressions, female gender was associated with a higher risk of COPD (adjusted risk ratio 1.47, 95% CI 1.30 to 1.65) and the relative risk was similar for respondents both with and without a history of smoking. Moreover, there was no significant interaction between gender and smoking status or gender and pack-year exposure relating to COPD prevalence. CONCLUSIONS Among adults aged ≥40 years, women had a roughly 50% greater risk of COPD than men. Higher susceptibility to cigarette smoking in women did not explain the difference.
Collapse
Affiliation(s)
| | - Jenny E Ozga
- Behavioral Health & Health Policy, Westat, Rockville, Maryland, USA
| | - Zhiqun Tang
- Behavioral Health & Health Policy, Westat, Rockville, Maryland, USA
| | | | - James D Sargent
- Pediatrics, Dartmouth College Geisel School of Medicine, Hanover, New Hampshire, USA
| | - Laura M Paulin
- Pulmonary and Critical Care, Dartmouth College Geisel School of Medicine, Hanover, New Hampshire, USA
| |
Collapse
|
3
|
Larcombe AN, Chivers EK, Landwehr KR, Berry LJ, de Jong E, Huxley RR, Musk A, Franklin PJ, Mullins BJ. Partial amelioration of a chronic cigarette-smoke-induced phenotype in mice by switching to electronic cigarettes. Arch Toxicol 2025:10.1007/s00204-025-04055-7. [PMID: 40249508 DOI: 10.1007/s00204-025-04055-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Accepted: 04/03/2025] [Indexed: 04/19/2025]
Abstract
Electronic cigarettes ("e-cigarettes") are often marketed as smoking cessation tools and are used by smokers to reduce/quit cigarette smoking. The objective of this study was to assess the health effects of switching to e-cigarettes after long-term smoking in a mouse model and compare these effects with continued smoking, or quitting entirely. Adult BALB/c mice were whole-body exposed to mainstream cigarette smoke (2 h/day, 5 days/week) for 12 weeks prior to switching to flavoured e-cigarette aerosol (50:50 propylene glycol and glycerine) containing 18 mg/mL nicotine (2 h/day and 5 days/week), continuing cigarette smoking (2 h/day and 5 days/week), or quitting entirely for an additional 2 weeks. We then assessed a range of respiratory health outcomes including lung function and structure, pulmonary inflammation and changes in gene expression in the lung. Switching to e-cigarettes led to improvements in some aspects of respiratory health in mice compared with continued smoking, such as reduced neutrophilic inflammation in the lung. However, total cellular lung inflammation was still elevated and lung function was still impaired, in terms of airway responsiveness to methacholine, for e-cigarette use compared with quitting. Larger effects were typically seen in female mice compared to male. This study shows that switching to e-cigarettes after long-term cigarette smoking leads to improvements in some aspects of respiratory health, such as neutrophilic inflammation and the volume dependence of lung function compared with continued smoking. However, switching to e-cigarettes was not as effective as quitting smoking entirely.
Collapse
Affiliation(s)
- Alexander N Larcombe
- Respiratory Environmental Health, Wal-yan Respiratory Research Centre, The Kids Research Institute Australia, 15 Hospital Avenue, Nedlands, Perth, WA, 6009, Australia.
- Occupation, Environment and Safety, School of Population Health, Curtin University, Perth, WA, Australia.
- School of Human Sciences, The University of Western Australia, Perth, WA, Australia.
| | - Emily K Chivers
- Respiratory Environmental Health, Wal-yan Respiratory Research Centre, The Kids Research Institute Australia, 15 Hospital Avenue, Nedlands, Perth, WA, 6009, Australia
| | - Katherine R Landwehr
- Respiratory Environmental Health, Wal-yan Respiratory Research Centre, The Kids Research Institute Australia, 15 Hospital Avenue, Nedlands, Perth, WA, 6009, Australia
- Occupation, Environment and Safety, School of Population Health, Curtin University, Perth, WA, Australia
| | - Luke J Berry
- Respiratory Environmental Health, Wal-yan Respiratory Research Centre, The Kids Research Institute Australia, 15 Hospital Avenue, Nedlands, Perth, WA, 6009, Australia
| | - Emma de Jong
- Centre for Health Research, The Kids Research Institute Australia, The University of Western Australia, Perth, WA, Australia
| | - Rachel R Huxley
- The George Institute for Global Health, University of New South Wales, Sydney, NSW, Australia
- Faculty of Health, Deakin University, Geelong, VIC, Australia
| | - Arthur Musk
- School of Population and Global Health, University of Western Australia, Perth, WA, Australia
| | - Peter J Franklin
- Respiratory Environmental Health, Wal-yan Respiratory Research Centre, The Kids Research Institute Australia, 15 Hospital Avenue, Nedlands, Perth, WA, 6009, Australia
- School of Population and Global Health, University of Western Australia, Perth, WA, Australia
| | - Benjamin J Mullins
- Occupation, Environment and Safety, School of Population Health, Curtin University, Perth, WA, Australia
| |
Collapse
|
4
|
Çolak Y, Nordestgaard BG, Lange P, Afzal S. Sex differences in COPD in relation to smoking exposure: a population-based cohort study. Thorax 2025:thorax-2024-222682. [PMID: 40185636 DOI: 10.1136/thorax-2024-222682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Accepted: 03/14/2025] [Indexed: 04/07/2025]
Abstract
BACKGROUND Sex discrepancies in the association between smoking and development and prognosis of chronic obstructive pulmonary disease (COPD) are controversial. We tested the hypothesis that females compared with males are more susceptible to the detrimental effects of smoking in relation to COPD. METHODS We identified 47 231 males and 57 806 females from the Copenhagen General Population Study. Smoking amount was assessed with sex interaction against COPD-related outcomes, including the cross-sectional association with airway obstruction, chronic bronchitis and dyspnoea, assessed using logistic regression analyses, and longitudinal association with exacerbation and mortality, assessed using Cox proportional hazard regression adjusted for potential confounders. RESULTS The increase in risk of airway obstruction (N=7367), chronic bronchitis (N=9206) and dyspnoea (N=8541) with higher smoking amount was greater in females compared with males. During 15 years' follow-up (median 9.3 years), the increase in risk of exacerbation (events=2756), respiratory mortality (events=711) and all-cause mortality (events=10 658) with higher smoking was greater for females compared with males. Compared with never-smokers, adjusted HRs for exacerbation increased from 4.64 (95% CI 2.83 to 7.61) in females with 10 pack-years to 41.6 (95% CI 28.8 to 60.2) in females with ≥50 pack-years, and from 2.21 (95% CI 0.92 to 5.32) in males with 10 pack-years to 23.7 (95% CI 12.9 to 43.5) in males with ≥50 pack-years. Corresponding HR increases for respiratory mortality were 2.04 (95% CI 1.27 to 3.26) to 11.1 (95% CI 7.39 to 16.8) in females and 1.09 (95% CI 0.62 to 1.92) to 5.66 (95% CI 3.96 to 8.11) in males, and for all-cause mortality, HR increases were 1.50 (95% CI 1.34 to 1.67) to 3.53 (95% CI 3.11 to 4.00) in females and 1.62 (1.45-1.81) to 2.94 (2.69-3.21) in males, respectively. CONCLUSIONS Females seem more susceptible to the detrimental effects of smoking in development and prognosis of COPD compared with males.
Collapse
Affiliation(s)
- Yunus Çolak
- Department of Respiratory Medicine and the Copenhagen General Population Study, Copenhagen University Hospital, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Børge G Nordestgaard
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Clinical Biochemistry and the Copenhagen General Population Study, Copenhagen University Hospital, Copenhagen, Denmark
| | - Peter Lange
- Department of Respiratory Medicine and the Copenhagen General Population Study, Copenhagen University Hospital, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Public Health, Section of Epidemiology, University of Copenhagen, Copenhagen, Denmark
| | - Shoaib Afzal
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Clinical Biochemistry and the Copenhagen General Population Study, Copenhagen University Hospital, Copenhagen, Denmark
| |
Collapse
|
5
|
Czarnota P, MacLeod JL, Gupta N, Manichaikul A, Shim YM. Sex Differences in Chronic Obstructive Pulmonary Disease: Implications for Pathogenesis, Diagnosis, and Treatment. Int J Mol Sci 2025; 26:2747. [PMID: 40141389 PMCID: PMC11942865 DOI: 10.3390/ijms26062747] [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: 01/09/2025] [Revised: 03/12/2025] [Accepted: 03/14/2025] [Indexed: 03/28/2025] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a leading chronic disease worldwide, with significant healthcare utilization, morbidity, and mortality. Irreversible airflow obstruction identified on spirometry establishes the diagnosis of COPD, but the disease entity encompasses a heterogeneous collection of lung diseases, including chronic bronchitis and emphysema. Despite the enormous burden of COPD, there are no pharmacological therapies that slow its progression or reduce mortality, indicating the need for a deeper understanding. There are sex differences concerning COPD prevalence, pathology, and symptoms. Historically thought to primarily affect males, its effect on females has increased significantly over time due to a rising prevalence of smoking and exposure to harmful pollutants among females. Over the past decade, the age-adjusted prevalence of COPD has been consistently higher in females than in males. Despite this, the impacts of biological sex continue to be confusing and poorly defined. The primary goal of this review is to organize and collate sex-dependent factors that may contribute to disease differences in males and females, thereby identifying future research questions in this area.
Collapse
Affiliation(s)
- Paulina Czarnota
- Department of Genome Sciences, University of Virginia School of Medicine, Charlottesville, VA 22908, USA;
| | - Jamie L. MacLeod
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, University of Virginia School of Medicine, Charlottesville, VA 22908, USA; (J.L.M.); (N.G.)
| | - Niya Gupta
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, University of Virginia School of Medicine, Charlottesville, VA 22908, USA; (J.L.M.); (N.G.)
| | - Ani Manichaikul
- Department of Genome Sciences, University of Virginia School of Medicine, Charlottesville, VA 22908, USA;
| | - Yun M. Shim
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, University of Virginia School of Medicine, Charlottesville, VA 22908, USA; (J.L.M.); (N.G.)
| |
Collapse
|
6
|
Knox-Brown B, Algharbi F, Mulhern O, Potts J, Harrabi I, Janson C, Nielsen R, Agarwal D, Malinovschi A, Juvekar S, Denguezli M, Gíslason T, Ahmed R, Nafees A, Koul PA, Obaseki D, Anand MP, Loh LC, Dias HB, Rodrigues F, Mannino D, Elbiaze M, El Rhazi K, Mejza F, Devereux G, Franssen FM, El Sony A, Wouters E, Al Ghobain M, Mortimer K, Rashid A, Osman R, Studnicka M, Cardoso J, Burney P, Amaral AF. Bronchodilator responsiveness and future chronic airflow obstruction: a multinational longitudinal study. EClinicalMedicine 2025; 81:103123. [PMID: 40083442 PMCID: PMC11905823 DOI: 10.1016/j.eclinm.2025.103123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Revised: 01/31/2025] [Accepted: 02/04/2025] [Indexed: 03/16/2025] Open
Abstract
Background Bronchodilator responsiveness testing is mainly used for diagnosing asthma. We aimed to investigate whether it is associated with progression to chronic airflow obstruction over time. Methods The multinational Burden of Obstructive Lung Disease cohort study surveyed adults, aged 40 years and above, at baseline and followed them up after a mean of 9.1 years. Recruitment took place between January 2, 2003 and December 26, 2016. Follow-up measurements were collected between January 29, 2019 and October 24, 2021. On both occasions, study participants provided information on respiratory symptoms, health status and several environmental and lifestyle exposures. They also underwent pre- and post-bronchodilator spirometry. We defined bronchodilator responsiveness at baseline using the American Thoracic Society and European Respiratory Society (ATS/ERS) 2022 definition, and the presence of chronic airflow obstruction at follow-up as a post-bronchodilator forced expiratory volume in 1 s to forced vital capacity ratio (FEV1/FVC) less than the lower limit of normal. We used multi-level regression models to estimate the association between baseline bronchodilator responsiveness and incident chronic airflow obstruction. We stratified analyses by gender and performed a sensitivity analysis in never smokers. Findings We analysed data from 3701 adults with 56% being women. Compared to those without bronchodilator responsiveness at baseline, those with bronchodilator responsiveness had 36% increased risk of developing chronic airflow obstruction (RR: 1.36, 95%CI 1.04, 1.80). This effect was stronger in women (RR: 1.45, 95%CI 1.09, 1.91) than men (RR: 1.07, 95%CI 0.51, 2.24). Never smokers with bronchodilator responsiveness also were at greater risk of incident chronic airflow obstruction (RR: 1.48, 95%CI 1.01, 2.20). Interpretation Bronchodilator responsiveness appears to be a risk factor for incident chronic airflow obstruction. It is important that future studies in other large population-based cohorts replicate these findings. Funding National Heart and Lung Institute, UK Medical Research Council, and Wellcome Trust.
Collapse
Affiliation(s)
- Ben Knox-Brown
- National Heart and Lung Institute, Imperial College London, London, UK
- Royal Papworth Hospital NHS FT, Cambridge, UK
| | - Fahad Algharbi
- National Heart and Lung Institute, Imperial College London, London, UK
- Respiratory Services, Royal Commission Health Services Program, Jubail, Saudi Arabia
| | - Octavia Mulhern
- National Heart and Lung Institute, Imperial College London, London, UK
- Guttmacher Institute, New York, USA
| | - James Potts
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Imed Harrabi
- Ibn El Jazzar Faculty of Medicine of Sousse, University of Sousse, Sousse, Tunisia
| | - Christer Janson
- Department of Medical Sciences: Respiratory, Allergy and Sleep Research, Uppsala University, Uppsala, Sweden
| | - Rune Nielsen
- Department of Clinical Science, Faculty of Medicine, University of Bergen, Bergen, Norway
| | - Dhiraj Agarwal
- Vadu Rural Health Program, KEM Hospital Research Centre, Pune, India
| | - Andrei Malinovschi
- Department of Medical Sciences, Clinical Physiology, Uppsala University, Uppsala, Sweden
| | - Sanjay Juvekar
- Vadu Rural Health Program, KEM Hospital Research Centre, Pune, India
- Dr. D. Y. Patil Medical College, Hospital and Research Centre, India
| | - Miriam Denguezli
- Université de Sousse, Faculté de Médecine de Sousse, LR19ES09, Sousse 4000, Tunisia
| | - Thorarinn Gíslason
- Faculty of Medicine, University of Iceland, Reykjavík, Iceland
- Department of Sleep, Landspitali - The National University Hospital of Iceland, Reykjavik, Iceland
| | - Rana Ahmed
- The Epidemiological Laboratory, Khartoum, Sudan
| | - Asaad Nafees
- Department of Community Health Sciences, Aga Khan University, Karachi, Pakistan
| | - Parvaiz A. Koul
- Sher-i-Kashmir Institute of Medical Sciences, Srinagar, J&K, India
| | | | | | - Li Cher Loh
- RCSI and UCD Malaysia Campus, Penang, Malaysia
| | - Hermínia Brites Dias
- Escola Superior de Tecnologia da Saúde de Lisboa, Politécnico de Lisboa (Lisbon School of Health Technology, Polytechnic of Lisbon), Lisbon, Portugal
| | - Fátima Rodrigues
- Pulmonology Department, Santa Maria Local Health Unit, Lisbon, Portugal
- Institute of Environmental Health, Lisbon Medical School, Lisbon University, Lisbon, Portugal
| | - David Mannino
- University of Kentucky, Lexington, KY, USA
- COPD Foundation, Miami, FL, USA
| | - Mohammed Elbiaze
- Faculté de Médecine et de Pharmacie et de Médecine dentaire Fès, Morocco
- Directeur du Centre de Médecine Universitaire du Sommeil et Spécialiste Pneumologie CHU Hassan II Fès, Morocco
| | - Karima El Rhazi
- Epidemiology and Research in Health Sciences Laboratory, Faculty of Medicine, Pharmacy and Dentistry, Sidi Mohamed Ben Abdillah University, Morocco
- Hassan Il University Hospital Centre of Fez, Morocco
| | - Filip Mejza
- Centre for Evidence Based Medicine, 2nd Department of Internal Medicine, Jagiellonian University Medical College, Kraków, Poland
| | - Graham Devereux
- Clinical Sciences, Liverpool School of Tropical Medicine, Pembroke Place, Liverpool, UK
| | | | | | - Emiel Wouters
- Maastricht University Medical Centre, Maastricht, the Netherlands
- Ludwig Boltzmann Institute for Lung Health, Vienna, Austria
| | - Mohammed Al Ghobain
- King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
- King Abdullah International Medical Research Centre, Riyadh, Saudi Arabia
| | - Kevin Mortimer
- University of Cambridge, Cambridge, UK
- Liverpool University Hospitals NHS Foundation Trust, Liverpool, UK
| | | | | | - Michael Studnicka
- Department of Pulmonary Medicine, Paracelsus Medical University, Salzburg, Austria
| | - Joao Cardoso
- Pulmonology Department, Centro Hospitalar Universitário de Lisboa Central, Lisboa, Portugal
- NOVA Medical School, Nova University Lisbon, Lisboa, Portugal
| | - Peter Burney
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Andre F.S. Amaral
- National Heart and Lung Institute, Imperial College London, London, UK
- NIHR Imperial Biomedical Research Centre, London
| |
Collapse
|
7
|
Yang JJ, Liu HJ, Wang YX, Wang LP, Gu JJ, Gao JY, Ren KQ, Min LF. Oxidative Stress and Epithelial-Mesenchymal Transition: The Impact of Ubiquitin C-terminal Hydrolase L1 in Cigarette Smoke-Induced COPD. Lung 2025; 203:36. [PMID: 40000498 DOI: 10.1007/s00408-025-00790-x] [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: 12/09/2024] [Accepted: 02/06/2025] [Indexed: 02/27/2025]
Abstract
PURPOSE Cigarette smoke (CS) has been demonstrated to mediate oxidative stress (OS) and epithelial-mesenchymal transition (EMT) in bronchial epithelial cells, thereby contributing to airway remodeling in chronic obstructive pulmonary disease (COPD). Studies have shown upregulation of Ubiquitin C-terminal hydrolase L1 (UCHL1), a deubiquitinating enzyme, in the airway epithelium of smokers. Many studies indicate that UCHL1's regulation of EMT and OS has a complex role in various cell types, including respiratory epithelium. Thus, we aimed to investigate UCHL1's regulation of EMT, OS, and related mechanisms in cigarette smoke-exposed airway epithelium. METHODS Exposure to cigarette smoke (CS) or cigarette smoke extract (CSE) was employed to establish both animal and cellular models. Protein expression was analyzed using immunohistochemistry, immunofluorescence, and Western blotting. Lentiviral UCHL1 or GPX1-siRNA was used to modulate UCHL1 or GPX1 expression, respectively. Transwell assays were employed to evaluate cell migration and EMT-related alterations. Oxidative stress levels were assessed using specific assay kits. RESULTS This study validated that exposure to CS induces UCHL1 expression in bronchial epithelial cells both in vitro and in vivo, a phenomenon positively correlated with increased OS and EMT in the airway. Notably, UCHL1 overexpression counteracted CSE's impact on EMT markers, cell migration, and oxidative stress in BEAS-2B cells, while UCHL1 knockdown exacerbated these effects. Furthermore, in BEAS-2B cells treated with CSE, upregulation of UCHL1 was found to enhance the expression of glutathione peroxidase 1 (GPX1), an antioxidant enzyme. The effect of UCHL1 overexpression on EMT-related protein markers and cell migration was reversed upon GPX1 silencing via siRNA. CONCLUSIONS These findings suggest that UCHL1-mediated regulation of GPX1 expression alleviates cigarette smoke-induced EMT-related protein markers change and cell migration in BEAS-2B cell.
Collapse
Affiliation(s)
- Jing Jing Yang
- Department of Pulmonary and Critical Care Medicine, Northern Jiangsu People's Hospital, Clinical Medical College, Yangzhou University, Yangzhou, Jiangsu, China
| | - Hong Jun Liu
- Department of Respiratory Medicine, Jingjiang People's Hospital Affiliated to Yangzhou University, Taizhou, Jiangsu, China
| | - Yu Xiu Wang
- Department of Pulmonary and Critical Care Medicine, Northern Jiangsu People's Hospital, Clinical Medical College, Yangzhou University, Yangzhou, Jiangsu, China
| | - Li Ping Wang
- Biospecimen Library, Northern Jiangsu People's Hospital, Clinical Medical College, Yangzhou University, Yangzhou, Jiangsu, China
| | - Jian Jun Gu
- Department of Cardiology, Institute of Translational Medicine, Clinical Medical College, Yangzhou University, Yangzhou, Jiangsu, China
| | - Jun Yin Gao
- Pulmonary and Critical Care Medicine, Yancheng No.1 People's Hospital, 224000, Yancheng, Jiangsu, China
| | - Kai Qi Ren
- Department of Pulmonary and Critical Care Medicine, Northern Jiangsu People's Hospital, Clinical Medical College, Yangzhou University, Yangzhou, Jiangsu, China
| | - Ling Feng Min
- Department of Pulmonary and Critical Care Medicine, Northern Jiangsu People's Hospital, Clinical Medical College, Yangzhou University, Yangzhou, Jiangsu, China.
| |
Collapse
|
8
|
Taylor-Blair HC, Siu ACW, Haysom-McDowell A, Kokkinis S, Bani Saeid A, Chellappan DK, Oliver BGG, Paudel KR, De Rubis G, Dua K. The impact of airborne particulate matter-based pollution on the cellular and molecular mechanisms in chronic obstructive pulmonary disease (COPD). THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 954:176413. [PMID: 39322084 DOI: 10.1016/j.scitotenv.2024.176413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 08/26/2024] [Accepted: 09/18/2024] [Indexed: 09/27/2024]
Abstract
Inhalation of particulate matter (PM), one of the many components of air pollution, is associated with the development and exacerbation of chronic respiratory diseases, such as chronic obstructive pulmonary disease (COPD). COPD is one of the leading causes of global mortality and morbidity, with a paucity of therapeutic options and a significant contributor to global health expenditure. This review aims to provide a mechanistic understanding of the cellular and molecular pathways that lead to the development of COPD following chronic PM exposure. Our review describes how the inhalation of PM can lead to lung parenchymal destruction and cellular senescence due to chronic pulmonary inflammation and oxidative stress. Following inhalation of PM, significant increases in a range of pro-inflammatory cytokines, mediated by the nuclear factor kappa B pathway are reported. This review also highlights how the inhalation of PM can lead to deleterious chronic oxidative stress persisting in the lung post-exposure. Furthermore, our work summarises how PM inhalation can lead to airway remodelling, with increases in pro-fibrotic cytokines and collagen deposition, typical of COPD. This paper also accentuates the interconnection and possible synergism between the pathophysiological mechanisms leading to COPD. Our work emphasises the serious health consequences of PM exposure on respiratory health. Elucidation of the cellular and molecular mechanisms can provide insight into possible therapeutic options. Finally, this review should serve as a stark reminder of the need for genuine action on air pollution to decrease the associated health burden on our growing global population.
Collapse
Affiliation(s)
- Hudson C Taylor-Blair
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Ultimo, NSW 2007, Australia; Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Ultimo, NSW 2007, Australia
| | - Alexander Chi Wang Siu
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Ultimo, NSW 2007, Australia
| | - Adam Haysom-McDowell
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Ultimo, NSW 2007, Australia; Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, NSW 2007, Australia
| | - Sofia Kokkinis
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Ultimo, NSW 2007, Australia; Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, NSW 2007, Australia
| | - Ayeh Bani Saeid
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Ultimo, NSW 2007, Australia; Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, NSW 2007, Australia
| | - Dinesh Kumar Chellappan
- Department of Life Sciences, School of Pharmacy, International Medical University, Bukit Jalil, 57000 Kuala Lumpur, Malaysia
| | - Brian G G Oliver
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Ultimo, NSW 2007, Australia; Respiratory Cellular and Molecular Biology, Woolcock Institute of Medical Research, Macquarie Park, NSW 2113, Australia
| | - Keshav Raj Paudel
- Centre for Inflammation, Centenary Institute, University of Technology Sydney, School of Life Sciences, Faculty of Science, Ultimo, NSW 2007, Australia
| | - Gabriele De Rubis
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Ultimo, NSW 2007, Australia; Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, NSW 2007, Australia.
| | - Kamal Dua
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Ultimo, NSW 2007, Australia; Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, NSW 2007, Australia.
| |
Collapse
|
9
|
Wang Z, Deng M, Xu W, Li C, Zheng Z, Li J, Liao L, Zhang Q, Bian Y, Li R, Miao J, Wang K, Yin Y, Li Y, Zhou X, Hou G. DKK3 as a diagnostic marker and potential therapeutic target for sarcopenia in chronic obstructive pulmonary disease. Redox Biol 2024; 78:103434. [PMID: 39571512 PMCID: PMC11617289 DOI: 10.1016/j.redox.2024.103434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 10/11/2024] [Accepted: 11/16/2024] [Indexed: 12/08/2024] Open
Abstract
Sarcopenia, characterized by the progressive loss of muscle mass and function, significantly affects patients with chronic obstructive pulmonary disease (COPD) and worsens their morbidity and mortality. The pathogenesis of muscle atrophy in patients with COPD involves complex mechanisms, including protein imbalance and mitochondrial dysfunction, which have been identified in the muscle tissues of patients with COPD. DKK3 (Dickkopf-3) is a secreted glycoprotein involved in the process of myogenesis. However, the role of DKK3 in the regulation of muscle mass is largely unknown. This study investigated the role of DKK3 in COPD-related sarcopenia. DKK3 was found to be overexpressed in cigarette smoking-induced muscle atrophy and in patients with COPD. Importantly, plasma DKK3 levels in COPD patients with sarcopenia were significantly higher than those without sarcopenia, and plasma DKK3 levels could effectively predict sarcopenia in patients with COPD based on two independent cohorts. Mechanistically, DKK3 is secreted by skeletal muscle cells that acts in autocrine and paracrine manners and interacts with the cell surface-activated receptor cytoskeleton-associated protein 4 (CKAP4) to induce mitochondrial dysfunction and myotube atrophy. The inhibition of DKK3 by genetic ablation prevented cigarette smoking-induced skeletal muscle dysfunction. These results suggest that DKK3 is a potential target for the diagnosis and treatment of sarcopenia in patients with COPD.
Collapse
Affiliation(s)
- Zilin Wang
- National Center for Respiratory Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, National Clinical Research Center for Respiratory Diseases, Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China
| | - Mingming Deng
- National Center for Respiratory Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, National Clinical Research Center for Respiratory Diseases, Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China
| | - Weidong Xu
- National Center for Respiratory Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, National Clinical Research Center for Respiratory Diseases, Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China
| | - Chang Li
- National Center for Respiratory Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, National Clinical Research Center for Respiratory Diseases, Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China
| | - Ziwen Zheng
- National Center for Respiratory Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, National Clinical Research Center for Respiratory Diseases, Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China
| | - Jiaye Li
- National Center for Respiratory Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, National Clinical Research Center for Respiratory Diseases, Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China
| | - Liwei Liao
- National Center for Respiratory Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, National Clinical Research Center for Respiratory Diseases, Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China
| | - Qin Zhang
- National Center for Respiratory Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, National Clinical Research Center for Respiratory Diseases, Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China
| | - Yiding Bian
- National Center for Respiratory Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, National Clinical Research Center for Respiratory Diseases, Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China
| | - Ruixia Li
- National Center for Respiratory Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, National Clinical Research Center for Respiratory Diseases, Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China; Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Harbin Medical University, Harbin Medical University, Harbin, China
| | - Jinrui Miao
- National Center for Respiratory Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, National Clinical Research Center for Respiratory Diseases, Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China; Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Harbin Medical University, Harbin Medical University, Harbin, China
| | - Kai Wang
- National Center for Respiratory Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, National Clinical Research Center for Respiratory Diseases, Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China
| | - Yan Yin
- Department of Pulmonary and Critical Care Medicine, First Hospital of China Medical University, Shenyang, China
| | - Yanxia Li
- Respiratory Department, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Xiaoming Zhou
- Department of Pulmonary and Critical Care Medicine, Disease, Fuwai Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Gang Hou
- National Center for Respiratory Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, National Clinical Research Center for Respiratory Diseases, Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China.
| |
Collapse
|
10
|
Gong KQ, Brune JE, Guo X, Manicone AM. MAP2K1 dampens cigarette smoke-induced inflammation via suppression of type I interferon pathway activation. Am J Physiol Lung Cell Mol Physiol 2024; 327:L740-L748. [PMID: 39316676 PMCID: PMC11563587 DOI: 10.1152/ajplung.00080.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 08/29/2024] [Accepted: 09/11/2024] [Indexed: 09/26/2024] Open
Abstract
Chronic obstructive pulmonary disease (COPD), comprised of chronic bronchitis and emphysema, is a leading cause of morbidity and mortality worldwide. Mitogen-activated protein 2 kinase (MAP2K) pathway activation is present in COPD lung tissue and a genetic polymorphism in Map2k1 associates with FEV1 decline in COPD, suggesting it may contribute to disease pathogenesis. To test the functional contribution of Map2k1 in cigarette smoke (CS)-induced lung inflammation, we used a short-term CS exposure model in mice deficient in myeloid Map2k1 (LysmCre+Mek1fl) and wild-type mice (Mek1fl). Mice deficient in myeloid Map2k1 had enhanced CS-induced lung inflammation characterized by increased neutrophil recruitment, vascular leak, augmented expression of elastolytic matrix metalloproteinases, and increased type I interferon-stimulated gene expression. The augmented neutrophilic inflammatory response could be abrogated by IFNAR1 blockade. These findings indicate that myeloid Map2k1 regulates the immune response to CS via inhibition of the type I interferon pathway. Overall, these results suggest that Map2k1 is a critical determinant in modulating the severity of CS-induced lung inflammation and its expression is protective.NEW & NOTEWORTHY Activation of the mitogen-activated protein kinases (MAPK)-ERK1/2 pathway is present in COPD lung tissue compared with healthy lungs. Our study using mice deficient in myeloid Map2k1 reveals that Map2k1 is a critical determinant in modulating the severity of CS-induced lung inflammation via suppression of type I interferon responses, and its expression is protective.
Collapse
Affiliation(s)
- Ke-Qin Gong
- Center for Lung Biology, University of Washington, Seattle, Washington, United States
- Division of Pulmonary, Critical Care and Sleep Medicine, University of Washington, Seattle, Washington, United States
| | - Jourdan E Brune
- Center for Lung Biology, University of Washington, Seattle, Washington, United States
- Department of Comparative Medicine, University of Washington, Seattle, Washington, United States
| | - Xiaoyun Guo
- Center for Lung Biology, University of Washington, Seattle, Washington, United States
- Division of Pulmonary, Critical Care and Sleep Medicine, University of Washington, Seattle, Washington, United States
| | - Anne M Manicone
- Center for Lung Biology, University of Washington, Seattle, Washington, United States
- Division of Pulmonary, Critical Care and Sleep Medicine, University of Washington, Seattle, Washington, United States
| |
Collapse
|
11
|
Guo J, Yang J, Wang J, Liu W, Kang Y, Li Z, Hao C, Qi S. Exploring Gender Differences in the Association Between TyG Index and COPD: A Cross-Sectional Study from NHANES 1999-2018. Int J Chron Obstruct Pulmon Dis 2024; 19:2001-2010. [PMID: 39253040 PMCID: PMC11381934 DOI: 10.2147/copd.s473089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Accepted: 08/25/2024] [Indexed: 09/11/2024] Open
Abstract
Purpose This study examined gender differences in the association of Triglyceride-Glucose (TyG) index with the prevalence of chronic obstructive pulmonary disease (COPD), particularly in a non-diabetic population. Methods The study leveraged data from the National Health and Nutrition Examination Survey (NHANES), spanning from 1999 to 2018, with a cohort of 23,456 participants. Logistic regression and restricted cubic spline analyses were employed to explore the relationship between the TyG index and COPD prevalence. Results Statistical analyses revealed a significant positive association between the TyG index and COPD prevalence among non-diabetic women after adjustment for all covariates (OR=1.50; 95% CI, 1.08-2.08), supported by a linear relationship (P for non-linearity=0.298). No equivalent significant association was found in non-diabetic men (OR=1.00; 95% CI, 0.67-1.48). Within the diabetic group, the TyG index did not show a significant association with COPD prevalence, regardless of gender. Conclusion Our study reveals a significant positive correlation between the TyG index and COPD prevalence in the non-diabetic population, marked by notable gender differences.
Collapse
Affiliation(s)
- Jun Guo
- General Surgery, Bethune International Peace Hospital, Shijiazhuang, People's Republic of China
| | - Jie Yang
- Cadre Ward, Bethune International Peace Hospital, Shijiazhuang, People's Republic of China
| | - Jinghua Wang
- Nephrology, Bethune International Peace Hospital, Shijiazhuang, People's Republic of China
| | - Wei Liu
- General Surgery, Bethune International Peace Hospital, Shijiazhuang, People's Republic of China
| | - Yingjie Kang
- General Surgery, Hebei Yiling Hospital, Shijiazhuang, People's Republic of China
| | - Zhi Li
- General Surgery, Bethune International Peace Hospital, Shijiazhuang, People's Republic of China
| | - Chen Hao
- General Surgery, Bethune International Peace Hospital, Shijiazhuang, People's Republic of China
| | - Shuai Qi
- General Surgery, Bethune International Peace Hospital, Shijiazhuang, People's Republic of China
| |
Collapse
|
12
|
Lopes-Ramos CM, Shutta KH, Ryu MH, Huang Y, Saha E, Ziniti J, Chase R, Hobbs BD, Yun JH, Castaldi P, Hersh CP, Glass K, Silverman EK, Quackenbush J, DeMeo DL. Sex-biased Regulation of Extracellular Matrix Genes in COPD. Am J Respir Cell Mol Biol 2024; 72:72-81. [PMID: 39102858 PMCID: PMC11707671 DOI: 10.1165/rcmb.2024-0226oc] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Accepted: 08/05/2024] [Indexed: 08/07/2024] Open
Abstract
Compared to men, women often develop COPD at an earlier age with worse respiratory symptoms despite lower smoking exposure. However, most preventive, and therapeutic strategies ignore biological sex differences in COPD. Our goal was to better understand sex-specific gene regulatory processes in lung tissue and the molecular basis for sex differences in COPD onset and severity. We analyzed lung tissue gene expression and DNA methylation data from 747 individuals in the Lung Tissue Research Consortium (LTRC), and 85 individuals in an independent dataset. We identified sex differences in COPD-associated gene regulation using gene regulatory networks. We used linear regression to test for sex-biased associations of methylation with lung function, emphysema, smoking, and age. Analyzing gene regulatory networks in the control group, we identified that genes involved in the extracellular matrix (ECM) have higher transcriptional factor targeting in females than in males. However, this pattern is reversed in COPD, with males showing stronger regulatory targeting of ECM-related genes than females. Smoking exposure, age, lung function, and emphysema were all associated with sex-specific differential methylation of ECM-related genes. We identified sex-based gene regulatory patterns of ECM-related genes associated with lung function and emphysema. Multiple factors including epigenetics, smoking, aging, and cell heterogeneity influence sex-specific gene regulation in COPD. Our findings underscore the importance of considering sex as a key factor in disease susceptibility and severity.
Collapse
Affiliation(s)
- Camila M. Lopes-Ramos
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
- Channing Division of Network Medicine, Brigham and Women’s Hospital, Boston, Massachusetts
- Department of Medicine, Harvard Medical School, Boston, Massachusetts; and
| | - Katherine H. Shutta
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
- Channing Division of Network Medicine, Brigham and Women’s Hospital, Boston, Massachusetts
| | - Min Hyung Ryu
- Channing Division of Network Medicine, Brigham and Women’s Hospital, Boston, Massachusetts
- Department of Medicine, Harvard Medical School, Boston, Massachusetts; and
| | - Yichen Huang
- Channing Division of Network Medicine, Brigham and Women’s Hospital, Boston, Massachusetts
| | - Enakshi Saha
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | - John Ziniti
- Channing Division of Network Medicine, Brigham and Women’s Hospital, Boston, Massachusetts
| | - Robert Chase
- Channing Division of Network Medicine, Brigham and Women’s Hospital, Boston, Massachusetts
| | - Brian D. Hobbs
- Channing Division of Network Medicine, Brigham and Women’s Hospital, Boston, Massachusetts
| | - Jeong H. Yun
- Channing Division of Network Medicine, Brigham and Women’s Hospital, Boston, Massachusetts
- Department of Medicine, Harvard Medical School, Boston, Massachusetts; and
| | - Peter Castaldi
- Channing Division of Network Medicine, Brigham and Women’s Hospital, Boston, Massachusetts
- Department of Medicine, Harvard Medical School, Boston, Massachusetts; and
| | - Craig P. Hersh
- Channing Division of Network Medicine, Brigham and Women’s Hospital, Boston, Massachusetts
- Department of Medicine, Harvard Medical School, Boston, Massachusetts; and
| | - Kimberly Glass
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
- Channing Division of Network Medicine, Brigham and Women’s Hospital, Boston, Massachusetts
- Department of Medicine, Harvard Medical School, Boston, Massachusetts; and
| | - Edwin K. Silverman
- Channing Division of Network Medicine, Brigham and Women’s Hospital, Boston, Massachusetts
- Department of Medicine, Harvard Medical School, Boston, Massachusetts; and
| | - John Quackenbush
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
- Channing Division of Network Medicine, Brigham and Women’s Hospital, Boston, Massachusetts
- Department of Data Science, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Dawn L. DeMeo
- Channing Division of Network Medicine, Brigham and Women’s Hospital, Boston, Massachusetts
- Department of Medicine, Harvard Medical School, Boston, Massachusetts; and
| |
Collapse
|
13
|
Firoozi Z, Shahi A, Mohammadisoleimani E, Afzali S, Mansoori B, Bahmanyar M, Mohaghegh P, Dastsooz H, Pezeshki B, Nikfar G, Kouhpayeh SA, Mansoori Y. CircRNA-associated ceRNA networks (circCeNETs) in chronic obstructive pulmonary disease (COPD). Life Sci 2024; 349:122715. [PMID: 38740326 DOI: 10.1016/j.lfs.2024.122715] [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: 09/24/2023] [Revised: 04/29/2024] [Accepted: 05/09/2024] [Indexed: 05/16/2024]
Abstract
Chronic obstructive pulmonary disease (COPD), a chronic airway disorder, which is mostly brought on by cigarette smoke extract (CSE), is a leading cause of death which has a high frequency. In COPD patients, smoking cigarette could also trigger the epithelial-mesenchymal transition (EMT) of airway remodeling. One of the most significant elements of environmental contaminants that is linked to pulmonary damage is fine particulate matter (PM2.5). However, the basic processes of lung injury brought on by environmental contaminants and cigarette smoke are poorly understood, particularly the molecular pathways involved in inflammation. For the clinical management of COPD, investigating the molecular process and identifying workable biomarkers will be important. According to newly available research, circular RNAs (circRNAs) are aberrantly produced and serve as important regulators in the pathological processes of COPD. This class of non-coding RNAs (ncRNAs) functions as microRNA (miRNA) sponges to control the levels of gene expression, changing cellular phenotypes and advancing disease. These findings led us to concentrate our attention in this review on new studies about the regulatory mechanism and potential roles of circRNA-associated ceRNA networks (circCeNETs) in COPD.
Collapse
Affiliation(s)
- Zahra Firoozi
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran; Noncommunicable Diseases Research Center, Fasa University of Medical Sciences, Fasa, Iran
| | - Abbas Shahi
- Noncommunicable Diseases Research Center, Fasa University of Medical Sciences, Fasa, Iran; Student Research Committee, Fasa University of Medical Sciences, Fasa, Iran; Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Elham Mohammadisoleimani
- Noncommunicable Diseases Research Center, Fasa University of Medical Sciences, Fasa, Iran; Department of Medical Microbiology (Bacteriology & Virology), Afzalipour Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Shima Afzali
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Behnam Mansoori
- Noncommunicable Diseases Research Center, Fasa University of Medical Sciences, Fasa, Iran
| | - Maryam Bahmanyar
- Pediatrics Department, School of Medicine, Fasa University of Medical Sciences, Fasa, Iran
| | - Poopak Mohaghegh
- Pediatrics Department, School of Medicine, Fasa University of Medical Sciences, Fasa, Iran
| | - Hassan Dastsooz
- Department of Life Sciences and Systems Biology, University of Turin, Turin, Italy; Candiolo, C/o IRCCS, IIGM-Italian Institute for Genomic Medicine, Turin, Italy; Candiolo Cancer (IT), FPO-IRCCS, Candiolo Cancer Institute, Turin, Italy
| | - Babak Pezeshki
- Noncommunicable Diseases Research Center, Fasa University of Medical Sciences, Fasa, Iran
| | - Ghasem Nikfar
- Noncommunicable Diseases Research Center, Fasa University of Medical Sciences, Fasa, Iran
| | - Seyed Amin Kouhpayeh
- Department of Pharmacology, Faculty of Medicine, Fasa University of Medical Sciences, Fasa, Iran
| | - Yaser Mansoori
- Noncommunicable Diseases Research Center, Fasa University of Medical Sciences, Fasa, Iran; Department of Medical Genetics, Fasa University of Medical Sciences, Fasa, Iran.
| |
Collapse
|
14
|
Xin L, An TM, Ying L, Rong DW, Lei H. Prevalence and risk factors for obstructive pulmonary dysfunction caused by silica dust exposure: a multicenter cross-sectional study. BMC Pulm Med 2024; 24:297. [PMID: 38918735 PMCID: PMC11197187 DOI: 10.1186/s12890-024-03106-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Accepted: 06/18/2024] [Indexed: 06/27/2024] Open
Abstract
OBJECTIVE To understand the prevalence rate of obstructive pulmonary dysfunction in workers exposed to silica dust and analyze its risk factors, so as to provide reference for the formulation of diagnostic criteria for chronic obstructive pulmonary disease caused by occupational dust. METHODS Data collection and structured questionnaire were used to collect the data of 2064 workers exposed to silica dust who underwent health examination in Hunan Occupational Disease Prevention and Control Hospital and Yuanling Second People's Hospital from January 1, 2021 to June 30, 2022. The prevalence rate of obstructive pulmonary ventilation dysfunction was analyzed and the risk factors were analyzed. RESULTS The prevalence rate of obstructive pulmonary ventilation dysfunction (FEV1/FVC < 70%) was 2.3% in 2064 silica dust exposed workers. The prevalence of restrictive pulmonary ventilation dysfunction (FVC/Pre < 80%) was 8.1%. The prevalence of obstructive pulmonary ventilation dysfunction in the high level exposure group was higher than that in the low level exposure group, 8.2 vs0.9% (P < 0.05). The rate of obstructive pulmonary ventilation dysfunction in female group was higher than that in male group (5.3% vs. 1.7%, p = 0.00). Workers with obstructive pulmonary dysfunction were older and worked longer than workers without obstructive pulmonary dysfunction, but there was no statistical difference. Multivariate regression analysis showed that high exposure level was a risk factor for obstructive pulmonary ventilation dysfunction in silica dust exposed workers (P < 0.05). Females were the risk factors for obstructive pulmonary ventilation dysfunction (P < 0.05). CONCLUSION Silica dust exposure can cause obstructive pulmonary ventilation dysfunction and lead to chronic obstructive pulmonary disease. High level of exposure is a risk factor for obstructive pulmonary ventilation dysfunction. Women exposed to dust are more prone to obstructive pulmonary ventilation dysfunction than men. Early diagnosis of chronic obstructive pulmonary disease caused by silica dust and timely intervention measures are very important to delay the decline of lung function and protect the health of workers.
Collapse
Affiliation(s)
- Li Xin
- Hunan Prevention and Treatment Institute for Occupational Diseases, Changsha, China
| | - Tang Mei An
- Hunan Prevention and Treatment Institute for Occupational Diseases, Changsha, China
| | - Li Ying
- Hunan Prevention and Treatment Institute for Occupational Diseases, Changsha, China
| | - Dai Wei Rong
- Hunan Prevention and Treatment Institute for Occupational Diseases, Changsha, China
| | - Huang Lei
- Hunan Prevention and Treatment Institute for Occupational Diseases, Changsha, China.
| |
Collapse
|
15
|
Negasi ZH, Nommi N, Liu C, Tesfaigzi Y. Persistence of emphysema following cessation of cigarette smoke exposure requires a susceptibility factor. Am J Physiol Lung Cell Mol Physiol 2024; 326:L431-L439. [PMID: 38349118 PMCID: PMC11281787 DOI: 10.1152/ajplung.00342.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 02/01/2024] [Accepted: 02/02/2024] [Indexed: 02/21/2024] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is caused by cigarette smoke (CS) exposure but can often be progressive even in former smokers. Exposure of mice to CS for 22 wk causes emphysema, but whether emphysema persists after cessation of CS exposure is not clear. The purpose of this study was to determine whether emphysema persists in mice following a recovery period of 22 wk and whether a susceptibility factor, such as deficiency in the Bcl-2-interacting killer (Bik), is required for this persistence. Therefore, bik+/+ and bik-/- mice at 6-10 wk of age were exposed to 250 mg/m3 total particulate matter of CS or filtered air (FA) for 3 or 22 wk and were kept in FA for an additional 22 wk. Lungs were lavaged to quantify inflammatory cells, and sections were stained with hematoxylin and eosin to assess severity of emphysema. Exposure to CS for 3 wk increased the number of inflammatory cells in bik-/- mice compared with bik+/+ mice but not at 22 wk of exposure. At 22 wk of CS exposure, extent of emphysema was similar in bik+/+ and bik-/- mice. However, when mice were exposed to CS over the first 22 wk and were kept in FA for an additional 22 wk, emphysema remained similar in bik+/+ mice but was enhanced in bik-/- mice. These findings link increased inflammation with persistent emphysematous changes even after smoking cessation and demonstrate that a preexisting susceptibility condition is required to sustain enhanced emphysema that was initiated by long-term CS exposure.NEW & NOTEWORTHY Exposure of mice to cigarette smoke (CS) for 22 wk causes emphysema, but whether emphysema persists after an additional period of 6 mo after cessation of CS exposure has not been reported. In addition, the role of preexisting susceptibility in enhancing the persistence of CS-induced emphysema after exposure to CS has stopped has not been shown. The present study shows that a preexisting susceptibility must be present to enhance CS-induced emphysema after cessation of CS exposure.
Collapse
Affiliation(s)
- Zerihun Hailemariam Negasi
- Pulmonary Critical Care Medicine Division, Brigham and Women's Hospital and Harvard Medical School, Harvard University, Boston, Massachusetts, United States
| | - Naomi Nommi
- Pulmonary Critical Care Medicine Division, Brigham and Women's Hospital and Harvard Medical School, Harvard University, Boston, Massachusetts, United States
| | - Congjian Liu
- Pulmonary Critical Care Medicine Division, Brigham and Women's Hospital and Harvard Medical School, Harvard University, Boston, Massachusetts, United States
| | - Yohannes Tesfaigzi
- Pulmonary Critical Care Medicine Division, Brigham and Women's Hospital and Harvard Medical School, Harvard University, Boston, Massachusetts, United States
- Chronic Obstructive Pulmonary Disease Program, Lovelace Respiratory Research Institute, Albuquerque, New Mexico, United States
| |
Collapse
|
16
|
Milne KM, Mitchell RA, Ferguson ON, Hind AS, Guenette JA. Sex-differences in COPD: from biological mechanisms to therapeutic considerations. Front Med (Lausanne) 2024; 11:1289259. [PMID: 38572156 PMCID: PMC10989064 DOI: 10.3389/fmed.2024.1289259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Accepted: 02/29/2024] [Indexed: 04/05/2024] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a heterogeneous respiratory condition characterized by symptoms of dyspnea, cough, and sputum production. We review sex-differences in disease mechanisms, structure-function-symptom relationships, responses to therapies, and clinical outcomes in COPD with a specific focus on dyspnea. Females with COPD experience greater dyspnea and higher morbidity compared to males. Imaging studies using chest computed tomography scans have demonstrated that females with COPD tend to have smaller airways than males as well as a lower burden of emphysema. Sex-differences in lung and airway structure lead to critical respiratory mechanical constraints during exercise at a lower absolute ventilation in females compared to males, which is largely explained by sex differences in maximum ventilatory capacity. Females experience similar benefit with respect to inhaled COPD therapies, pulmonary rehabilitation, and smoking cessation compared to males. Ongoing re-assessment of potential sex-differences in COPD may offer insights into the evolution of patterns of care and clinical outcomes in COPD patients over time.
Collapse
Affiliation(s)
- Kathryn M. Milne
- Centre for Heart Lung Innovation, The University of British Columbia and Providence Research, St. Paul’s Hospital, Vancouver, BC, Canada
- Division of Respiratory Medicine, The University of British Columbia, Vancouver, BC, Canada
| | - Reid A. Mitchell
- Centre for Heart Lung Innovation, The University of British Columbia and Providence Research, St. Paul’s Hospital, Vancouver, BC, Canada
| | - Olivia N. Ferguson
- Centre for Heart Lung Innovation, The University of British Columbia and Providence Research, St. Paul’s Hospital, Vancouver, BC, Canada
| | - Alanna S. Hind
- Centre for Heart Lung Innovation, The University of British Columbia and Providence Research, St. Paul’s Hospital, Vancouver, BC, Canada
| | - Jordan A. Guenette
- Centre for Heart Lung Innovation, The University of British Columbia and Providence Research, St. Paul’s Hospital, Vancouver, BC, Canada
- Division of Respiratory Medicine, The University of British Columbia, Vancouver, BC, Canada
- Department of Physical Therapy, The University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
17
|
Zhang J, Yi Q, Zhou C, Luo Y, Wei H, Ge H, Liu H, Zhang J, Li X, Xie X, Pan P, Yi M, Cheng L, Zhou H, Liu L, Aili A, Liu Y, Peng L, Pu J, Zhou H. Characteristics, treatments, in-hospital and long-term outcomes among inpatients with acute exacerbation of chronic obstructive pulmonary disease in China: sex differences in a large cohort study. BMC Pulm Med 2024; 24:125. [PMID: 38468263 DOI: 10.1186/s12890-024-02948-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Accepted: 03/04/2024] [Indexed: 03/13/2024] Open
Abstract
BACKGROUND Data related to the characteristics, treatments and clinical outcomes of acute exacerbation of chronic obstructive pulmonary disease (AECOPD) patients in China are limited, and sex differences are still a neglected topic. METHODS The patients hospitalized for AECOPD were prospectively enrolled from ten medical centers in China between September 2017 and July 2021. Patients from some centers received follow-up for 3 years. Data regarding the characteristics, treatments and in-hospital and long-term clinical outcomes from male and female AECOPD patients included in the cohort were analyzed and compared. RESULTS In total, 14,007 patients with AECOPD were included in the study, and 11,020 (78.7%) were males. Compared with males, female patients were older (74.02 ± 10.79 vs. 71.86 ± 10.23 years, P < 0.001), and had more comorbidities (2.22 ± 1.64 vs. 1.73 ± 1.56, P < 0.001), a higher frequency of altered mental status (5.0% vs. 2.9%, P < 0.001), lower diastolic blood pressure (78.04 ± 12.96 vs. 79.04 ± 12.47 mmHg, P < 0.001). In addition, there were also significant sex differences in a range of laboratory and radiographic findings. Females were more likely to receive antibiotics, high levels of respiratory support and ICU admission than males. The in-hospital and 3-year mortality were not significantly different between males and females (1.4% vs. 1.5%, P = 0.711; 35.3% vs. 31.4%, P = 0.058), while female smokers with AECOPD had higher in-hospital mortality than male smokers (3.3% vs. 1.2%, P = 0.002) and male smokers exhibited a trend toward higher 3-year mortality compared to female smokers (40.7% vs. 33.1%, P = 0.146). CONCLUSIONS In AECOPD inpatients, females and males had similar in-hospital and long-term survival despite some sex differences in clinical characteristics and treatments, but female smokers had significantly worse in-hospital outcomes than male smokers. CLINICAL TRIAL REGISTRATION Retrospectively registered, registration number is ChiCTR2100044625, date of registration 21/03/2021. URL: http://www.chictr.org.cn/showproj.aspx?proj=121626 .
Collapse
Affiliation(s)
- Jiarui Zhang
- Department of Respiratory and Critical Care Medicine, West China Hospital, Sichuan University, Guo-xue-xiang 37#, Wuhou District, 610041, Chengdu, Sichuan Province, China
| | - Qun Yi
- Department of Respiratory and Critical Care Medicine, West China Hospital, Sichuan University, Guo-xue-xiang 37#, Wuhou District, 610041, Chengdu, Sichuan Province, China
- Sichuan Cancer Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan Province, China
| | - Chen Zhou
- West China School of Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Yuanming Luo
- State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, Guangdong Province, China
| | - Hailong Wei
- Department of Respiratory and Critical Care Medicine, People's Hospital of Leshan, Leshan, Sichuan Province, China
| | - Huiqing Ge
- Department of Respiratory and Critical Care Medicine, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
| | - Huiguo Liu
- Department of Respiratory and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Jianchu Zhang
- Department of Respiratory and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Xianhua Li
- Department of Respiratory and Critical Care Medicine, the First People's Hospital of Neijiang City, Neijiang, Sichuan Province, China
| | - Xiufang Xie
- Department of Respiratory and Critical Care Medicine, the First People's Hospital of Neijiang City, Neijiang, Sichuan Province, China
| | - Pinhua Pan
- Department of Respiratory and Critical Care Medicine, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Mengqiu Yi
- Department of Emergency, First People's Hospital of Jiujiang, Jiujiang, Jiangxi Province, China
| | - Lina Cheng
- Department of Emergency, First People's Hospital of Jiujiang, Jiujiang, Jiangxi Province, China
| | - Hui Zhou
- Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Chengdu University, Chengdu, Sichuan Province, China
| | - Liang Liu
- Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Chengdu University, Chengdu, Sichuan Province, China
| | - Adila Aili
- Department of Respiratory and Critical Care Medicine, West China Hospital, Sichuan University, Guo-xue-xiang 37#, Wuhou District, 610041, Chengdu, Sichuan Province, China
| | - Yu Liu
- Department of Respiratory and Critical Care Medicine, West China Hospital, Sichuan University, Guo-xue-xiang 37#, Wuhou District, 610041, Chengdu, Sichuan Province, China
| | - Lige Peng
- Department of Respiratory and Critical Care Medicine, West China Hospital, Sichuan University, Guo-xue-xiang 37#, Wuhou District, 610041, Chengdu, Sichuan Province, China
| | - Jiaqi Pu
- Department of Respiratory and Critical Care Medicine, West China Hospital, Sichuan University, Guo-xue-xiang 37#, Wuhou District, 610041, Chengdu, Sichuan Province, China
| | - Haixia Zhou
- Department of Respiratory and Critical Care Medicine, West China Hospital, Sichuan University, Guo-xue-xiang 37#, Wuhou District, 610041, Chengdu, Sichuan Province, China.
| |
Collapse
|
18
|
Odimba U, Senthilselvan A, Farrell J, Gao Z. Sex-Specific Genetic Determinants of Asthma-COPD Phenotype and COPD in Middle-Aged and Older Canadian Adults: An Analysis of CLSA Data. COPD 2023; 20:233-247. [PMID: 37466093 DOI: 10.1080/15412555.2023.2229906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 05/22/2023] [Accepted: 06/20/2023] [Indexed: 07/20/2023]
Abstract
The etiology of sex differences in the risk of asthma-COPD phenotype and COPD is still not completely understood. Genetic and environmental risk factors are commonly believed to play an important role. This study aims to identify sex-specific genetic markers associated with asthma-COPD phenotype and COPD using the Canadian Longitudinal Study on Aging (CLSA) Baseline Comprehensive and Genomic data. There were a total of 1,415 COPD cases. Out of them, 504 asthma-COPD phenotype cases were identified. 20,524 participants without a diagnosis of asthma and COPD served as controls. We performed genome-wide SNP-by-sex interaction analysis. SNPs with an interaction p-value < 10-5 were included in a sex-stratified multivariable logistic regression for asthma-COPD phenotype and COPD outcomes. 18 and 28 SNPs had a significant interaction term p-value < 10-5 with sex in the regression analyses of asthma-COPD phenotype and COPD outcomes, respectively. Sex-stratified multivariable analysis of asthma-COPD phenotype showed that 7 SNPs in/near SMYD3, FHIT, ZNF608, RIMBP2, ZNF133, BPIFB1, and S100B loci were significant in males. Sex-stratified multivariable analysis of COPD showed that 8 SNPs in/near MAGI1, COX18, OSTC, ELOVL5, C7orf72 FGF14, and NKAIN4 were significant in males, and 4 SNPs in/near genes CAMTA1, SATB2, PDE10A, and LINC00908 were significant in females. An SNP in the ZPBP gene was associated with COPD in both males and females. Identification of sex-specific loci associated with asthma-COPD phenotype and COPD may offer valuable evidence toward a better understanding of the sex-specific differences in the pathophysiology of the diseases.
Collapse
Affiliation(s)
- Ugochukwu Odimba
- Clinical Epidemiology Unit, Division of Community Health and Humanities, Faculty of Medicine, Memorial University, St. John's, Canada
| | | | - Jamie Farrell
- Clinical Epidemiology Unit, Division of Community Health and Humanities, Faculty of Medicine, Memorial University, St. John's, Canada
- Faculty of Medicine, Health Sciences Centre (Respirology Department), Memorial University, St John's, Newfoundland and Labrador, Canada
| | - Zhiwei Gao
- Clinical Epidemiology Unit, Division of Community Health and Humanities, Faculty of Medicine, Memorial University, St. John's, Canada
| |
Collapse
|
19
|
Zhao J, Ma X, Li S, Liu C, Liu Y, Tan J, Yu L, Li X, Li W. Berberine hydrochloride ameliorates PM2.5-induced pulmonary fibrosis in mice through inhibiting oxidative stress and inflammatory. Chem Biol Interact 2023; 386:110731. [PMID: 37839514 DOI: 10.1016/j.cbi.2023.110731] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 09/18/2023] [Accepted: 09/25/2023] [Indexed: 10/17/2023]
Abstract
Elevated levels of respirable particulate matter (PM) have been strongly linked to disease incidence and mortality in population-based epidemiological studies. Berberine hydrochloride (BBR), an isoquinoline alkaloid found in Coptis chinensis, exhibits antipyretic, anti-inflammatory, and antioxidant properties. However, the protective effects and underlying mechanism of BBR against pulmonary fibrosis remain unclear. This study aimed to investigate the protective effect of BBR on lung tissue damage using a mouse model of PM2.5-induced pulmonary fibrosis. SPF grade C57BL/6 mice were randomly assigned to four groups, each consisting of 10 mice. The mice were pretreated with BBR (50 mg/kg) by gavage for 45 consecutive days. A tracheal drip of PM2.5 suspension (8 mg/kg) was administered once every three days for a total of 15 times to induce lung fibrosis. Moreover, the results demonstrated that PM2.5 was found to inhibit the PPARγ signaling pathway, increase ROS expression, upregulate protein levels of IL-6, IL-1β, TNF-α, as well as regulation of gene expression of STAT3 and SOCS3. Importantly, PM2.5 induced lung fibrosis by promoting collagen deposition, upregulating gene expression of fibrosis markers (TGF-β1, FN, α-SMA, COL-1, and COL-3), and downregulating E-cadherin expression. Remarkably, our findings suggest that these injuries could be reversed by BBR pretreatment. BBR acts as a PPARγ agonist in PM2.5-induced pulmonary fibrosis, activating the PPARγ signaling pathway to mitigate oxidative and inflammatory factor-mediated lung injury. This study provides valuable insights for the future prevention and treatment of pulmonary fibrosis.
Collapse
Affiliation(s)
- Jiali Zhao
- School of Public Health, Weifang Medical University, Weifang, 261053, China
| | - Xuan Ma
- School of Public Health, Weifang Medical University, Weifang, 261053, China
| | - Siqi Li
- School of Public Health, Weifang Medical University, Weifang, 261053, China
| | - Chen Liu
- School of Public Health, Weifang Medical University, Weifang, 261053, China
| | - Yumei Liu
- School of Public Health, Weifang Medical University, Weifang, 261053, China; Weifang Key Laboratory of Health Inspection and Quarantine, Weifang, 261053, China
| | - Jinfeng Tan
- Weifang Environmental Monitoring Station, Weifang, 261044, China
| | - Li Yu
- School of Basic Medicine, Weifang Medical University, Weifang, 261053, China
| | - Xiaohong Li
- School of Public Health, Weifang Medical University, Weifang, 261053, China; "Healthy Shandong" Major Social Risk Prediction and Management Collaborative Innovation Center, Weifang, 261053, China; Weifang Key Laboratory of Health Inspection and Quarantine, Weifang, 261053, China.
| | - Wanwei Li
- School of Public Health, Weifang Medical University, Weifang, 261053, China; "Healthy Shandong" Major Social Risk Prediction and Management Collaborative Innovation Center, Weifang, 261053, China; Weifang Key Laboratory of Health Inspection and Quarantine, Weifang, 261053, China.
| |
Collapse
|
20
|
Zhang Y, Barupal DK, Fan S, Gao B, Zhu C, Flenniken AM, McKerlie C, Nutter LMJ, Lloyd KCK, Fiehn O. Sexual Dimorphism of the Mouse Plasma Metabolome Is Associated with Phenotypes of 30 Gene Knockout Lines. Metabolites 2023; 13:947. [PMID: 37623890 PMCID: PMC10456929 DOI: 10.3390/metabo13080947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 08/03/2023] [Accepted: 08/08/2023] [Indexed: 08/26/2023] Open
Abstract
Although metabolic alterations are observed in many monogenic and complex genetic disorders, the impact of most mammalian genes on cellular metabolism remains unknown. Understanding the effect of mouse gene dysfunction on metabolism can inform the functions of their human orthologues. We investigated the effect of loss-of-function mutations in 30 unique gene knockout (KO) lines on plasma metabolites, including genes coding for structural proteins (11 of 30), metabolic pathway enzymes (12 of 30) and protein kinases (7 of 30). Steroids, bile acids, oxylipins, primary metabolites, biogenic amines and complex lipids were analyzed with dedicated mass spectrometry platforms, yielding 827 identified metabolites in male and female KO mice and wildtype (WT) controls. Twenty-two percent of 23,698 KO versus WT comparison tests showed significant genotype effects on plasma metabolites. Fifty-six percent of identified metabolites were significantly different between the sexes in WT mice. Many of these metabolites were also found to have sexually dimorphic changes in KO lines. We used plasma metabolites to complement phenotype information exemplified for Dhfr, Idh1, Mfap4, Nek2, Npc2, Phyh and Sra1. The association of plasma metabolites with IMPC phenotypes showed dramatic sexual dimorphism in wildtype mice. We demonstrate how to link metabolomics to genotypes and (disease) phenotypes. Sex must be considered as critical factor in the biological interpretation of gene functions.
Collapse
Affiliation(s)
- Ying Zhang
- West Coast Metabolomics Center, University of California Davis, Davis, CA 95616, USA
- Department of Chemistry, University of California Davis, Davis, CA 95616, USA
| | - Dinesh K. Barupal
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA;
| | - Sili Fan
- West Coast Metabolomics Center, University of California Davis, Davis, CA 95616, USA
| | - Bei Gao
- School of Marine Sciences, Nanjing University of Information Science and Technology, Nanjing 210044, China
| | - Chao Zhu
- College of Medicine & Nursing, Dezhou University, Dezhou 253023, China
| | - Ann M. Flenniken
- The Centre for Phenogenomics, Toronto, ON M5T 3H7, Canada; (A.M.F.); (C.M.); (L.M.J.N.)
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON M5G 1X5, Canada
| | - Colin McKerlie
- The Centre for Phenogenomics, Toronto, ON M5T 3H7, Canada; (A.M.F.); (C.M.); (L.M.J.N.)
- The Hospital for Sick Children, Toronto, ON M5G 1X8, Canada
| | - Lauryl M. J. Nutter
- The Centre for Phenogenomics, Toronto, ON M5T 3H7, Canada; (A.M.F.); (C.M.); (L.M.J.N.)
- The Hospital for Sick Children, Toronto, ON M5G 1X8, Canada
| | - Kevin C. Kent Lloyd
- Department of Surgery, School of Medicine, and Mouse Biology Program, University of California Davis, Davis, CA 95616, USA;
| | - Oliver Fiehn
- West Coast Metabolomics Center, University of California Davis, Davis, CA 95616, USA
| |
Collapse
|
21
|
Burrowes KS, Ruppage M, Lowry A, Zhao D. Sex matters: the frequently overlooked importance of considering sex in computational models. Front Physiol 2023; 14:1186646. [PMID: 37520817 PMCID: PMC10374267 DOI: 10.3389/fphys.2023.1186646] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 06/27/2023] [Indexed: 08/01/2023] Open
Abstract
Personalised medicine and the development of a virtual human or a digital twin comprises visions of the future of medicine. To realise these innovations, an understanding of the biology and physiology of all people are required if we wish to apply these technologies at a population level. Sex differences in health and biology is one aspect that has frequently been overlooked, with young white males being seen as the "average" human being. This has not been helped by the lack of inclusion of female cells and animals in biomedical research and preclinical studies or the historic exclusion, and still low in proportion, of women in clinical trials. However, there are many known differences in health between the sexes across all scales of biology which can manifest in differences in susceptibility to diseases, symptoms in a given disease, and outcomes to a given treatment. Neglecting these important differences in the development of any health technologies could lead to adverse outcomes for both males and females. Here we highlight just some of the sex differences in the cardio-respiratory systems with the goal of raising awareness that these differences exist. We discuss modelling studies that have considered sex differences and touch on how and when to create sex-specific models. Scientific studies should ensure sex differences are included right from the study planning phase and results reported using sex as a biological variable. Computational models must have sex-specific versions to ensure a movement towards personalised medicine is realised.
Collapse
Affiliation(s)
- K. S. Burrowes
- Auckland Bioengineering Institute, University of Auckland, Auckland, New Zealand
| | - M. Ruppage
- Department of Nursing, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - A. Lowry
- Auckland Bioengineering Institute, University of Auckland, Auckland, New Zealand
| | - D. Zhao
- Auckland Bioengineering Institute, University of Auckland, Auckland, New Zealand
| |
Collapse
|
22
|
Terada S, Tanabe N, Maetani T, Shiraishi Y, Sakamoto R, Shima H, Oguma T, Sato A, Kanasaki M, Masuda I, Sato S, Hirai T. Association of age with computed tomography airway tree morphology in male and female never smokers without lung disease history. Respir Med 2023; 214:107278. [PMID: 37196749 DOI: 10.1016/j.rmed.2023.107278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 04/09/2023] [Accepted: 05/06/2023] [Indexed: 05/19/2023]
Abstract
BACKGROUND Sex and aging may affect the airway tree structure in patients with airway diseases and even healthy subjects. Using chest computed tomography (CT), this study sought to determine whether age is associated with airway morphological features differently in healthy males and females. METHODS This retrospective cross-sectional study consecutively incorporated lung cancer screening CT data of asymptomatic never smokers (n = 431) without lung disease history. Luminal areas were measured at the trachea, main bronchi, bronchus intermedius, segmental and subsegmental bronchus, and the ratio of their geometric mean to total lung volume (airway-to-lung size ratio, ALR) was determined. Airway fractal dimension (AFD) and total airway count (TAC) were calculated for the segmented airway tree resolved on CT. RESULTS The lumen areas of the trachea, main bronchi, segmental and subsegmental airways, AFD and TAC visible on CT were smaller in females (n = 220) than in males (n = 211) after adjusting for age, height, and body mass index, while ALR or count of the 1st to 5th generation airways did not differ. Furthermore, in males but not in females, older age was associated with larger lumen sizes of the main bronchi, segmental and subsegmental airways, and ALR. In contrast, neither male nor female had any associations between age and AFD or TAC on CT. CONCLUSION Older age was associated with larger lumen size of the relatively central airways and ALR exclusively in males. Aging may have a more profound effect on airway lumen tree caliber in males than in females.
Collapse
Affiliation(s)
- Satoru Terada
- Department of Respiratory Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan.
| | - Naoya Tanabe
- Department of Respiratory Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan; Rehabilitation Unit, Kyoto University Hospital, Kyoto, Japan.
| | - Tomoki Maetani
- Department of Respiratory Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan.
| | - Yusuke Shiraishi
- Department of Respiratory Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan.
| | - Ryo Sakamoto
- Department of Diagnostic Imaging and Nuclear Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan.
| | - Hiroshi Shima
- Department of Respiratory Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan.
| | - Tsuyoshi Oguma
- Department of Respiratory Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan.
| | - Atsuyasu Sato
- Department of Respiratory Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan.
| | | | - Izuru Masuda
- Medical Examination Center, Takeda Hospital, Kyoto, Japan; Department of Endocrinology, Metabolism and Hypertension Research, Clinical Research Institute, National Hospital Organization, Kyoto Medical Center, Japan.
| | - Susumu Sato
- Department of Respiratory Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan; Department of Respiratory Care and Sleep Control Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan.
| | - Toyohiro Hirai
- Department of Respiratory Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan.
| |
Collapse
|
23
|
Zhang Y, Zhang H, Su X, Wang Y, Gao G, Wang X, Zhang T. Analysis of influencing factors and a predictive model of small airway dysfunction in adults. BMC Pulm Med 2023; 23:141. [PMID: 37098545 PMCID: PMC10131465 DOI: 10.1186/s12890-023-02416-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Accepted: 04/04/2023] [Indexed: 04/27/2023] Open
Abstract
BACKGROUND Small airway dysfunction (SAD) is a widespread but less typical clinical manifestation of respiratory dysfunction. In lung diseases, SAD can have a higher-than-expected impact on lung function. The aim of this study was to explore risk factors for SAD and to establish a predictive model. METHODS We included 1233 patients in the pulmonary function room of TangDu Hospital from June 2021 to December 2021. We divided the subjects into a small airway disorder group and a non-small airway disorder group, and all participants completed a questionnaire. We performed univariate and multivariate analyses to identify the risk factors for SAD. Multivariate logistic regression was performed to construct the nomogram. The performance of the nomogram was assessed and validated by the Area under roc curve (AUC), calibration curves, and Decision curve analysis (DCA). RESULTS One. The risk factors for small airway disorder were advanced age (OR = 7.772,95% CI 2.284-26.443), female sex (OR = 1.545,95% CI 1.103-2.164), family history of respiratory disease (OR = 1.508,95% CI 1.069-2.126), history of occupational dust exposure (OR = 1.723,95% CI 1.177-2.521), history of smoking (OR = 1.732,95% CI 1.231-2.436), history of pet exposure (OR = 1.499,95% CI 1.065-2.110), exposure to O3 (OR = 1.008,95% CI 1.003-1.013), chronic bronchitis (OR = 1.947,95% CI 1.376-2.753), emphysema (OR = 2.190,95% CI 1.355-3.539) and asthma (OR = 7.287,95% CI 3.546-14.973). 2. The AUCs of the nomogram were 0.691 in the training set and 0.716 in the validation set. Both nomograms demonstrated favourable clinical consistency. 3.There was a dose‒response relationship between cigarette smoking and SAD; however, quitting smoking did not reduce the risk of SAD. CONCLUSION Small airway disorders are associated with age, sex, family history of respiratory disease, occupational dust exposure, smoking history, history of pet exposure, exposure to O3, chronic bronchitis, emphysema, and asthma. The nomogram based on the above results can effectively used in the preliminary risk prediction.
Collapse
Affiliation(s)
- Yifan Zhang
- Department of Thoracic Surgery, Tangdu Hospital, Air Force Medical University, Xi'an, 710032, China
| | - Haihua Zhang
- Department of Thoracic Surgery, Tangdu Hospital, Air Force Medical University, Xi'an, 710032, China
| | - Xuan Su
- Department of Respiratory Medicine, Tangdu Hospital, Air Force Medical University, Xi'an, 710032, China
| | - Ying Wang
- Department of Respiratory Medicine, Tangdu Hospital, Air Force Medical University, Xi'an, 710032, China
| | - Guizhou Gao
- Department of Thoracic Surgery, Tangdu Hospital, Air Force Medical University, Xi'an, 710032, China
| | - Xiaodong Wang
- Department of Thoracic Surgery, Tangdu Hospital, Air Force Medical University, Xi'an, 710032, China
| | - Tao Zhang
- Department of Thoracic Surgery, Tangdu Hospital, Air Force Medical University, Xi'an, 710032, China.
| |
Collapse
|
24
|
Reddy KD, Oliver BGG. Sexual dimorphism in chronic respiratory diseases. Cell Biosci 2023; 13:47. [PMID: 36882807 PMCID: PMC9993607 DOI: 10.1186/s13578-023-00998-5] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2023] [Accepted: 02/23/2023] [Indexed: 03/09/2023] Open
Abstract
Sex differences in susceptibility, severity, and progression are prevalent for various diseases in multiple organ systems. This phenomenon is particularly apparent in respiratory diseases. Asthma demonstrates an age-dependent pattern of sexual dimorphism. However, marked differences between males and females exist in other pervasive conditions such as chronic obstructive pulmonary disease (COPD) and lung cancer. The sex hormones estrogen and testosterone are commonly considered the primary factors causing sexual dimorphism in disease. However, how they contribute to differences in disease onset between males and females remains undefined. The sex chromosomes are an under-investigated fundamental form of sexual dimorphism. Recent studies highlight key X and Y-chromosome-linked genes that regulate vital cell processes and can contribute to disease-relevant mechanisms. This review summarises patterns of sex differences in asthma, COPD and lung cancer, highlighting physiological mechanisms causing the observed dimorphism. We also describe the role of the sex hormones and present candidate genes on the sex chromosomes as potential factors contributing to sexual dimorphism in disease.
Collapse
Affiliation(s)
- Karosham Diren Reddy
- Respiratory and Cellular Molecular Biology Group, Woolcock Institute of Medical Research, Glebe, NSW, 2037, Australia.
- School of Life Science, University of Technology Sydney, Ultimo, NSW, 2007, Australia.
| | - Brian Gregory George Oliver
- Respiratory and Cellular Molecular Biology Group, Woolcock Institute of Medical Research, Glebe, NSW, 2037, Australia
- School of Life Science, University of Technology Sydney, Ultimo, NSW, 2007, Australia
| |
Collapse
|
25
|
Cui J, Tang W, Wang W, Yi L, Teng F, Xu F, Li M, Ma M, Dong J. Acteoside alleviates asthma by modulating ROS-responsive NF-κB/MAPK signaling pathway. Int Immunopharmacol 2023. [DOI: 10.1016/j.intimp.2023.109806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/19/2023]
|
26
|
Odimba U, Senthilselvan A, Farrell J, Gao Z. Identification of Sex-Specific Genetic Polymorphisms Associated with Asthma in Middle-Aged and Older Canadian Adults: An Analysis of CLSA Data. J Asthma Allergy 2023; 16:553-566. [PMID: 37197194 PMCID: PMC10184860 DOI: 10.2147/jaa.s404670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Accepted: 04/29/2023] [Indexed: 05/19/2023] Open
Abstract
Purpose Asthma is a chronic heterogeneous respiratory disease resulting from a complex interplay between genetic variations and environmental exposures. There are sex disparities in the prevalence and severity of asthma in males and females. Asthma prevalence is higher in males during childhood but increases in females in adulthood. The mechanisms underlying these sex differences are not well understood; nevertheless, genetic variations, hormonal changes, and environmental influences are thought to play important roles. This study aimed to identify sex-specific genetic variants associated with asthma using CLSA genomic and questionnaire data. Methods First, we conducted a genome-wide SNP-by-sex interaction analysis on 23,323 individuals, examining 416,562 single nucleotide polymorphisms (SNPs) after quality control, followed by sex-stratified survey logistic regression of SNPs with interaction p-value less than 10¯5. Results Out of the 49 SNPs with interaction p-value less than 10-5, a sex-stratified survey logistic regression showed that five male-specific SNPs (rs6701638, rs17071077, rs254804, rs6013213, and rs2968822) in/near KIF26B, NMBR, PEPD, RTN4, and NFATC2 loci, and three female-specific SNPs (rs2968801, rs2864052, and rs9525931) in/near RTN4, and SERP2 loci were significantly associated with asthma after Bonferroni correction. An SNP (rs36213) in the EPHB1 gene was significantly associated with an increased risk of asthma in males [OR=1.35, 95% CI (1.14, 1.60)] but with a reduced risk of asthma in females [OR=0.84, 95% CI (0.76, 0.92)] after Bonferroni correction. Conclusion We discovered novel sex-specific genetic markers in/near the KIF26B, RTN4, EPHB1, NMBR, SERP2, PEPD, and NFATC2 genes that could potentially shed light on the sex differences in asthma susceptibility in males and females. Future mechanistic studies are required to understand better the underlying sex-related pathways of the identified loci in asthma development.
Collapse
Affiliation(s)
- Ugochukwu Odimba
- Clinical Epidemiology Unit, Division of Community Health and Humanities, Faculty of Medicine, Memorial University, St. John’s, Newfoundland and Labrador, Canada
| | | | - Jamie Farrell
- Clinical Epidemiology Unit, Division of Community Health and Humanities, Faculty of Medicine, Memorial University, St. John’s, Newfoundland and Labrador, Canada
- Faculty of Medicine, Health Science Centre (Respirology Department), Memorial University, St John’s, Newfoundland and Labrador, Canada
| | - Zhiwei Gao
- Clinical Epidemiology Unit, Division of Community Health and Humanities, Faculty of Medicine, Memorial University, St. John’s, Newfoundland and Labrador, Canada
- Correspondence: Zhiwei Gao, Clinical Epidemiology Unit, Division of Community Health and Humanities, Faculty of Medicine, Memorial University of Newfoundland, St. John’s, Newfoundland and Labrador, A1B 3V6, Canada, Tel +17098646523, Email
| |
Collapse
|
27
|
Ding K, Jiang W, Zhan W, Xiong C, Chen J, Wang Y, Jia H, Lei M. The therapeutic potential of quercetin for cigarette smoking-induced chronic obstructive pulmonary disease: a narrative review. Ther Adv Respir Dis 2023; 17:17534666231170800. [PMID: 37154390 PMCID: PMC10170608 DOI: 10.1177/17534666231170800] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/10/2023] Open
Abstract
Quercetin is a flavonoid with antioxidant and anti-inflammatory properties. Quercetin has potentially beneficial therapeutic effects for several diseases, including cigarette smoking-induced chronic obstructive pulmonary disease (CS-COPD). Many studies have shown that quercetin's antioxidant and anti-inflammatory properties have positive therapeutic potential for CS-COPD. In addition, quercetin's immunomodulatory, anti-cellular senescence, mitochondrial autophagy-modulating, and gut microbiota-modulating effects may also have therapeutic value for CS-COPD. However, there appears to be no review of the possible mechanisms of quercetin for treating CS-COPD. Moreover, the combination of quercetin with common therapeutic drugs for CS-COPD needs further refinement. Therefore, in this article, after introducing the definition and metabolism of quercetin, and its safety, we comprehensively presented the pathogenesis of CS-COPD related to oxidative stress, inflammation, immunity, cellular senescence, mitochondrial autophagy, and gut microbiota. We then reviewed quercetin's anti-CS-COPD effects, performed by influencing these mechanisms. Finally, we explored the possibility of using quercetin with commonly used drugs for treating CS-COPD, providing a basis for future screening of excellent drug combinations for treating CS-COPD. This review has provided meaningful information on quercetin's mechanisms and clinical use in treating CS-COPD.
Collapse
Affiliation(s)
- Kaixi Ding
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Wei Jiang
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Wenling Zhan
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Chunping Xiong
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jieling Chen
- Shehong Hospital of Traditional Chinese Medicine, Shehong, China
| | - Yu Wang
- Jiangsu Provincial Hospital of Traditional Chinese Medicine, Nanjing, China
| | - Huanan Jia
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China
| | - Ming Lei
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China
| |
Collapse
|
28
|
Knox-Brown B, Patel J, Potts J, Ahmed R, Aquart-Stewart A, Cherkaski HH, Denguezli M, Elbiaze M, Elsony A, Franssen FME, Ghobain MA, Harrabi I, Janson C, Jõgi R, Juvekar S, Lawin H, Mannino D, Mortimer K, Nafees AA, Nielsen R, Obaseki D, Paraguas SNM, Rashid A, Loh LC, Salvi S, Seemungal T, Studnicka M, Tan WC, Wouters EEFM, Barbara C, Gislason T, Gunasekera K, Burney P, Amaral AFS. Small airways obstruction and its risk factors in the Burden of Obstructive Lung Disease (BOLD) study: a multinational cross-sectional study. Lancet Glob Health 2023; 11:e69-e82. [PMID: 36521955 DOI: 10.1016/s2214-109x(22)00456-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 09/21/2022] [Accepted: 10/10/2022] [Indexed: 12/15/2022]
Abstract
BACKGROUND Small airways obstruction is a common feature of obstructive lung diseases. Research is scarce on small airways obstruction, its global prevalence, and risk factors. We aimed to estimate the prevalence of small airways obstruction, examine the associated risk factors, and compare the findings for two different spirometry parameters. METHODS The Burden of Obstructive Lung Disease study is a multinational cross-sectional study of 41 municipalities in 34 countries across all WHO regions. Adults aged 40 years or older who were not living in an institution were eligible to participate. To ensure a representative sample, participants were selected from a random sample of the population according to a predefined site-specific sampling strategy. We included participants' data in this study if they completed the core study questionnaire and had acceptable spirometry according to predefined quality criteria. We excluded participants with a contraindication for lung function testing. We defined small airways obstruction as either mean forced expiratory flow rate between 25% and 75% of the forced vital capacity (FEF25-75) less than the lower limit of normal or forced expiratory volume in 3 s to forced vital capacity ratio (FEV3/FVC ratio) less than the lower limit of normal. We estimated the prevalence of pre-bronchodilator (ie, before administration of 200 μg salbutamol) and post-bronchodilator (ie, after administration of 200 μg salbutamol) small airways obstruction for each site. To identify risk factors for small airways obstruction, we performed multivariable regression analyses within each site and pooled estimates using random-effects meta-analysis. FINDINGS 36 618 participants were recruited between Jan 2, 2003, and Dec 26, 2016. Data were collected from participants at recruitment. Of the recruited participants, 28 604 participants had acceptable spirometry and completed the core study questionnaire. Data were available for 26 443 participants for FEV3/FVC ratio and 25 961 participants for FEF25-75. Of the 26 443 participants included, 12 490 were men and 13 953 were women. Prevalence of pre-bronchodilator small airways obstruction ranged from 5% (34 of 624 participants) in Tartu, Estonia, to 34% (189 of 555 participants) in Mysore, India, for FEF25-75, and for FEV3/FVC ratio it ranged from 5% (31 of 684) in Riyadh, Saudi Arabia, to 31% (287 of 924) in Salzburg, Austria. Prevalence of post-bronchodilator small airways obstruction was universally lower. Risk factors significantly associated with FEV3/FVC ratio less than the lower limit of normal included increasing age, low BMI, active and passive smoking, low level of education, working in a dusty job for more than 10 years, previous tuberculosis, and family history of chronic obstructive pulmonary disease. Results were similar for FEF25-75, except for increasing age, which was associated with reduced odds of small airways obstruction. INTERPRETATION Despite the wide geographical variation, small airways obstruction is common and more prevalent than chronic airflow obstruction worldwide. Small airways obstruction shows the same risk factors as chronic airflow obstruction. However, further research is required to investigate whether small airways obstruction is also associated with respiratory symptoms and lung function decline. FUNDING National Heart and Lung Institute and Wellcome Trust. TRANSLATIONS For the Dutch, Estonian, French, Icelandic, Malay, Marathi, Norwegian, Portuguese, Swedish and Urdu translations of the abstract see Supplementary Materials section.
Collapse
Affiliation(s)
- Ben Knox-Brown
- National Heart and Lung Institute, Imperial College London, London, UK.
| | - Jaymini Patel
- National Heart and Lung Institute, Imperial College London, London, UK
| | - James Potts
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Rana Ahmed
- Epidemiological Laboratory for Public Health, Research and Development, Khartoum, Sudan
| | | | - Hamid Hacene Cherkaski
- Department of Pneumology, Faculty of Medicine Annaba, University Badji Mokhtar of Annaba, Annaba, Algeria
| | - Meriam Denguezli
- Faculté de Médecine Dentaire de Monastir, Université de Monastir, Monastir, Tunisia
| | - Mohammed Elbiaze
- Department of Respiratory Medicine, Faculty of Medicine, Mohammed Ben Abdellah University, University Hospital, Fes, Morocco
| | - Asma Elsony
- Epidemiological Laboratory for Public Health, Research and Development, Khartoum, Sudan
| | - Frits M E Franssen
- Department of Respiratory Medicine, Maastricht University Medical Centre, Maastricht, Netherlands; Department of Research and Education, CIRO, Horn, Netherlands
| | - Mohammed Al Ghobain
- King Abdullah International Medical Research Centre, King Saud Bin Abdulaziz University for Health Sciences, King Abdulaziz Medical City, Ministry of National Guard-Health Affairs, Riyadh, Saudi Arabia
| | - Imed Harrabi
- Ibn El Jazzar Faculty of Medicine of Sousse, University of Sousse, Sousse, Tunisia
| | - Christer Janson
- Department of Medical Sciences: Respiratory, Allergy and Sleep Research, Uppsala University, Uppsala, Sweden
| | - Rain Jõgi
- Lung Clinic, Tartu University Hospital, Tartu, Estonia
| | - Sanjay Juvekar
- Vadu Rural Health Program, KEM Hospital Research Centre, Pune, India
| | - Herve Lawin
- Unit of Teaching and Research in Occupational and Environmental Health, University of Abomey-Calavi, Cotonou, Benin
| | - David Mannino
- University of Kentucky, Lexington, KY, USA; COPD Foundation, Miami, FL, USA
| | - Kevin Mortimer
- University of Cambridge, Cambridge, UK; Liverpool University Hospitals NHS Foundation Trust, Liverpool, UK
| | - Asaad Ahmed Nafees
- Department of Community Health Sciences, Aga Khan University, Karachi, Pakistan
| | - Rune Nielsen
- Department of Thoracic Medicine, Haukeland University Hospital, Bergen, Norway; Department of Clinical Science, University of Bergen, Bergen, Norway
| | | | - Stefanni Nonna M Paraguas
- Philippine College of Chest Physicians, Quezon City, Philippines; Philippine Heart Centre, Quezon City, Philippines
| | | | - Li-Cher Loh
- RCSI and UCD Malaysia Campus, Penang, Malaysia
| | - Sundeep Salvi
- Pulmocare Research and Education Foundation, Pune, India; Symbiosis International (Deemed University), Pune, India
| | - Terence Seemungal
- Faculty of Medical Sciences, University of the West Indies, Trinidad and Tobago
| | - Michael Studnicka
- University Clinic for Pneumology, Paracelsus Medical University Salzburg, Salzburg, Austria
| | - Wan C Tan
- Centre for Heart Lung Innovation, University of British Columbia, Vancouver, BC, Canada
| | - Emiel E F M Wouters
- Department of Respiratory Medicine, Maastricht University Medical Centre, Maastricht, Netherlands; Ludwig Boltzmann Institute for Lung Health, Vienna, Austria
| | - Cristina Barbara
- Instituto de Saúde Ambiental, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal; Serviço de Pneumologia, Centro Hospitalar Universitário Lisboa Norte, Lisbon, Portugal
| | - Thorarinn Gislason
- Department of Sleep, Landspitali University Hospital, Reykjavik, Iceland; Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | - Kirthi Gunasekera
- Medical Research Institute, Central Chest Clinic, Colombo, Sri Lanka
| | - Peter Burney
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Andre F S Amaral
- National Heart and Lung Institute, Imperial College London, London, UK
| | | |
Collapse
|
29
|
Kaleem Ullah M, Malamardi S, Siddaiah JB, A T, Prashant A, Vishwanath P, Riley LW, Madhivanan P, Mahesh PA. Trends in the Bacterial Prevalence and Antibiotic Resistance Patterns in the Acute Exacerbation of Chronic Obstructive Pulmonary Disease in Hospitalized Patients in South India. Antibiotics (Basel) 2022; 11:1577. [PMID: 36358232 PMCID: PMC9686600 DOI: 10.3390/antibiotics11111577] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 11/02/2022] [Accepted: 11/07/2022] [Indexed: 11/11/2022] Open
Abstract
Exacerbation due to antimicrobial-drug-resistant bacteria among chronic obstructive pulmonary disease (AECOPD) patients contributes to mortality and morbidity. We examined the prevalence of the bacterial organisms and trends in drug resistance in AECOPD. In this retrospective study, between January 2016 to December 2020, among 3027 AECOPD patients, 432 (14.3%) had bacteria isolated. The regression and generalized estimating equations (GEE) were used for trends in the resistance patterns over five years, adjusting for age, gender, and comorbidities. Klebsiella pneumoniae (32.4%), Pseudomonas aeruginosa (17.8%), Acinetobacter baumannii (14.4%), Escherichia coli (10.4%), and Staphylococcus aureus (2.5%) were common. We observed high levels of drug resistance in AECOPD patients admitted to ICU (87.8%) and non-ICU (86.5%). A Cox proportional hazard analysis, observed infection with Acinetobacter baumannii and female sex as independent predictors of mortality. Acinetobacter baumannii had 2.64 (95% confidence interval (CI): 1.08−6.43) higher odds of death, compared to Klebsiella pneumoniae. Females had 2.89 (95% CI: 1.47−5.70) higher odds of death, compared to males. A high proportion of bacterial AECOPD was due to drug-resistant bacteria. An increasing trend in drug resistance was observed among females.
Collapse
Affiliation(s)
- Mohammed Kaleem Ullah
- Centre for Excellence in Molecular Biology and Regenerative Medicine, Department of Biochemistry, JSS Medical College, JSS Academy of Higher Education & Research, Mysore 570015, Karnataka, India
- Division of Infectious Disease and Vaccinology, School of Public Health, University of California, Berkeley, CA 94720, USA
| | - Sowmya Malamardi
- Department of Respiratory Medicine, JSS Medical College, JSS Academy of Higher Education & Research, Mysuru 570015, Karnataka, India
- School of Psychology & Public Health, College of Science Health and Engineering, La Trobe University, Melbourne 3086, Australia
| | - Jayaraj Biligere Siddaiah
- Department of Respiratory Medicine, JSS Medical College, JSS Academy of Higher Education & Research, Mysuru 570015, Karnataka, India
| | - Tejashree A
- Department of Microbiology, JSS Medical College, JSS Academy of Higher Education & Research, Mysuru 570015, Karnataka, India
| | - Akila Prashant
- Centre for Excellence in Molecular Biology and Regenerative Medicine, Department of Biochemistry, JSS Medical College, JSS Academy of Higher Education & Research, Mysore 570015, Karnataka, India
| | - Prashant Vishwanath
- Centre for Excellence in Molecular Biology and Regenerative Medicine, Department of Biochemistry, JSS Medical College, JSS Academy of Higher Education & Research, Mysore 570015, Karnataka, India
| | - Lee W. Riley
- Division of Infectious Disease and Vaccinology, School of Public Health, University of California, Berkeley, CA 94720, USA
| | - Purnima Madhivanan
- Department of Health Promotion Sciences, Mel & Enid Zuckerman College of Public Health, University of Arizona, Tucson, AZ 85724, USA
- Division of Infectious Diseases, College of Medicine, University of Arizona, Tucson, AZ 85724, USA
- Public Health Research Institute of India, Mysuru 570020, Karnataka, India
| | - Padukudru Anand Mahesh
- Department of Respiratory Medicine, JSS Medical College, JSS Academy of Higher Education & Research, Mysuru 570015, Karnataka, India
| |
Collapse
|
30
|
Alobaidi NY, Almeshari M, Stockley J, Stockley RA, Sapey E. Small airway function measured using forced expiratory flow between 25% and 75% of vital capacity and its relationship to airflow limitation in symptomatic ever-smokers: a cross-sectional study. BMJ Open Respir Res 2022; 9:9/1/e001385. [PMID: 36202407 PMCID: PMC9540854 DOI: 10.1136/bmjresp-2022-001385] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Accepted: 09/17/2022] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND Chronic obstructive pulmonary disease (COPD) is diagnosed and its severity graded by traditional spirometric parameters (forced expiratory volume in 1 s (FEV1)/forced vital capacity (FVC) and FEV1, respectively) but these parameters are considered insensitive for identifying early pathology. Measures of small airway function, including forced expiratory flow between 25% and 75% of vital capacity (FEF25-75), may be more valuable in the earliest phases of COPD. This study aimed to determine the prevalence of low FEF25-75 in ever-smokers with and without airflow limitation (AL) and to determine whether FEF25-75 relates to AL severity. METHOD A retrospective analysis of lung function data of 1458 ever-smokers suspected clinically of having COPD. Low FEF25-75 was defined by z-score<-0.8345 and AL was defined by FEV1/FVC z-scores<-1.645. The severity of AL was evaluated using FEV1 z-scores. Participants were placed into three groups: normal FEF25-75/ no AL (normal FEF25-75/AL-); low FEF25-75/ no AL (low FEF25-75/AL-) and low FEF25-75/ AL (low FEF25-75/AL+). RESULTS Low FEF25-75 was present in 99.9% of patients with AL, and 50% of those without AL. Patients in the low FEF25-75/AL- group had lower spirometric measures (including FEV1 FEF25-75/FVC and FEV3/FVC) than those in the normal FEF25-75/AL- group. FEF25-75 decreased with AL severity. A logistic regression model demonstrated that in the absence of AL, the presence of low FEF25-75 was associated with lower FEV1 and FEV1/FVC even when smoking history was accounted for. CONCLUSIONS Low FEF25-75 is a physiological trait in patients with conventional spirometric AL and likely reflects early evidence of impairment in the small airways when spirometry is within the 'normal range'. FEF25-75 likely identifies a group of patients with early evidence of pathological lung damage who warrant careful monitoring and reinforced early intervention to abrogate further lung injury.
Collapse
Affiliation(s)
- Nowaf Y Alobaidi
- Birmingham Acute Care Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
- Respiratory Therapy Department, College of Applied Medical Sciences, King Saud bin Abdulaziz University for Health Sciences, Alahsa, Saudi Arabia
- King Abdullah International Medical Research Center, Alahsa, Saudi Arabia
| | - Mohammed Almeshari
- Birmingham Acute Care Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
- Rehabilitation Health Sciences Department, College of Applied Medical Sciences, King Saud University, Riyadh, Saudi Arabia
| | - James Stockley
- Lung Function & Sleep Department, Respiratory Medicine, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - Robert Andrew Stockley
- Lung Function & Sleep Department, Respiratory Medicine, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - Elizabeth Sapey
- Birmingham Acute Care Research Group, Institute of Inflammation and Ageing, University of Birmingham College of Medical and Dental Sciences, Birmingham, UK
- Acute Medicine, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| |
Collapse
|
31
|
Miao Y, Wu J, Wu R, Wang E, Wang J. Circ_0040929 Serves as Promising Biomarker and Potential Target for Chronic Obstructive Pulmonary Disease. Int J Chron Obstruct Pulmon Dis 2022; 17:2079-2092. [PMID: 36101791 PMCID: PMC9464637 DOI: 10.2147/copd.s364553] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 07/04/2022] [Indexed: 11/23/2022] Open
Abstract
Background Circular RNAs (circRNAs) can act as essential regulators in many diseases, including chronic obstructive pulmonary disease (COPD). We aimed to explore the role and underlying mechanism of circ_0040929 in COPD. Methods A cellular model of COPD was constructed by treating human bronchial epithelial cells (16HBE) with cigarette smoke extract (CSE). The levels of circ_0040929, microRNA-515-5p (miR-515-5p) and insulin-like growth factor-binding protein 3 (IGFBP3) were measured by quantitative real-time PCR. Cell proliferation was assessed by Cell Counting Kit-8 and 5-ethynyl-2'-deoxyuridine assays. Cell apoptosis was evaluated by flow cytometry. Protein expression was measured using Western blot assay. The levels of inflammatory factors and airway remodeling were assayed via enzyme-linked immunosorbent assay. The interaction between miR-515-5p and circ_0040929/IGFBP3 was confirmed by dual-luciferase reporter, RNA pull-down and RNA immunoprecipitation assays. Exosomes were detected using transmission electron microscopy. Results Circ_0040929 expression and IGFBP3 expression were upregulated in the serum of smokers (n = 22) compared to non-smokers (n = 22) and more significantly upregulated in the serum of COPD patients (n = 22). However, miR-515-5p expression was decreased in the serum of smokers compared to non-smokers and further reduced in the serum of COPD. Circ_0040929 knockdown attenuated CSE-induced cell injury by increasing proliferation and reducing apoptosis, inflammation, and airway remodeling in 16HBE cells. MiR-515-5p was a direct target of circ_0040929, and miR-515-5p inhibition reversed the effect of circ_0040929 knockdown in CSE-treated 16HBE cells. IGFBP3 was a direct target of miR-515-5p, and miR-515-5p overexpression alleviated CSE-induced cell injury via targeting IGFBP3. Moreover, circ_0040929 regulated IGFBP3 expression by targeting miR-515-5p. Importantly, circ_0040929 was upregulated in serum exosomes from COPD patients. Conclusion Circ_0040929 played a promoting role in CSE-induced COPD by regulating miR-515-5p/IGFBP3 axis, suggesting that it might be a novel potential target for COPD treatment.
Collapse
Affiliation(s)
- Yi Miao
- Department of Respiratory Medicine, Shaanxi Provincial People's Hospital, Xi'an City, 710068, People's Republic of China
| | - Junfang Wu
- Department of Respiratory Medicine, Shaanxi Provincial People's Hospital, Xi'an City, 710068, People's Republic of China
| | - Runmiao Wu
- Department of Respiratory Medicine, Shaanxi Provincial People's Hospital, Xi'an City, 710068, People's Republic of China
| | - Enguang Wang
- Department of Respiratory and Critical Care, the Fifth Affiliated Hospital of Xinjiang Medical University, Urumqi City, 830000, People's Republic of China
| | - Jing Wang
- Department of Clinical Laboratory, Shaanxi Provincial People's Hospital, Xi'an City, 710068, People's Republic of China
| |
Collapse
|
32
|
Sodhi A, Pisani M, Glassberg MK, Bourjeily G, D'Ambrosio C. Sex and Gender in Lung Disease and Sleep Disorders: A State-of-the-Art Review. Chest 2022; 162:647-658. [PMID: 35300976 PMCID: PMC9808608 DOI: 10.1016/j.chest.2022.03.006] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 03/08/2022] [Accepted: 03/08/2022] [Indexed: 01/13/2023] Open
Abstract
The terms sex and gender often are used interchangeably, but have specific meaning when it comes to their effects on lung disease. Ample evidence is now available that sex and gender affect the incidence, susceptibility, presentation, diagnosis, and severity of many lung diseases. Some conditions are more prevalent in women, such as asthma. Other conditions are seen almost exclusively in women, like lymphangioleiomyomatosis. Some life stages-such as pregnancy-are unique to women and can affect the onset and course of lung disease. Clinical presentation may differ as well, such as higher number of exacerbations experienced by women with COPD and greater cardiovascular morbidity in women with sleep-disordered breathing. In addition, response to therapy and medication safety may also differ by sex, and yet, pharmacogenomic factors often are not addressed adequately in clinical trials. Various aspects of lung and sleep biology and pathobiology are impacted by female sex and female reproductive transitions. Differential gene expression or organ development can be impacted by these biological differences. Understanding these differences is the first step in moving toward precision medicine for women. This article is a state-of-the-art review of specific effects of sex and gender focused on epidemiology, disease presentation, risk factors, and management of lung diseases. Pathobiological mechanisms explaining sex differences in these diseases are beyond the scope of this article. We review the literature and focus on recent guidelines about using sex and gender in research. We also review sex and gender differences in lung diseases.
Collapse
Affiliation(s)
- Amik Sodhi
- Division of Allergy, Pulmonary and Critical Care Medicine, University of Wisconsin, Madison, WI
| | - Margaret Pisani
- Division of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, CT
| | - Marilyn K Glassberg
- Division of Pulmonary, Critical Care and Sleep Medicine, University of Arizona College of Medicine Phoenix, Phoenix, AR
| | - Ghada Bourjeily
- Division of Pulmonary, Critical Care and Sleep Medicine, Brown University, Providence, RI
| | - Carolyn D'Ambrosio
- Division of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, CT.
| |
Collapse
|
33
|
Wei D, Wang Q, Liu S, Tan X, Chen L, Tu R, Liu Q, Jia Y, Liu S. Influences of Two FEV1 Reference Equations (GLI-2012 and GIRH-2017) on Airflow Limitation Classification Among COPD Patients. Int J Chron Obstruct Pulmon Dis 2022; 17:2053-2065. [PMID: 36081764 PMCID: PMC9447406 DOI: 10.2147/copd.s373834] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Accepted: 08/17/2022] [Indexed: 11/23/2022] Open
Abstract
Objective Methods Results Conclusion
Collapse
Affiliation(s)
- Dafei Wei
- Department of Pediatrics, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, People’s Republic of China
| | - Qi Wang
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, People’s Republic of China
| | - Shasha Liu
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, People’s Republic of China
| | - Xiaowu Tan
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, People’s Republic of China
| | - Lin Chen
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, People’s Republic of China
| | - Rongfang Tu
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, People’s Republic of China
| | - Qing Liu
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, People’s Republic of China
| | - Yuanhang Jia
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, People’s Republic of China
| | - Sha Liu
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, People’s Republic of China
- Correspondence: Sha Liu, Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, People’s Republic of China, Email
| |
Collapse
|
34
|
Hernandez L, Laucyte-Cibulskiene A, Ward LJ, Kautzky-Willer A, Herrero MT, Norris CM, Raparelli V, Pilote L, Stenvinkel P, Kublickiene K. Gender dimension in cardio-pulmonary continuum. Front Cardiovasc Med 2022; 9:916194. [PMID: 36003909 PMCID: PMC9393639 DOI: 10.3389/fcvm.2022.916194] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 07/11/2022] [Indexed: 11/17/2022] Open
Abstract
Cardio-pulmonary diseases, which were once regarded as a man's illness, have been one of the leading causes of morbidity and mortality for both men and women in many countries in recent years. Both gender and sex influence the functional and structural changes in the human body and therefore play an important role in disease clinical manifestation, treatment choice, and/or response to treatment and prognosis of health outcomes. The gender dimension integrates sex and gender analysis in health sciences and medical research, however, it is still relatively overlooked suggesting the need for empowerment in the medical research community. Latest advances in the field of cardiovascular research have provided supportive evidence that the application of biological variables of sex has led to the understanding that heart disease in females may have different pathophysiology compared to males, particularly in younger adults. It has also resulted in new diagnostic techniques and a better understanding of symptomatology, while gender analysis has informed more appropriate risk stratification and prevention strategies. The existing knowledge in the pulmonary field shows the higher prevalence of pulmonary disorders among females, however, the role of gender as a socio-cultural construct has yet to be explored for the implementation of targeted interventions. The purpose of this review is to introduce the concept of gender dimension and its importance for the cardiopulmonary continuum with a focus on shared pathophysiology and disease presentation in addition to interrelation with chronic kidney disease. The review presents basic knowledge of what gender dimension means, and the application of sex and gender aspects in cardiovascular medicine with a specific focus on early pulmonary development, pulmonary hypertension, and chronic obstructive pulmonary disease (COPD). Early vascular aging and inflammation have been presented as a potential pathophysiological link, with further interactions between the cardiopulmonary continuum and chronic kidney disease. Finally, implications for potential future research have been provided to increase the impact of gender dimension on research excellence that would add value to everybody, foster toward precision medicine and ultimately improve human health.
Collapse
Affiliation(s)
- Leah Hernandez
- Division of Renal Medicine, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| | - Agne Laucyte-Cibulskiene
- Division of Renal Medicine, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
- Department of Nephrology, Lund University, Skåne University Hospital, Malmö, Sweden
| | - Liam J. Ward
- Division of Renal Medicine, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
- Department of Forensic Genetics and Forensic Toxicology, National Board of Forensic Medicine, Linköping, Sweden
| | - Alexandra Kautzky-Willer
- Division of Endocrinology and Metabolism, Department of Medicine III, Medical University of Vienna, Vienna, Austria
| | - Maria-Trinidad Herrero
- Clinical and Experimental Neuroscience, Institutes for Aging Research and Bio-Health Research of Murcia, School of Medicine, University of Murcia, Murcia, Spain
| | - Colleen M. Norris
- Faculty of Nursing, University of Alberta, Edmonton, AB, Canada
- Cardiovascular and Stroke Strategic Clinical Network, Alberta Health Services, Edmonton, AB, Canada
| | - Valeria Raparelli
- Faculty of Nursing, University of Alberta, Edmonton, AB, Canada
- Department of Translational Medicine, University of Ferrara, Ferrara, Italy
- University Center for Studies on Gender Medicine, University of Ferrara, Ferrara, Italy
| | - Louise Pilote
- Division of Clinical Epidemiology, Research Institute of McGill University Health Centre, McGill University, Montreal, QC, Canada
| | - Peter Stenvinkel
- Division of Renal Medicine, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| | - Karolina Kublickiene
- Division of Renal Medicine, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| | | |
Collapse
|
35
|
Milad N, Pineault M, Bouffard G, Maranda-Robitaille M, Lechasseur A, Beaulieu MJ, Aubin S, Jensen BAH, Morissette MC. Recombinant human β-defensin 2 delivery improves smoking-associated lung neutrophilia and bacterial exacerbation. Am J Physiol Lung Cell Mol Physiol 2022; 323:L37-L47. [DOI: 10.1152/ajplung.00027.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Treatment of the cigarette smoke-associated lung disease has largely focused on broad-spectrum anti-inflammatory therapies. However, these therapies, such as high-dose inhaled corticosteroids, enhance patient susceptibility to lung infection and exacerbation. Our objective was to assess whether the host-defense peptide, human beta-defensin 2 (hBD-2), can simultaneously reduce pulmonary inflammation in cigarette smoke-exposed mice while maintaining immune competence during bacterial exacerbation. Mice were exposed to cigarette smoke acutely (4 days) or chronically (5 days/week for 7 weeks) and administered hBD-2 intranasally or by gavage. In a separate model of acute exacerbation, chronically exposed mice treated with hBD-2 were infected with non-typeable Haemophilus influenzae prior to sacrifice. In the acute exposure model, cigarette smoke-associated pulmonary neutrophilia was significantly blunted by both local and systemic hBD-2 administration. Similarly, chronically exposed mice administered hBD-2 therapeutically exhibited reduced pulmonary neutrophil infiltration and downregulated pro-inflammatory signaling in the lungs compared to vehicle-treated mice. Finally, in a model of acute bacterial exacerbation, hBD-2 administration effectively limited neutrophil infiltration in the lungs while markedly reducing pulmonary bacterial load. This study shows that hBD-2 treatment can significantly attenuate lung neutrophilia induced by cigarette smoke exposure while preserving immune competence and promoting an appropriate host-defense response to bacterial stimuli.
Collapse
Affiliation(s)
- Nadia Milad
- Quebec Heart and Lung Institute, Université Laval, Quebec City, QC, Canada
- Faculty of Medicine, Université Laval, Quebec City, QC, Canada
| | - Marie Pineault
- Quebec Heart and Lung Institute, Université Laval, Quebec City, QC, Canada
- Faculty of Medicine, Université Laval, Quebec City, QC, Canada
| | - Gabrielle Bouffard
- Quebec Heart and Lung Institute, Université Laval, Quebec City, QC, Canada
- Faculty of Medicine, Université Laval, Quebec City, QC, Canada
| | - Michael Maranda-Robitaille
- Quebec Heart and Lung Institute, Université Laval, Quebec City, QC, Canada
- Faculty of Medicine, Université Laval, Quebec City, QC, Canada
| | - Ariane Lechasseur
- Quebec Heart and Lung Institute, Université Laval, Quebec City, QC, Canada
- Faculty of Medicine, Université Laval, Quebec City, QC, Canada
| | | | - Sophie Aubin
- Quebec Heart and Lung Institute, Université Laval, Quebec City, QC, Canada
| | - Benjamin A. H. Jensen
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Mathieu C. Morissette
- Quebec Heart and Lung Institute, Université Laval, Quebec City, QC, Canada
- Department of Medicine, Université Laval, Quebec City, QC, Canada
| |
Collapse
|
36
|
Peters CM, Peters RC, Lee AD, Lane P, Lam S, Sin DD, McKenzie DC, William Sheel A. software development TO OPTIMIZE THE minimal detectable difference IN huMAN AIRWAY IMAGES CAPTURED USING optical coherence tomography. Clin Physiol Funct Imaging 2022; 42:308-319. [PMID: 35522086 DOI: 10.1111/cpf.12762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Accepted: 05/04/2022] [Indexed: 11/03/2022]
Abstract
Optical coherence tomography (OCT) is an imaging methodology that can be used to assess human airways. OCT avoids the harmful effects of ionizing radiation and has a high spatial resolution making it well suited for imaging the structure of small airways. Analysis of OCT airway images has typically been performed manually by tracing the airway with a relatively high coefficient of variation. The purpose of this study was to develop an analysis tool to reduce the inter- and intra-observer reproducibility of OCT and improve the ability to detect differences in airways. OCT images from healthy, young human volunteers were used to develop and test the OCT software. Measurement software was developed to allow the conversion of the original image into a grayscale image and was followed by an enhancement operation to brighten the image, and contour measurement. A total of 140 OCT images, 70 small (< 2 mm) and 70 medium (2-4 mm) sized airways, were analyzed. The inter- and intra- observer reproducibility of airway measurements ranged for strong to very strong in the small sized airways. For medium sized airways the reproducibility was considered moderate. Bland-Altman bias was low between observers and observations for all measures. The minimal detectable differences in the airway measurements with our semi-automated software were lower relative to manual tracing in medium-sized airways. Our software improves the ability to perform quantitative OCT analysis and may help to quantify the extent of airway remodelling in respiratory disease or elite athletes in future studies. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Carli M Peters
- School of Kinesiology, University of British Columbia, Vancouver, BC, Canada
| | - Robert C Peters
- School of Kinesiology, University of British Columbia, Vancouver, BC, Canada
| | - Anthony D Lee
- Department of Integrative Oncology, BC Cancer Research Institute, Vancouver, BC, Canada
| | - Pierre Lane
- Department of Integrative Oncology, BC Cancer Research Institute, Vancouver, BC, Canada
| | - Stephen Lam
- Department of Integrative Oncology, BC Cancer Research Institute, Vancouver, BC, Canada
| | - Don D Sin
- Centre for Heart Lung Innovation, St. Paul's Hospital and Department of Medicine (Respirology), University of British Columbia, Vancouver, BC, Canada
| | - Donald C McKenzie
- School of Kinesiology, University of British Columbia, Vancouver, BC, Canada
| | - A William Sheel
- School of Kinesiology, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
37
|
Lin H, Wang C, Yu H, Liu Y, Tan L, He S, Li Z, Wang C, Wang F, Li P, Liu J. Protective effect of total Saponins from American ginseng against cigarette smoke-induced COPD in mice based on integrated metabolomics and network pharmacology. Biomed Pharmacother 2022; 149:112823. [PMID: 35334426 DOI: 10.1016/j.biopha.2022.112823] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Revised: 03/11/2022] [Accepted: 03/11/2022] [Indexed: 11/02/2022] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a prevalent respiratory disease. Aiming at assessing the effect of total saponins from American ginseng on COPD, both the chemical composition and anti-COPD activity of total saponins from wild-simulated American ginseng (TSW) and field-grown American ginseng (TSF) were investigated in this study. Firstly, a HPLC-ELSD chromatographic method was established to simultaneously determine the contents of 22 saponins in TSW and TSF. Secondly, CS-induced COPD mouse model was established to evaluate the activity of TSW and TSF. The results indicated that both TSW and TSF had the protective effect against COPD by alleviating oxidative stress and inflammatory response. TSW showed a stronger effect than TSF. Thirdly, an integrated approach involving metabolomics and network pharmacology was used to construct the "biomarker-reaction-enzyme-target" correlation network aiming at further exploring the observed effects. As the results, 15 biomarkers, 9 targets and 5 pathways were identified to play vital roles in the treatment of TSW and TSF on COPD. Fourthly, based on network pharmacology and the CS-stimulated A549 cell model, ginsenoside Rgl, Rc, oleanolic acid, notoginsenoside R1, Fe, silphioside B were certified to be the material basis for the stronger effect of TSW than TSF. Finally, the molecular docking were performed to visualize the binding modes. Our findings suggested that both TSW and TSF could effectively ameliorate the progression of COPD and might be used for the treatment of COPD.
Collapse
Affiliation(s)
- Hongqiang Lin
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China
| | - Caixia Wang
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China
| | - Hui Yu
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China
| | - Yunhe Liu
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China
| | - Luying Tan
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China
| | - Shanmei He
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China
| | - Zhuoqiao Li
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China
| | - Cuizhu Wang
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China; Research Center of Natural Drug, Jilin University, Changchun 130021, China
| | - Fang Wang
- College of Basic Medical Sciences, Jilin University, Changchun 130021, China
| | - Pingya Li
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China; Research Center of Natural Drug, Jilin University, Changchun 130021, China
| | - Jinping Liu
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China; Research Center of Natural Drug, Jilin University, Changchun 130021, China.
| |
Collapse
|
38
|
Association between Smoking Status and Incident Non-Cystic Fibrosis Bronchiectasis in Young Adults: A Nationwide Population-Based Study. J Pers Med 2022; 12:jpm12050691. [PMID: 35629114 PMCID: PMC9144886 DOI: 10.3390/jpm12050691] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 04/22/2022] [Accepted: 04/24/2022] [Indexed: 12/27/2022] Open
Abstract
Smoking traditionally has not been considered as a cause of bronchiectasis. However, few studies have evaluated the association between smoking and bronchiectasis. This study aimed to investigate the association between smoking status and bronchiectasis development in young adults. This study included 6,861,282 adults aged 20−39 years from the Korean National Health Insurance Service database 2009−2012 who were followed-up until the date of development of bronchiectasis, death, or 31 December 2018. We evaluated the incidence of bronchiectasis according to smoking status. During a mean of 7.4 years of follow-up, 23,609 (0.3%) participants developed bronchiectasis. In multivariable Cox regression analysis, ex-smokers (adjusted hazard ratio (aHR) = 1.07, 95% confidence interval (CI) = 1.03−1.13) and current-smokers (aHR = 1.06, 95% CI = 1.02−1.10) were associated with incident bronchiectasis, with the highest HR in ≥ 10 pack-years current smokers (aHR = 1.12, 95% CI = 1.06−1.16). The association of smoking with bronchiectasis was more profound in females than in males (p for interaction < 0.001), in younger than in older participants (p for interaction = 0.036), and in the overweight and obese than in the normal weight or underweight (p for interaction = 0.023). In conclusion, our study shows that smoking is associated with incident bronchiectasis in young adults. The association of smoking with bronchiectasis development was stronger in females, 20−29 year-olds, and the overweight and obese than in males, 30−40-year-olds, and the normal weight or underweight, respectively.
Collapse
|
39
|
Somayaji R, Chalmers JD. Just breathe: a review of sex and gender in chronic lung disease. Eur Respir Rev 2022; 31:31/163/210111. [PMID: 35022256 DOI: 10.1183/16000617.0111-2021] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Accepted: 08/20/2021] [Indexed: 01/08/2023] Open
Abstract
Chronic lung diseases are the third leading cause of death worldwide and are increasing in prevalence over time. Although much of our traditional understanding of health and disease is derived from study of the male of the species - be it animal or human - there is increasing evidence that sex and gender contribute to differences in disease risk, prevalence, presentation, severity, treatment approach, response and outcomes. Chronic obstructive pulmonary disease, asthma and bronchiectasis represent the most prevalent and studied chronic lung diseases and have key sex- and gender-based differences which are critical to consider and incorporate into clinical and research approaches. Mechanistic differences present opportunities for therapeutic development whereas behavioural and clinical differences on the part of patients and providers present opportunities for greater education and understanding at multiple levels. In this review, we seek to summarise the sex- and gender-based differences in key chronic lung diseases and outline the clinical and research implications for stakeholders.
Collapse
Affiliation(s)
- Ranjani Somayaji
- Dept of Medicine, Cumming School of Medicine, University of Calgary, Calgary, Canada .,Dept of Microbiology, Immunology and Infectious Disease, University of Calgary, Calgary, Canada.,Dept of Community Health Sciences, University of Calgary, Calgary, Canada
| | - James D Chalmers
- Division of Molecular and Clinical Medicine, University of Dundee, Ninewells Hospital and Medical School, Dundee, UK
| |
Collapse
|
40
|
Ghosh AJ, Hobbs BD. Recent advancements in understanding the genetic involvement of alpha-1 antitrypsin deficiency associated lung disease: a look at future precision medicine approaches. Expert Rev Respir Med 2022; 16:173-182. [PMID: 35025710 PMCID: PMC8983484 DOI: 10.1080/17476348.2022.2027755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
INTRODUCTION Alpha-1 antitrypsin deficiency occurs in individuals with deleterious genetic mutations on both chromosomes (maternal and paternal) in SERPINA1, the gene encoding the alpha-1 antitrypsin protein. There has been substantial progress in understanding the genetic variation that underlies the heterogeneous penetrance of lung disease in alpha-1 antitrypsin deficiency. AREAS COVERED This review will cover SERPINA1 gene structure and genetic variation, population genetics, genome-wide genetic modifiers of lung disease, alternative mechanisms of disease, and emerging therapeutics - including gene and cell therapy - related to alpha-1 antitrypsin deficiency-associated lung disease. EXPERT OPINION There remains ample opportunity to employ precision medicine in the diagnosis, prognosis, and therapy of alpha-1 antitrypsin deficiency-associated lung disease. In particular, a genome-wide association study and subsequent polygenic risk score is an important first step in identifying genome-wide genetic modifiers contributing to the variability of lung disease in severe alpha-1 antitrypsin deficiency.
Collapse
Affiliation(s)
- Auyon J. Ghosh
- Assistant Professor of Medicine, Division of Pulmonary, Critical Care, and Sleep Medicine, SUNY Upstate Medical University, 750 E. Adams St, Syracuse, NY, 13210
| | - Brian D. Hobbs
- Assistant Professor of Medicine, Channing Division of Network Medicine, Brigham and Women’s Hospital, 181 Longwood Ave, Boston, MA, 02115,Division of Pulmonary and Critical Care Medicine, Brigham and Women’s Hospital,Harvard Medical School
| |
Collapse
|
41
|
Zhang M, Lu Y, Liu L, Zhang X, Ning J. Role and mechanism of miR-181a-5p in mice with chronic obstructive pulmonary disease by regulating HMGB1 and the NF-κB pathway. Cells Tissues Organs 2022:000522155. [DOI: 10.1159/000522155] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Accepted: 01/16/2022] [Indexed: 11/19/2022] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a common respiratory disease. This study explored the mechanism of miR-181a-5p in the inflammatory response in COPD mice. COPD mouse models were established by cigarette smoke (CS) exposure following pretreatment with recombinant adeno-associated virus (rAAv)-miR-181a-5p, si-HMGB1 (high mobility group box 1), and NF-κB pathway inhibitor PDTC, respectively. Pathological changes of lung tissues were determined by HE staining. BALF was collected to count total cells, neutrophils and lymphocytes using a Countess II automatic cell counter. Expressions of NE and inflammatory factors (TNF-α, IL-6, IL-8 and IFN-γ) were detected by ELISA. Binding relationship between miR-181a-5p and HMGB1 was predicted on Starbase (http://starbase.sysu.edu.cn/index.php) and validated by dual-luciferase assay. miR-181a-5p expression was detected by RT-qPCR, and expressions of HMGB1, IκBα, p-IκBα were detected by Western blot. The expression level of miR-181a-5p was lower in lung tissues. miR-181a-5p overexpression alleviated inflammatory response and pathological changes of lung tissues in COPD mice, with decreased pulmonary inflammation scores, total cells, neutrophils, and lymphocytes and expressions of NE and inflammatory factors. HMGB1 expression level was increased in COPD mice. miR-181a-5p targeted HMGB1. si-HMGB1 relieved inflammatory responses in COPD mice. NF-κB was activated in COPD mice, evidenced by degraded IκBα and increased p-IκBα level. si-HMGB1 significantly restrained the activation of NF-κB pathway. Briefly, miR-181a-5p targets HMGB1 to inhibit the NF-κB pathway, thus alleviating the inflammatory response in COPD mice.
Collapse
|
42
|
Choudhary I, Vo T, Paudel K, Wen X, Gupta R, Kesimer M, Patial S, Saini Y. Vesicular and extravesicular protein analyses from the airspaces of ozone-exposed mice revealed signatures associated with mucoinflammatory lung disease. Sci Rep 2021; 11:23203. [PMID: 34853335 PMCID: PMC8636509 DOI: 10.1038/s41598-021-02256-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Accepted: 11/08/2021] [Indexed: 12/16/2022] Open
Abstract
Lung epithelial lining fluid (ELF) harbors a variety of proteins that influence homeostatic and stress responses in the airspaces. Exosomes, nano-sized extracellular vesicles, contain many proteins that vary in abundance and composition based on the prevailing conditions. Ozone causes inflammatory responses in the airspaces of experimental animals and humans. However, the exosomal protein signatures contained within the ELF from ozone-exposed lung airspaces remain poorly characterized. To explore this, we hypothesized that ozone triggers the release of exosome-bound inflammatory proteins from various cells that reflect mucoobstructive lung disease. Accordingly, we repetitively exposed adult male and female C57BL/6 mice to HEPA-filtered air (air) or 0.8 ppm ozone (4 h per day) for 14 days (five consecutive days of exposure, 2 days of rest, five consecutive days of exposure, 2 days of rest, four consecutive days of exposure). Exosome-bound proteomic signatures, as well as the levels of soluble inflammatory mediators in the bronchoalveolar lavage fluid (BALF), were determined 12-16 h after the last exposure. Principal component analyses of the exosome-bound proteome revealed a clear distinction between air-exposed and ozone-exposed mice, as well as between ozone-exposed males and ozone-exposed females. In addition to 575 proteins that were enriched in both sexes upon ozone exposure, 243 and 326 proteins were enriched uniquely in ozone-exposed males and females, respectively. Ingenuity pathway analyses on enriched proteins between ozone- and air-exposed mice revealed enrichment of pro-inflammatory pathways. More specifically, macrophage activation-related proteins were enriched in exosomes from ozone-exposed mice. Cytokine analyses on the BALF revealed elevated levels of G-CSF, KC, IP-10, IL-6, and IL-5 in ozone-exposed mice. Finally, the histopathological assessment revealed significantly enhanced intracellular localization of mucoinflammatory proteins including MUC5B and FIZZ1 in ozone-exposed mice in a cell-specific manner indicating the cellular sources of the proteins that are ferried in the exosomes upon ozone-induced lung injury. Collectively, this study identified exosomal, secretory, and cell-specific proteins and biological pathways following repetitive exposure of mice to ozone.
Collapse
Affiliation(s)
- Ishita Choudhary
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, 1909 Skip Bertman Drive, Baton Rouge, LA, 70803, USA
| | - Thao Vo
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, 1909 Skip Bertman Drive, Baton Rouge, LA, 70803, USA
| | - Kshitiz Paudel
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, 1909 Skip Bertman Drive, Baton Rouge, LA, 70803, USA
| | - Xue Wen
- Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, 70803, USA
| | - Richa Gupta
- Department of Pathology and Laboratory Medicine, UNC School of Medicine, Chapel Hill, NC, 27510, USA
| | - Mehmet Kesimer
- Department of Pathology and Laboratory Medicine, UNC School of Medicine, Chapel Hill, NC, 27510, USA
| | - Sonika Patial
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, 1909 Skip Bertman Drive, Baton Rouge, LA, 70803, USA
| | - Yogesh Saini
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, 1909 Skip Bertman Drive, Baton Rouge, LA, 70803, USA.
| |
Collapse
|
43
|
Peng J, Wu F, Tian H, Yang H, Zheng Y, Deng Z, Wang Z, Xiao S, Wen X, Huang P, Lu L, Dai C, Zhao N, Huang S, Ran P, Zhou Y. Clinical characteristics of and risk factors for small airway dysfunction detected by impulse oscillometry. Respir Med 2021; 190:106681. [PMID: 34784563 DOI: 10.1016/j.rmed.2021.106681] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2021] [Revised: 10/14/2021] [Accepted: 11/07/2021] [Indexed: 02/08/2023]
Abstract
BACKGROUND Small airway dysfunction (SAD) is an early lesion of chronic respiratory disease that is best detected using impulse oscillometry (IOS). Few studies have investigated risk factors for IOS-defined SAD (IOS-SAD) in a large population. We aimed to explore the clinical features of and risk factors for IOS-SAD in a community-based population. METHODS We divided subjects into IOS-SAD and non-SAD groups based on a cutoff of >0.07 kPa/L/s in the difference between the resistance at 5 Hz versus the resistance at 20 Hz (R5-R20). All participants underwent spirometry, IOS, and completed a questionnaire; some participants underwent computed tomography (CT). We analyzed the risk factors for SAD based on binary logistic regression. RESULTS The total cohort comprised 1327 subjects. The prevalence of IOS-SAD was 32.9% (437/1327). Compared with the non-SAD group, the IOS-SAD group was older (64.0 ± 7.8 vs. 59.6 ± 7.8 years, p < 0.001), included less never-smokers (30.2% vs. 35.8%, p < 0.001), had greater airway resistance and worse lung function, indicated by a larger R5-R20 (0.15 ± 0.08 vs. 0.03 ± 0.02 kPa/L/s, p < 0.001) and smaller forced expiratory volume in 1 s to forced vital capacity after bronchodilation (60.2 ± 14.4% vs. 72.6 ± 10.0%, p < 0.001); on CT, the IOS-SAD group had higher prevalences of emphysema and gas trapping. Risk factors for SAD were older age, high BMI, smoking, childhood cough, and asthma. CONCLUSION Subjects with IOS-SAD had increased airway resistance and visible CT changes. Individuals with smoking exposure, advanced age, high BMI, childhood cough, and asthma were more prone to SAD. CLINICAL TRIAL REGISTRATION ChiCTR1900024643.
Collapse
Affiliation(s)
- Jieqi Peng
- National Center for Respiratory Medicine, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Fan Wu
- National Center for Respiratory Medicine, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Heshen Tian
- National Center for Respiratory Medicine, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Huajing Yang
- National Center for Respiratory Medicine, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Youlan Zheng
- National Center for Respiratory Medicine, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Zhishan Deng
- National Center for Respiratory Medicine, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Zihui Wang
- National Center for Respiratory Medicine, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Shan Xiao
- National Center for Respiratory Medicine, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xiang Wen
- National Center for Respiratory Medicine, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Peiyu Huang
- National Center for Respiratory Medicine, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Lifei Lu
- National Center for Respiratory Medicine, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Cuiqiong Dai
- National Center for Respiratory Medicine, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Ningning Zhao
- National Center for Respiratory Medicine, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Suyin Huang
- National Center for Respiratory Medicine, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Pixin Ran
- National Center for Respiratory Medicine, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.
| | - Yumin Zhou
- National Center for Respiratory Medicine, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.
| |
Collapse
|
44
|
Reinke SN, Naz S, Chaleckis R, Gallart-Ayala H, Kolmert J, Kermani NZ, Tiotiu A, Broadhurst DI, Lundqvist A, Olsson H, Ström M, Wheelock ÅM, Gómez C, Ericsson M, Sousa AR, Riley JH, Bates S, Scholfield J, Loza M, Baribaud F, Bakke PS, Caruso M, Chanez P, Fowler SJ, Geiser T, Howarth P, Horváth I, Krug N, Montuschi P, Behndig A, Singer F, Musial J, Shaw DE, Dahlén B, Hu S, Lasky-Su J, Sterk PJ, Chung KF, Djukanovic R, Dahlén SE, Adcock IM, Wheelock CE. Urinary metabotype of severe asthma evidences decreased carnitine metabolism independent of oral corticosteroid treatment in the U-BIOPRED study. Eur Respir J 2021; 59:13993003.01733-2021. [PMID: 34824054 PMCID: PMC9245194 DOI: 10.1183/13993003.01733-2021] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Accepted: 10/28/2021] [Indexed: 11/05/2022]
Abstract
INTRODUCTION Asthma is a heterogeneous disease with poorly defined phenotypes. Severe asthmatics often receive multiple treatments including oral corticosteroids (OCS). Treatment may modify the observed metabotype, rendering it challenging to investigate underlying disease mechanisms. Here, we aimed to identify dysregulated metabolic processes in relation to asthma severity and medication. METHODS Baseline urine was collected prospectively from healthy participants (n=100), mild-to-moderate asthmatics (n=87) and severe asthmatics (n=418) in the cross-sectional U-BIOPRED cohort; 12-18-month longitudinal samples were collected from severe asthmatics (n=305). Metabolomics data were acquired using high-resolution mass spectrometry and analysed using univariate and multivariate methods. RESULTS Ninety metabolites were identified, with 40 significantly altered (p<0.05, FDR<0.05) in severe asthma and 23 by OCS use. Multivariate modelling showed that observed metabotypes in healthy participants and mild-to-moderate asthmatics differed significantly from severe asthmatics (p=2.6×10-20), OCS-treated asthmatics differed significantly from non-treated (p=9.5×10-4), and longitudinal metabotypes demonstrated temporal stability. Carnitine levels evidenced the strongest OCS-independent decrease in severe asthma. Reduced carnitine levels were associated with mitochondrial dysfunction via decreases in pathway enrichment scores of fatty acid metabolism and reduced expression of the carnitine transporter SLC22A5 in sputum and bronchial brushings. CONCLUSIONS This is the first large-scale study to delineate disease- and OCS-associated metabolic differences in asthma. The widespread associations with different therapies upon the observed metabotypes demonstrate the necessity to evaluate potential modulating effects on a treatment- and metabolite-specific basis. Altered carnitine metabolism is a potentially actionable therapeutic target that is independent of OCS treatment, highlighting the role of mitochondrial dysfunction in severe asthma.
Collapse
Affiliation(s)
- Stacey N Reinke
- Division of Physiological Chemistry 2, Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, Sweden.,Centre for Integrative Metabolomics & Computational Biology, School of Science, Edith Cowan University, Perth, Australia.,equal contribution
| | - Shama Naz
- Division of Physiological Chemistry 2, Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, Sweden.,equal contribution
| | - Romanas Chaleckis
- Division of Physiological Chemistry 2, Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, Sweden.,Gunma Initiative for Advanced Research (GIAR), Gunma University, Maebashi, Japan
| | - Hector Gallart-Ayala
- Division of Physiological Chemistry 2, Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, Sweden
| | - Johan Kolmert
- Division of Physiological Chemistry 2, Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, Sweden.,The Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | | | - Angelica Tiotiu
- National Heart and Lung Institute, Imperial College, London, U.K.,Department of Pulmonology, University Hospital of Nancy, Nancy, France
| | - David I Broadhurst
- Centre for Integrative Metabolomics & Computational Biology, School of Science, Edith Cowan University, Perth, Australia
| | - Anders Lundqvist
- Respiratory & Immunology, BioPharmaceuticals R&D, DMPK, Research and Early Development, AstraZeneca, Gothenburg, Sweden
| | - Henric Olsson
- Translational Science and Experimental Medicine, Research and Early Development, AstraZeneca, Gothenburg, Sweden
| | - Marika Ström
- Respiratory Medicine Unit, K2 Department of Medicine Solna and Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden.,Department of Respiratory Medicine and Allergy, Karolinska University Hospital, Stockholm, Sweden
| | - Åsa M Wheelock
- Respiratory Medicine Unit, K2 Department of Medicine Solna and Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden.,Department of Respiratory Medicine and Allergy, Karolinska University Hospital, Stockholm, Sweden
| | - Cristina Gómez
- Division of Physiological Chemistry 2, Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, Sweden.,The Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Magnus Ericsson
- Department of Clinical Pharmacology, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
| | | | | | | | - James Scholfield
- Faculty of Medicine, Southampton University and NIHR Southampton Respiratory Biomedical Research Center, University Hospital Southampton, Southampton, U.K
| | - Matthew Loza
- Janssen Research and Development, High Wycombe, U.K
| | | | - Per S Bakke
- Institute of Medicine, University of Bergen, Bergen, Norway
| | - Massimo Caruso
- Department of Biomedical and Biotechnological Sciences and Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Pascal Chanez
- Assistance Publique des Hôpitaux de Marseille, Clinique des Bronches, Allergies et Sommeil, Aix Marseille Université, Marseille, France
| | - Stephen J Fowler
- Division of Infection, Immunity and Respiratory Medicine, Faculty of Biology, School of Biological Sciences, Medicine and Health, University of Manchester, and Manchester Academic Health Science Centre and NIHR Biomedical Research Centre, Manchester University Hospitals NHS Foundation Trust, Manchester, U.K
| | - Thomas Geiser
- Department of Pulmonary Medicine, University Hospital, University of Bern, Switzerland
| | - Peter Howarth
- Faculty of Medicine, Southampton University and NIHR Southampton Respiratory Biomedical Research Center, University Hospital Southampton, Southampton, U.K
| | - Ildikó Horváth
- Department of Pulmonology, Semmelweis University, Budapest, Hungary
| | - Norbert Krug
- Fraunhofer Institute for Toxicology and Experimental Medicine, Hannover, Germany
| | - Paolo Montuschi
- Pharmacology, Catholic University of the Sacred Heart, Rome, Italy
| | - Annelie Behndig
- Department of Public Health and Clinical Medicine, Section of Medicine, Umeå University, Umeå, Sweden
| | - Florian Singer
- Division of Paediatric Respiratory Medicine and Allergology, Department of Paediatrics, Inselspital, Bern University Hospital, University of Bern, Switzerland
| | - Jacek Musial
- Dept of Medicine, Jagiellonian University Medical College, Krakow, Poland
| | - Dominick E Shaw
- Nottingham NIHR Biomedical Research Centre, University of Nottingham, U.K
| | - Barbro Dahlén
- Department of Respiratory Medicine and Allergy, Karolinska University Hospital, Stockholm, Sweden
| | - Sile Hu
- Data Science Institute, Imperial College, London, U.K
| | - Jessica Lasky-Su
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Peter J Sterk
- Department of Respiratory Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Kian Fan Chung
- National Heart and Lung Institute, Imperial College, London, U.K
| | - Ratko Djukanovic
- Faculty of Medicine, Southampton University and NIHR Southampton Respiratory Biomedical Research Center, University Hospital Southampton, Southampton, U.K
| | - Sven-Erik Dahlén
- The Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden.,Department of Respiratory Medicine and Allergy, Karolinska University Hospital, Stockholm, Sweden
| | - Ian M Adcock
- National Heart and Lung Institute, Imperial College, London, U.K
| | - Craig E Wheelock
- Division of Physiological Chemistry 2, Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, Sweden .,Gunma Initiative for Advanced Research (GIAR), Gunma University, Maebashi, Japan.,Department of Respiratory Medicine and Allergy, Karolinska University Hospital, Stockholm, Sweden
| | | |
Collapse
|
45
|
Gregory AD, Tran KC, Tamaskar AS, Wei J, Zhao J, Zhao Y. USP13 Deficiency Aggravates Cigarette-smoke-induced Alveolar Space Enlargement. Cell Biochem Biophys 2021; 79:485-491. [PMID: 34032995 PMCID: PMC8887808 DOI: 10.1007/s12013-021-01000-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/16/2021] [Indexed: 11/26/2022]
Abstract
Alveolar enlargement is a pathological feature of emphysema. Long-term exposure to cigarette smoke (CS) is a high-risk factor for the development of emphysema. Abnormal protein ubiquitination has been implicated to regulate the development of human disorders, however, the role of protein ubiquitination in emphysema has not been well-studied. In this study, we attempted to investigate if a deubiquitinase, USP13, regulates the development of emphysema. Under a mild CS exposure condition, USP13-deficient mice show significant increases in alveolar chord length, indicating that USP13-deficient mice are susceptible to CS-induced alveolar enlargement. It has been shown that USP13 knockout reduced fibronectin expression in lungs. Here, we found that collagen levels were reduced in USP13 siRNA-transfected lung fibroblast cells. This suggests that a loss of extracellular matrix in connective tissues contributes to alveolar enlargement in USP13-deficient mice in response to CS exposure. Further, we investigated the role of USP13 in the expression of oxidative stress markers TXNIP and HMOX1. An increase in HMOX1 abundance was observed in USP13 knockdown lung fibroblast and epithelial cells. Overexpression of USP13 reduced HMOX1 protein levels in lung fibroblast cells, suggesting that modulation of USP13 levels may affect oxidative stress. Knockdown of USP13 significantly reduced TXNIP levels in lungs or lung fibroblast cells. A protein stability pulse-chase assay showed that TXNIP is instable within USP13 knockdown lung fibroblast cells. Further, the reduction of TXNIP was observed in USP13 inhibitor-treated lung epithelial cells. USP13-deficient mice also show higher levels of IgG in bronchoalveolar lavage fluid. This study provides evidence showing that USP13 deficiency plays a role in alveolar space enlargement.
Collapse
Affiliation(s)
- Alyssa D Gregory
- Department of Medicine, The University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Kevin C Tran
- Department of Physiology and Cell Biology, The Ohio State University Wexner Medical Center, Columbus, OH, USA
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Arya S Tamaskar
- Department of Physiology and Cell Biology, The Ohio State University Wexner Medical Center, Columbus, OH, USA
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Jianxin Wei
- Department of Medicine, The University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Jing Zhao
- Department of Physiology and Cell Biology, The Ohio State University Wexner Medical Center, Columbus, OH, USA
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, USA
- Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Yutong Zhao
- Department of Physiology and Cell Biology, The Ohio State University Wexner Medical Center, Columbus, OH, USA.
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, USA.
- Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, USA.
| |
Collapse
|
46
|
Reddy KD, Rutting S, Tonga K, Xenaki D, Simpson JL, McDonald VM, Plit M, Malouf M, Zakarya R, Oliver BG. Sexually dimorphic production of interleukin-6 in respiratory disease. Physiol Rep 2021; 8:e14459. [PMID: 32472750 PMCID: PMC7260763 DOI: 10.14814/phy2.14459] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 04/30/2020] [Accepted: 05/03/2020] [Indexed: 12/27/2022] Open
Abstract
Diverging susceptibility and severity in respiratory diseases is prevalent between males and females. Sex hormones have inconclusively been attributed as the cause of these differences, however, strong evidence exists promoting genetic factors leading to sexual dimorphism. As such, we investigate differential proinflammatory cytokine (interleukin (IL)‐6 and CXCL8) release from TNF‐α stimulated primary human lung fibroblasts in vitro. We present, for the first time, in vitro evidence supporting clinical findings of differential production of IL‐6 between males and females across various respiratory diseases. IL‐6 was found to be produced approximately two times more from fibroblasts derived from females compared to males. As such we demonstrate sexual dimorphism in cytokine production of IL‐6 outside the context of biological factors in the human body. As such, our data highlight that differences exist between males and females in the absence of sex hormones. We, for the first time, demonstrate inherent in vitro differences exist between males and females in pulmonary fibroblasts.
Collapse
Affiliation(s)
- Karosham D Reddy
- School of Life Sciences, University of Technology Sydney, Sydney, NSW, Australia.,Respiratory Cellular and Molecular Biology, Woolcock Institute of Medical Research, University of Sydney, Sydney, NSW, Australia
| | - Sandra Rutting
- Respiratory Cellular and Molecular Biology, Woolcock Institute of Medical Research, University of Sydney, Sydney, NSW, Australia
| | - Katrina Tonga
- Respiratory Cellular and Molecular Biology, Woolcock Institute of Medical Research, University of Sydney, Sydney, NSW, Australia.,St Vincent's Hospital Sydney and St Vincent's Clinical School, University of New South Wales, Darlinghurst, NSW, Australia
| | - Dikaia Xenaki
- Respiratory Cellular and Molecular Biology, Woolcock Institute of Medical Research, University of Sydney, Sydney, NSW, Australia
| | - Jodie L Simpson
- Priority Research Centre for Healthy Lungs, Faculty of Health and Medicine, University of Newcastle, Callaghan, NSW, Australia
| | - Vanessa M McDonald
- Priority Research Centre for Healthy Lungs, Faculty of Health and Medicine, University of Newcastle, Callaghan, NSW, Australia
| | - Marshall Plit
- St Vincent's Hospital Sydney and St Vincent's Clinical School, University of New South Wales, Darlinghurst, NSW, Australia
| | - Monique Malouf
- St Vincent's Hospital Sydney and St Vincent's Clinical School, University of New South Wales, Darlinghurst, NSW, Australia
| | - Razia Zakarya
- School of Life Sciences, University of Technology Sydney, Sydney, NSW, Australia.,Respiratory Cellular and Molecular Biology, Woolcock Institute of Medical Research, University of Sydney, Sydney, NSW, Australia
| | - Brian G Oliver
- School of Life Sciences, University of Technology Sydney, Sydney, NSW, Australia.,Respiratory Cellular and Molecular Biology, Woolcock Institute of Medical Research, University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
47
|
Bai S, Zhao L. Imbalance Between Injury and Defense in the COPD Emphysematous Phenotype. Front Med (Lausanne) 2021; 8:653332. [PMID: 34026786 PMCID: PMC8131650 DOI: 10.3389/fmed.2021.653332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Accepted: 03/10/2021] [Indexed: 11/15/2022] Open
Abstract
The chronic obstructive pulmonary disease (COPD) emphysematous phenotype is characterized by destruction of lung tissue structure. Patients with this phenotype usually present with typical emphysema-like changes on chest computed Tomography CT, experience higher mortality and poorer prognosis, and are insensitive to routine pharmacological COPD therapy. However, the pathogenesis for the COPD emphysematous phenotype remains unclear, resulting in diagnostic and therapeutic challenges. The imbalance between injury and defense mechanisms is essential in the progression of many pulmonary diseases. Thus, in this review, we focus on the pathogenesis of the COPD emphysematous phenotype and discuss the pathophysiological processes involved in disease progression, from the perspective of injury and defense imbalance.
Collapse
Affiliation(s)
- Shuang Bai
- Department of Pulmonary and Critical Care Medicine, Shengjing Hospital of China Medical University, Shenyang, China
| | - Li Zhao
- Department of Pulmonary and Critical Care Medicine, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
48
|
Yang Y, Di T, Zhang Z, Liu J, Fu C, Wu Y, Bian T. Dynamic evolution of emphysema and airway remodeling in two mouse models of COPD. BMC Pulm Med 2021; 21:134. [PMID: 33902528 PMCID: PMC8073949 DOI: 10.1186/s12890-021-01456-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Accepted: 03/03/2021] [Indexed: 12/22/2022] Open
Abstract
Background Establishment of a mouse model is important for investigating the mechanism of chronic obstructive pulmonary disease (COPD). In this study, we observed and compared the evolution of the pathology in two mouse models of COPD induced by cigarette smoke (CS) exposure alone or in combination with lipopolysaccharide (LPS). Methods One hundred eight wild-type C57BL/6 mice were equally divided into three groups: the (1) control group, (2) CS-exposed group (CS group), and (3) CS + LPS-exposed group (CS + LPS group). The body weight of the mice was recorded, and noninvasive lung function tests were performed monthly. Inflammation was evaluated by counting the number of inflammatory cells in bronchoalveolar lavage fluid and measuring the expression of the IL-6 mRNA in mouse lung tissue. Changes in pathology were assessed by performing hematoxylin and eosin and Masson staining of lung tissue sections. Results The two treatments induced emphysema and airway remodeling and decreased lung function. Emphysema was induced after 1 month of exposure to CS or CS + LPS, while airway remodeling was induced after 2 months of exposure to CS + LPS and 3 months of exposure to CS. Moreover, the mice in the CS + LPS group exhibited more severe inflammation and airway remodeling than the mice in the CS group, but the two treatments induced similar levels of emphysema. Conclusion Compared with the single CS exposure method, the CS + LPS exposure method is a more suitable model of COPD in airway remodeling research. Conversely, the CS exposure method is a more suitable model of COPD for emphysema research due to its simple operation. Supplementary Information The online version contains supplementary material available at 10.1186/s12890-021-01456-z.
Collapse
Affiliation(s)
- Yue Yang
- Department of Respiratory Medicine, Wuxi People's Hospital Affiliated With Nanjing Medical University, Wuxi, 214023, Jiangsu, People's Republic of China
| | - Tingting Di
- Department of Respiratory Medicine, Wuxi People's Hospital Affiliated With Nanjing Medical University, Wuxi, 214023, Jiangsu, People's Republic of China
| | - Zixiao Zhang
- Department of Respiratory Medicine, Wuxi People's Hospital Affiliated With Nanjing Medical University, Wuxi, 214023, Jiangsu, People's Republic of China
| | - Jiaxin Liu
- Department of Respiratory Medicine, Wuxi People's Hospital Affiliated With Nanjing Medical University, Wuxi, 214023, Jiangsu, People's Republic of China
| | - Congli Fu
- Respiratory and Critical Care Medicine, Zhejiang Province People's Hospital, Hangzhou, 310000, Zhejiang, People's Republic of China
| | - Yan Wu
- Department of Respiratory Medicine, Wuxi People's Hospital Affiliated With Nanjing Medical University, Wuxi, 214023, Jiangsu, People's Republic of China.
| | - Tao Bian
- Department of Respiratory Medicine, Wuxi People's Hospital Affiliated With Nanjing Medical University, Wuxi, 214023, Jiangsu, People's Republic of China.
| |
Collapse
|
49
|
Szafran BN, Borazjani A, Seay CN, Carr RL, Lehner R, Kaplan BLF, Ross MK. Effects of Chlorpyrifos on Serine Hydrolase Activities, Lipid Mediators, and Immune Responses in Lungs of Neonatal and Adult Mice. Chem Res Toxicol 2021; 34:1556-1571. [PMID: 33900070 DOI: 10.1021/acs.chemrestox.0c00488] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Chlorpyrifos (CPF) is an organophosphate (OP) pesticide that causes acute toxicity by inhibiting acetylcholinesterase (AChE) in the nervous system. However, endocannabinoid (eCB) metabolizing enzymes in brain of neonatal rats are more sensitive than AChE to inhibition by CPF, leading to increased levels of eCBs. Because eCBs are immunomodulatory molecules, we investigated the association between eCB metabolism, lipid mediators, and immune function in adult and neonatal mice exposed to CPF. We focused on lung effects because epidemiologic studies have linked pesticide exposures to respiratory diseases. CPF was hypothesized to disrupt lung eCB metabolism and alter lung immune responses to lipopolysaccharide (LPS), and these effects would be more pronounced in neonatal mice due to an immature immune system. We first assessed the biochemical effects of CPF in adult mice (≥8 weeks old) and neonatal mice after administering CPF (2.5 mg/kg, oral) or vehicle for 7 days. Tissues were harvested 4 h after the last CPF treatment and lung microsomes from both age groups demonstrated CPF-dependent inhibition of carboxylesterases (Ces), a family of xenobiotic and lipid metabolizing enzymes, whereas AChE activity was inhibited in adult lungs only. Activity-based protein profiling (ABPP)-mass spectrometry of lung microsomes identified 31 and 32 individual serine hydrolases in neonatal lung and adult lung, respectively. Of these, Ces1c/Ces1d/Ces1b isoforms were partially inactivated by CPF in neonatal lung, whereas Ces1c/Ces1b and Ces1c/BChE were partially inactivated in adult female and male lungs, respectively, suggesting age- and sex-related differences in their sensitivity to CPF. Monoacylglycerol lipase (MAGL) and fatty acid amide hydrolase (FAAH) activities in lung were unaffected by CPF. When LPS (1.25 mg/kg, i.p.) was administered following the 7-day CPF dosing period, little to no differences in lung immune responses (cytokines and immunophenotyping) were noted between the CPF and vehicle groups. However, a CPF-dependent increase in the amounts of dendritic cells and certain lipid mediators in female lung following LPS challenge was observed. Experiments in neonatal and adult Ces1d-/- mice yielded similar results as wild type mice (WT) following CPF treatment, except that CPF augmented LPS-induced Tnfa mRNA in adult Ces1d-/- mouse lungs. This effect was associated with decreased expression of Ces1c mRNA in Ces1d-/- mice versus WT mice in the setting of LPS exposure. We conclude that CPF exposure inactivates several Ces isoforms in mouse lung and, during an inflammatory response, increases certain lipid mediators in a female-dependent manner. However, it did not cause widespread altered lung immune effects in response to an LPS challenge.
Collapse
Affiliation(s)
- Brittany N Szafran
- Department of Comparative Biomedical Sciences, Center for Environmental Health Sciences, College of Veterinary Medicine, Mississippi State University, Mississippi State, Mississippi 39762, United States
| | - Abdolsamad Borazjani
- Department of Comparative Biomedical Sciences, Center for Environmental Health Sciences, College of Veterinary Medicine, Mississippi State University, Mississippi State, Mississippi 39762, United States
| | - Caitlin N Seay
- Department of Comparative Biomedical Sciences, Center for Environmental Health Sciences, College of Veterinary Medicine, Mississippi State University, Mississippi State, Mississippi 39762, United States
| | - Russell L Carr
- Department of Comparative Biomedical Sciences, Center for Environmental Health Sciences, College of Veterinary Medicine, Mississippi State University, Mississippi State, Mississippi 39762, United States
| | - Richard Lehner
- Departments of Cell Biology and Pediatrics, Group on Molecular & Cell Biology of Lipids, University of Alberta, Edmonton, Alberta T6G 2H7, Canada
| | - Barbara L F Kaplan
- Department of Comparative Biomedical Sciences, Center for Environmental Health Sciences, College of Veterinary Medicine, Mississippi State University, Mississippi State, Mississippi 39762, United States
| | - Matthew K Ross
- Department of Comparative Biomedical Sciences, Center for Environmental Health Sciences, College of Veterinary Medicine, Mississippi State University, Mississippi State, Mississippi 39762, United States
| |
Collapse
|
50
|
Zhu S, Huang S, Xia G, Wu J, Shen Y, Wang Y, Ostrom RS, Du A, Shen C, Xu C. Anti-inflammatory effects of α7-nicotinic ACh receptors are exerted through interactions with adenylyl cyclase-6. Br J Pharmacol 2021; 178:2324-2338. [PMID: 33598912 DOI: 10.1111/bph.15412] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2020] [Revised: 12/20/2020] [Accepted: 02/11/2021] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND AND PURPOSE Nicotinic ACh receptors containing the α7 sub-unit (α7-nAChRs) suppress inflammation through a wide range of pathways in immune cells. These receptors are thus potentially involved in a number of inflammatory diseases. However, the detailed mechanisms underlying the anti-inflammatory effects of α7-nAChRs remain to be described. EXPERIMENTAL APPROACH Anti-inflammatory effects of α7-nAChR agonists were assessed in both murine macrophages (RAW 264.7) and bone marrow-derived macrophages (BMDM), stimulated with LPS, using immunoblotting, RT-PCR and luciferase reporter assays. The role of adenylyl cyclase-6 in the degradation of Toll-like receptor 4 (TLR4) following endocytosis, was explored via overexpression and knockdown. A mouse model of chronic obstructive pulmonary disease (COPD) induced by porcine pancreatic elastase was used to confirm key findings. RESULTS Anti-inflammatory effects of α7-nAChRs were largely dependent on adenylyl cyclase-6 activation, as knockdown of adenylyl cyclase-6 considerably reduced the effects of α7-nAChR agonists while adenylyl cyclase-6 overexpression promoted them. We found that α7-nAChRs and adenylyl cyclase-6 are co-localized in lipid rafts of macrophages and directly interact. Activation of adenylyl cyclase-6 led to increased degradation of TLR4. Administration of the α7-nAChR agonist PNU-282987 attenuated pathological and inflammatory end points in a mouse model of COPD. CONCLUSION AND IMPLICATIONS The α7-nAChRs inhibit inflammation through activating adenylyl cyclase-6 and promoting degradation of TLR4. The use of α7-nAChR agonists may represent a novel therapeutic approach for treating COPD and possibly other inflammatory diseases.
Collapse
Affiliation(s)
- Simeng Zhu
- Department of Cardiology, Shanghai Jiaotong University Affiliated Sixth People's Hospital, Shanghai Jiaotong University School of Medicine (SJTUSM), Shanghai, China
| | - Shiqian Huang
- Department of Cardiology, Shanghai Jiaotong University Affiliated Sixth People's Hospital, Shanghai Jiaotong University School of Medicine (SJTUSM), Shanghai, China.,Shanghai Institute of Immunology, Institutes of Medical Sciences, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Guofang Xia
- Department of Cardiology, Shanghai Jiaotong University Affiliated Sixth People's Hospital, Shanghai Jiaotong University School of Medicine (SJTUSM), Shanghai, China
| | - Jin Wu
- Shanghai Institute for Pediatric Research, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Yan Shen
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Ying Wang
- Shanghai Institute of Immunology, Institutes of Medical Sciences, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Rennolds S Ostrom
- Department of Biomedical and Pharmaceutical Sciences, Chapman University, Irvine, California, United States
| | - Ailian Du
- Department of Neurology, Tongren Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Chengxing Shen
- Department of Cardiology, Shanghai Jiaotong University Affiliated Sixth People's Hospital, Shanghai Jiaotong University School of Medicine (SJTUSM), Shanghai, China
| | - Congfeng Xu
- Department of Cardiology, Shanghai Jiaotong University Affiliated Sixth People's Hospital, Shanghai Jiaotong University School of Medicine (SJTUSM), Shanghai, China.,Shanghai Institute of Immunology, Institutes of Medical Sciences, Shanghai Jiaotong University School of Medicine, Shanghai, China
| |
Collapse
|