1
|
Chen G, Meng Z, Wang P. Cigarette smoke-exposed microparticles released from T lymphocytes contribute to autophagy and apoptosis dysfunction in pulmonary microvascular endothelial cells. Mol Immunol 2025; 181:9-17. [PMID: 40048930 DOI: 10.1016/j.molimm.2025.02.020] [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: 01/25/2024] [Revised: 12/13/2024] [Accepted: 02/24/2025] [Indexed: 04/07/2025]
Abstract
Cigarette smoke can cause dysfunction of the vascular endothelium; however, the underlying mechanisms have not been fully elucidated. We hypothesized that T lymphocyte-derived microparticles (TLMPs) are involved in cigarette-related diseases, especially those involving the vascular endothelium. The effect of cigarette smoke on the release of microparticles from human lymphocytes was investigated. The contributions of TLMPs induced by cigarette smoke to endothelial proliferation/apoptosis, autophagy and cytokine levels were also measured. Notably, the potential mechanism of autophagy and apoptosis dysfunction in endothelial cells was further examined. Cigarette smoke promoted the release of microparticles from T lymphocytes. TLMPs attenuated endothelial proliferation but promoted endothelial apoptosis/autophagy and the expression of proinflammatory cytokines, especially when T lymphocytes were preexposed to cigarette smoke. The potential mechanism may involve disorders of oxidative stress and STAT3 phosphorylation. In conclusion, cigarette smoke-exposed microparticles released from T lymphocytes contribute to autophagy and apoptosis dysfunction in pulmonary microvascular endothelial cells.
Collapse
Affiliation(s)
- Gang Chen
- Department of Respiratory and Critical Care Medicine, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, Henan, China.
| | - Zhaoji Meng
- Department of Immune Allergy, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Pei Wang
- Department of Respiratory and Critical Care Medicine, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
2
|
Malvi A, Khatib MN, Ganesan S, Kaur M, Srivastava M, Barwal A, Siva Prasad GV, Rajput P, Syed R, Hooda RC, Mohan B, Shabil M, Jena D, Nanda S, Aneja A, Bushi G, Mehta R, Sah R, Satapathy P, Gaidhane S. Assessing the impact of electronic nicotine delivery systems on chronic obstructive pulmonary disease: A systematic review and meta-analysis. Respir Med 2025; 241:108059. [PMID: 40157397 DOI: 10.1016/j.rmed.2025.108059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Revised: 03/17/2025] [Accepted: 03/25/2025] [Indexed: 04/01/2025]
Abstract
BACKGROUND Electronic Nicotine Delivery Systems (ENDS), commonly known as e-cigarettes or vapes, have gained widespread popularity, particularly among young adults and former smokers. While marketed as a harm reduction tool, concerns have emerged regarding their long-term impact on respiratory health, particularly their association with chronic obstructive pulmonary disease (COPD). This systematic review and meta-analysis aimed to evaluate the relationship between ENDS use and COPD risk while considering tobacco smoking as a key confounder. METHODS A comprehensive search was conducted across PubMed, Embase, and Web of Science for studies published up to September 20, 2024. Observational studies assessing the association between ENDS use and COPD risk were included. A random-effects meta-analysis was performed using R statistical software (version 4.4). Tobacco smoking, a key confounder in COPD research, was accounted for in many included studies, with adjustments varying across studies. RESULTS Fifteen studies met the inclusion criteria. The pooled odds ratio (OR) for current ENDS use and COPD risk was 1.488 (95 % CI: 1.363-1.623). Former ENDS users had an OR of 1.839 (95 % CI: 1.513-2.234), and ever-users had an OR of 1.787 (95 % CI: 1.421-2.247). Sensitivity analyses confirmed the robustness of findings, and no publication bias was detected. CONCLUSION This meta-analysis provides evidence of a significant association between ENDS use and increased COPD risk, even after adjusting for tobacco smoking. Future research should standardize smoking adjustments and investigate the independent impact of ENDS use on COPD.
Collapse
Affiliation(s)
- Ajay Malvi
- National Institute of Pharmaceutical Education and Research, Guwahati, India.
| | - Mahalaqua Nazli Khatib
- Division of Evidence Synthesis, Global Consortium of Public Health and Research, Datta Meghe Institute of Higher Education, Wardha, India.
| | - Subbulakshmi Ganesan
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to Be University), Bangalore, Karnataka, India.
| | - Mandeep Kaur
- Department of Allied Healthcare and Sciences, Vivekananda Global University, Jaipur, Rajasthan, 303012, India.
| | | | - Amit Barwal
- Chandigarh Pharmacy College, Chandigarh Group of College, Jhanjeri, Mohali, 140307, Punjab, India.
| | - G V Siva Prasad
- Department of Chemistry, Raghu Engineering College, Visakhapatnam, Andhra Pradesh, 531162, India.
| | - Pranchal Rajput
- School of Applied and Life Sciences, Division of Research and Innovation, Uttaranchal University, Dehradun, India.
| | - Rukshar Syed
- IES Institute of Pharmacy, IES University, Bhopal, Madhya Pradesh, 462044, India.
| | - Ramesh Chander Hooda
- New Delhi Institute of Management, Tughlakabad Institutional Area, New Delhi, India.
| | - Brijendra Mohan
- Department of Emergency, Graphic Era Institute of Medical Sciences, Graphic Era (Deemed to Be University), Clement Town, Dehradun, India.
| | - Muhammed Shabil
- Noida Institute of Engineering and Technology (Pharmacy Institute), Greater Noida, India; University of Cyberjaya, Persiaran Bestari, Cyber 11, 63000, Cyberjaya, Selangor Darul Ehsan, Malaysia.
| | - Diptismitha Jena
- Center for Global Health Research, Saveetha Medical College and Hospital, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India.
| | - Simranjeet Nanda
- Centre of Research Impact and Outcome, Chitkara University, Rajpura, 140417, Punjab, India.
| | - Aseem Aneja
- Chitkara Centre for Research and Development, Chitkara University, Himachal Pradesh, 174103, India.
| | - Ganesh Bushi
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, India.
| | - Rachana Mehta
- Clinical Microbiology, RDC, Manav Rachna International Institute of Research and Studies, Faridabad, Haryana, 121004, India.
| | - Renu Sah
- Department of Paediatrics, Dr. D. Y. Patil Medical College, Hospital and Research Centre, Dr. D. Y. Patil Vidyapeeth, Pune, 411018, Maharashtra, India; Department of Public Health Dentistry, Dr. D.Y. Patil Dental College and Hospital, Dr. D.Y. Patil Vidyapeeth, Pune, 411018, Maharashtra, India.
| | - Prakasini Satapathy
- University Center for Research and Development, Chandigarh University, Mohali, Punjab, India; Medical Laboratories Techniques Department, AL-Mustaqbal University, 51001, Hillah, Babil, Iraq.
| | - Shilpa Gaidhane
- One Health Centre (COHERD), Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education, Wardha, India.
| |
Collapse
|
3
|
Wang M, Hao Y, He W, Jia H, Zhong Z, Xia S. Nebulized mesenchymal stem cell-derived exosomes attenuate airway inflammation in a rat model of chronic obstructive pulmonary disease. Cell Immunol 2025; 409-410:104933. [PMID: 40020434 DOI: 10.1016/j.cellimm.2025.104933] [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: 11/25/2024] [Revised: 01/20/2025] [Accepted: 02/20/2025] [Indexed: 03/03/2025]
Abstract
Chronic Obstructive Pulmonary Disease (COPD) is one of the leading causes of death worldwide, and current treatments fail to significantly halt its progression. Exosomes derived from mesenchymal stem cells (MSCs-Exos) have demonstrated promising potential in treating COPD due to their anti-inflammatory and regenerative biological properties. In this study, we investigated the potential anti-inflammatory effects of bone marrow mesenchymal stem cell-derived exosomes (BMSCs-Exos) in a COPD rat model and the possible mechanisms by which they inhibit airway remodeling, as well as identifying the optimal dosage and administration route. Our results show that nebulized BMSC-Exos significantly improve lung function in COPD rats while reducing pulmonary inflammatory infiltration, bronchial mucus secretion, and collagen deposition. Moreover, BMSC-Exos treatment notably decreased the expression of pro-inflammatory cytokines such as TNF-α, IL-6 and IL-1β, and the pro-fibrotic factor TGF-β1 in serum, bronchoalveolar lavage fluid (BALF), and lung tissue. The most pronounced therapeutic effect was observed at a low dose of exosomes. Furthermore, quantitative real-time PCR and immunohistochemical analyses revealed that nebulized BMSC-Exos significantly inhibited airway remodeling and epithelial-mesenchymal transition (EMT) by suppressing the Wnt/β-catenin signaling pathway. In conclusion, these findings indicate that nebulized BMSC-Exos offer a noninvasive therapeutic strategy for COPD by mitigating lung inflammation and airway remodeling through the suppression of abnormal Wnt/β-catenin pathway activation induced by cigarette smoke (CS) and lipopolysaccharide (LPS) in rats.
Collapse
Affiliation(s)
- Min Wang
- Graduate School of Dalian Medical University, Dalian 116044, China
| | - Yuxin Hao
- Graduate School of Shandong First Medical University, Jinan 271016, China
| | - Wei He
- Department of Respiratory and Critical Care Medicine, Central Hospital Affiliated to Shenyang Medical College, Shenyang 110024, China
| | - Hui Jia
- Department of Respiratory and Critical Care Medicine, Central Hospital Affiliated to Shenyang Medical College, Shenyang 110024, China
| | - Zhaoshuang Zhong
- Department of Respiratory and Critical Care Medicine, Central Hospital Affiliated to Shenyang Medical College, Shenyang 110024, China
| | - Shuyue Xia
- Graduate School of Dalian Medical University, Dalian 116044, China; Department of Respiratory and Critical Care Medicine, Central Hospital Affiliated to Shenyang Medical College, Shenyang 110024, China.
| |
Collapse
|
4
|
Chitteti R, Zuniga-Hertz JP, Masso-Silva JA, Shin J, Niesman I, Bojanowski CM, Kumar AJ, Hepokoski M, Crotty Alexander LE, Patel HH, Roth DM. E-cigarette-induced changes in cell stress and mitochondrial function. Free Radic Biol Med 2025; 228:329-338. [PMID: 39756490 DOI: 10.1016/j.freeradbiomed.2025.01.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 12/22/2024] [Accepted: 01/02/2025] [Indexed: 01/07/2025]
Abstract
Inhaling aerosols from electronic nicotine delivery systems, such as e-cigarettes (e-cigs), may pose health risks beyond those caused by nicotine intake. Exposure to e-cig aerosols can lead to the release of exosomes and metabolites into the bloodstream, potentially affecting mitochondrial physiology across the body, leading to chronic inflammatory diseases. In this study we assessed the effects of e-cig use by young healthy human subjects on the circulating exosome profile and markers of cell stress, and also defined the effects of e-cig user plasma on mitochondrial function in endothelial cells (EA. Hy 926) and epithelial cells (A549) via adoptive transfer. E-cig users had altered plasma exosome profiles, with significantly increased levels of cell free mitochondrial DNA (mtDNA), protein carbonyls, and 4-HNE relative to non-users. Plasma from e-cig users decreased maximal mitochondrial respiration and spare capacity of cells, while also increasing metabolic stress, as evidenced by changes in mitochondrial phenotype from basal to stressed in both endothelial and epithelial cells, which was corroborated by electron microscopy demonstrating structural changes in mitochondria. Mitochondrial membrane potential (MMP) and reactive oxygen species (ROS) levels significantly increased in e-cig plasma-subjected cells. Overall, we identified alterations in plasma exosome profiles and increased markers of mitochondrial stress in e-cig users and evidence that circulating factors within plasma from e-cig users drives metabolic stress in endothelial and epithelial cells. Our results imply that e-cig use adversely affects mitochondrial function, leading to stress and potentially chronic inflammation across the body.
Collapse
Affiliation(s)
- Ramamurthy Chitteti
- VA San Diego Healthcare System, San Diego, CA, USA; Department of Anesthesiology, School of Medicine, University of California San Diego, USA.
| | - Juan Pablo Zuniga-Hertz
- VA San Diego Healthcare System, San Diego, CA, USA; Department of Anesthesiology, School of Medicine, University of California San Diego, USA
| | - Jorge A Masso-Silva
- VA San Diego Healthcare System, San Diego, CA, USA; Division of Pulmonary, Critical Care and Sleep Medicine, University of California San Diego, USA
| | - John Shin
- Division of Pulmonary, Critical Care and Sleep Medicine, University of California San Diego, USA
| | - Ingrid Niesman
- San Diego State University, Electron Microscope Facility, 5500 Campanile Dr, San Diego, CA, 92182, USA
| | - Christine M Bojanowski
- Division of Pulmonary, Critical Care and Sleep Medicine, University of California San Diego, USA; Division of Pulmonary and Critical Care, Tulane University, New Orleans, LA, USA
| | - Avnee J Kumar
- VA San Diego Healthcare System, San Diego, CA, USA; Division of Pulmonary, Critical Care and Sleep Medicine, University of California San Diego, USA
| | - Mark Hepokoski
- VA San Diego Healthcare System, San Diego, CA, USA; Division of Pulmonary, Critical Care and Sleep Medicine, University of California San Diego, USA
| | - Laura E Crotty Alexander
- VA San Diego Healthcare System, San Diego, CA, USA; Division of Pulmonary, Critical Care and Sleep Medicine, University of California San Diego, USA
| | - Hemal H Patel
- VA San Diego Healthcare System, San Diego, CA, USA; Department of Anesthesiology, School of Medicine, University of California San Diego, USA
| | - David M Roth
- VA San Diego Healthcare System, San Diego, CA, USA; Department of Anesthesiology, School of Medicine, University of California San Diego, USA
| |
Collapse
|
5
|
REİS R, KOLCİ K, ÖZHAN Y, COŞKUN GP, SİPAHİ H. Third-Hand Smoke Exacerbates H 2O 2-Driven Airway Responses in A549 Cells. Turk J Pharm Sci 2024; 21:376-389. [PMID: 39569661 PMCID: PMC11600328 DOI: 10.4274/tjps.galenos.2024.36153] [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: 09/13/2023] [Accepted: 01/12/2024] [Indexed: 11/22/2024]
Abstract
Objectives Third-hand smoke (THS) is residual smoke after extinguishing a cigarette and adhering to surfaces. Re-emission into the air also makes THS a health concern for those who suffer from respiratory diseases. The present study aimed to elucidate the mechanistic pathways involved in THS-induced respiratory toxicity and the accelerative potential of THS in an H2O2-induced oxidative stress model of human airway epithelia in vitro. Materials and Methods THS extracted from terrycloth exposed to 3R4F cigarettes was assessed via 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay to identify cytotoxicity. The reactive oxygen species (ROS) level was determined via 2,7-dichlorofluorescein diacetate (DCFDA) fluorescence intensity in a flow cytometer, and glutathione (GSH), malondialdehyde (MDA), and catalase (CAT) activity were assessed spectrophotometrically. Interleukin-6 (IL-6) level was measured via enzyme-linked immunosorbent assay. Results THS 50% (v/v) with significant cytotoxicity in A549 cells upregulated intracellular ROS levels via a right-shifted fluorescence intensity of DCFDA compared with the control (p < 0.05), which was also amplified with H2O2 co-treatment. MDA levels remarkably increased with THS (p < 0.05). Both THS and THS + H2O2 led to notable GSH depletion, increased CAT activity, and increased IL-6 levels, which were attenuated by the negative control (N-acetylcysteine, 1 mM) (p < 0.05). Conclusion The induction of oxidative stress may be an important event in THS-induced airway toxicity that may contribute to the progression of respiratory diseases.
Collapse
Affiliation(s)
- Rengin REİS
- Acıbadem Mehmet Ali Aydınlar University Faculty of Pharmacy, Department of Toxicology, İstanbul, Türkiye
| | - Kübra KOLCİ
- Acıbadem Mehmet Ali Aydınlar University Faculty of Pharmacy, Department of Toxicology, İstanbul, Türkiye
- Yeditepe University Faculty of Pharmacy, Department of Toxicology, İstanbul, Türkiye
| | - Yağmur ÖZHAN
- Yeditepe University Faculty of Pharmacy, Department of Toxicology, İstanbul, Türkiye
| | - Göknil Pelin COŞKUN
- Acıbadem Mehmet Ali Aydınlar University Faculty of Pharmacy, Department of Pharmaceutical Chemistry, İstanbul, Türkiye
| | - Hande SİPAHİ
- Yeditepe University Faculty of Pharmacy, Department of Toxicology, İstanbul, Türkiye
| |
Collapse
|
6
|
Ringshausen FC, Baumann I, de Roux A, Dettmer S, Diel R, Eichinger M, Ewig S, Flick H, Hanitsch L, Hillmann T, Koczulla R, Köhler M, Koitschev A, Kugler C, Nüßlein T, Ott SR, Pink I, Pletz M, Rohde G, Sedlacek L, Slevogt H, Sommerwerck U, Sutharsan S, von Weihe S, Welte T, Wilken M, Rademacher J, Mertsch P. [Management of adult bronchiectasis - Consensus-based Guidelines for the German Respiratory Society (DGP) e. V. (AWMF registration number 020-030)]. Pneumologie 2024; 78:833-899. [PMID: 39515342 DOI: 10.1055/a-2311-9450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
Bronchiectasis is an etiologically heterogeneous, chronic, and often progressive respiratory disease characterized by irreversible bronchial dilation. It is frequently associated with significant symptom burden, multiple complications, and reduced quality of life. For several years, there has been a marked global increase in the prevalence of bronchiectasis, which is linked to a substantial economic burden on healthcare systems. This consensus-based guideline is the first German-language guideline addressing the management of bronchiectasis in adults. The guideline emphasizes the importance of thoracic imaging using CT for diagnosis and differentiation of bronchiectasis and highlights the significance of etiology in determining treatment approaches. Both non-drug and drug treatments are comprehensively covered. Non-pharmacological measures include smoking cessation, physiotherapy, physical training, rehabilitation, non-invasive ventilation, thoracic surgery, and lung transplantation. Pharmacological treatments focus on the long-term use of mucolytics, bronchodilators, anti-inflammatory medications, and antibiotics. Additionally, the guideline covers the challenges and strategies for managing upper airway involvement, comorbidities, and exacerbations, as well as socio-medical aspects and disability rights. The importance of patient education and self-management is also emphasized. Finally, the guideline addresses special life stages such as transition, family planning, pregnancy and parenthood, and palliative care. The aim is to ensure comprehensive, consensus-based, and patient-centered care, taking into account individual risks and needs.
Collapse
Affiliation(s)
- Felix C Ringshausen
- Klinik für Pneumologie und Infektiologie, Medizinische Hochschule Hannover (MHH), Hannover, Deutschland
- Biomedical Research in End-Stage and Obstructive Lung Disease (BREATH), Deutsches Zentrum für Lungenforschung (DZL), Hannover, Deutschland
- European Reference Network on Rare and Complex Respiratory Diseases (ERN-LUNG), Frankfurt, Deutschland
| | - Ingo Baumann
- Hals-, Nasen- und Ohrenklinik, Universitätsklinikum Heidelberg, Heidelberg, Deutschland
| | - Andrés de Roux
- Pneumologische Praxis am Schloss Charlottenburg, Berlin, Deutschland
| | - Sabine Dettmer
- Biomedical Research in End-Stage and Obstructive Lung Disease (BREATH), Deutsches Zentrum für Lungenforschung (DZL), Hannover, Deutschland
- Institut für Diagnostische und Interventionelle Radiologie, Medizinische Hochschule Hannover (MHH), Hannover, Deutschland
| | - Roland Diel
- Institut für Epidemiologie, Universitätsklinikum Schleswig-Holstein (UKSH), Kiel, Deutschland; LungenClinic Grosshansdorf, Airway Research Center North (ARCN), Deutsches Zentrum für Lungenforschung (DZL), Grosshansdorf, Deutschland
| | - Monika Eichinger
- Klinik für Diagnostische und Interventionelle Radiologie, Thoraxklinik am Universitätsklinikum Heidelberg, Heidelberg, Deutschland; Translational Lung Research Center Heidelberg (TLRC), Deutsches Zentrum für Lungenforschung (DZL), Heidelberg, Deutschland
| | - Santiago Ewig
- Thoraxzentrum Ruhrgebiet, Kliniken für Pneumologie und Infektiologie, EVK Herne und Augusta-Kranken-Anstalt Bochum, Bochum, Deutschland
| | - Holger Flick
- Klinische Abteilung für Pulmonologie, Universitätsklinik für Innere Medizin, LKH-Univ. Klinikum Graz, Medizinische Universität Graz, Graz, Österreich
| | - Leif Hanitsch
- Institut für Medizinische Immunologie, Charité - Universitätsmedizin Berlin, Freie Universität Berlin und Humboldt-Universität zu Berlin, Berlin, Deutschland
| | - Thomas Hillmann
- Ruhrlandklinik, Westdeutsches Lungenzentrum am Universitätsklinikum Essen, Essen, Deutschland
| | - Rembert Koczulla
- Abteilung für Pneumologische Rehabilitation, Philipps Universität Marburg, Marburg, Deutschland
| | | | - Assen Koitschev
- Klinik für Hals-, Nasen-, Ohrenkrankheiten, Klinikum Stuttgart - Olgahospital, Stuttgart, Deutschland
| | - Christian Kugler
- Abteilung Thoraxchirurgie, LungenClinic Grosshansdorf, Grosshansdorf, Deutschland
| | - Thomas Nüßlein
- Klinik für Kinder- und Jugendmedizin, Gemeinschaftsklinikum Mittelrhein gGmbH, Koblenz, Deutschland
| | - Sebastian R Ott
- Pneumologie/Thoraxchirurgie, St. Claraspital AG, Basel; Universitätsklinik für Pneumologie, Allergologie und klinische Immunologie, Inselspital, Universitätsspital und Universität Bern, Bern, Schweiz
| | - Isabell Pink
- Klinik für Pneumologie und Infektiologie, Medizinische Hochschule Hannover (MHH), Hannover, Deutschland
- Biomedical Research in End-Stage and Obstructive Lung Disease (BREATH), Deutsches Zentrum für Lungenforschung (DZL), Hannover, Deutschland
- European Reference Network on Rare and Complex Respiratory Diseases (ERN-LUNG), Frankfurt, Deutschland
| | - Mathias Pletz
- Institut für Infektionsmedizin und Krankenhaushygiene, Universitätsklinikum Jena, Jena, Deutschland
| | - Gernot Rohde
- Pneumologie/Allergologie, Medizinische Klinik 1, Universitätsklinikum Frankfurt, Goethe-Universität, Frankfurt am Main, Deutschland
| | - Ludwig Sedlacek
- Institut für Medizinische Mikrobiologie und Krankenhaushygiene, Medizinische Hochschule Hannover (MHH), Hannover, Deutschland
| | - Hortense Slevogt
- Klinik für Pneumologie und Infektiologie, Medizinische Hochschule Hannover (MHH), Hannover, Deutschland
- Biomedical Research in End-Stage and Obstructive Lung Disease (BREATH), Deutsches Zentrum für Lungenforschung (DZL), Hannover, Deutschland
- Center for Individualised Infection Medicine, Hannover, Deutschland
| | - Urte Sommerwerck
- Klinik für Pneumologie, Allergologie, Schlaf- und Beatmungsmedizin, Cellitinnen-Severinsklösterchen Krankenhaus der Augustinerinnen, Köln, Deutschland
| | | | - Sönke von Weihe
- Abteilung Thoraxchirurgie, LungenClinic Grosshansdorf, Grosshansdorf, Deutschland
| | - Tobias Welte
- Klinik für Pneumologie und Infektiologie, Medizinische Hochschule Hannover (MHH), Hannover, Deutschland
- Biomedical Research in End-Stage and Obstructive Lung Disease (BREATH), Deutsches Zentrum für Lungenforschung (DZL), Hannover, Deutschland
- European Reference Network on Rare and Complex Respiratory Diseases (ERN-LUNG), Frankfurt, Deutschland
| | | | - Jessica Rademacher
- Klinik für Pneumologie und Infektiologie, Medizinische Hochschule Hannover (MHH), Hannover, Deutschland
- Biomedical Research in End-Stage and Obstructive Lung Disease (BREATH), Deutsches Zentrum für Lungenforschung (DZL), Hannover, Deutschland
- European Reference Network on Rare and Complex Respiratory Diseases (ERN-LUNG), Frankfurt, Deutschland
| | - Pontus Mertsch
- Medizinische Klinik und Poliklinik V, Klinikum der Universität München (LMU), Comprehensive Pneumology Center (CPC), Deutsches Zentrum für Lungenforschung (DZL), München, Deutschland
| |
Collapse
|
7
|
Ghojazadeh M, Pourmanaf H, Fekri V, Nikoukheslat S, Nasoudi Y, Mills DE. The effects of aerobic exercise training on inflammatory markers in adult tobacco smokers: A systematic review and meta-analysis of randomized controlled trials. Respir Med 2024; 231:107732. [PMID: 38971338 DOI: 10.1016/j.rmed.2024.107732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 06/19/2024] [Accepted: 07/03/2024] [Indexed: 07/08/2024]
Abstract
INTRODUCTION Aerobic exercise training may reduce systemic inflammation, but the effects of this on systemic inflammatory markers in adult tobacco smokers has not been systematically reviewed. Therefore, we evaluated the effects of aerobic exercise training on C-reactive protein (CRP) and tumor necrosis factor-α (TNF-α) in adult tobacco smokers using a systematic review and meta-analysis of randomized controlled trials. METHODS A comprehensive literature search was carried out using PubMed/Medline, Web of Science, EMBASE, Google Scholar, and hand search of bibliographies of the retrieved English or Persian articles up to August 2023. This review only included randomized controlled trials which investigated the effect of aerobic exercise training on CRP and TNF-α in adult smokers, based on a predefined inclusion and exclusion criteria. RESULTS A total of 1641 articles were identified. Six studies were included in the review and four evaluated CRP and two evaluated TNF-α in only males. The meta-analysis demonstrated that aerobic exercise training significantly decreased TNF-α concentrations in males (MD = -6.68, 95 % CI = -13.90 to -0.54, P = 0.05). CRP concentrations did not decrease significantly when the data from the four studies were pooled (MD = -0.17, 95 % CI = -0.37 to 0.03, P = 0.09). CONCLUSION Aerobic exercise training may reduce the concentration of TNF-α in male smokers, but it does not have a significant effect on CRP concentrations. However, these findings are based upon a small number of studies, that enrolled either exclusively male or female participants, and further investigation is necessary to increase statistical inference.
Collapse
Affiliation(s)
- Morteza Ghojazadeh
- Road Traffic Injury Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hadi Pourmanaf
- Faculty of Physical Education and Sport Sciences, University of Tabriz, Tabriz, Iran
| | - Vahid Fekri
- Faculty of Physical Education and Sport Sciences, University of Tabriz, Tabriz, Iran
| | - Saeid Nikoukheslat
- Faculty of Physical Education and Sport Sciences, University of Tabriz, Tabriz, Iran
| | - Yasmin Nasoudi
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Dean E Mills
- School of Health and Medical Sciences, University of Southern Queensland, Ipswich, Queensland, Australia; Respiratory and Exercise Physiology Research Group, School of Health and Medical Sciences, University of Southern Queensland, Ipswich, Queensland, Australia; Centre for Health Research, Institute for Resilient Regions, University of Southern Queensland, Ipswich, Queensland, Australia.
| |
Collapse
|
8
|
Jiang Y, Li Z, Jiang W, Wei T, Chen B. Risk prediction model for postoperative pneumonia in esophageal cancer patients: A systematic review. Front Oncol 2024; 14:1419633. [PMID: 39161387 PMCID: PMC11330789 DOI: 10.3389/fonc.2024.1419633] [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: 04/18/2024] [Accepted: 07/18/2024] [Indexed: 08/21/2024] Open
Abstract
Background Numerous studies have developed or validated prediction models to estimate the likelihood of postoperative pneumonia (POP) in esophageal cancer (EC) patients. The quality of these models and the evaluation of their applicability to clinical practice and future research remains unknown. This study systematically evaluated the risk of bias and applicability of risk prediction models for developing POP in patients undergoing esophageal cancer surgery. Methods PubMed, Embase, Web of Science, Cochrane Library, Cumulative Index to Nursing and Allied Health Literature (CINAHL), China National Knowledge Infrastructure (CNKI), China Science and Technology Journal Database (VIP), WanFang Database and Chinese Biomedical Literature Database were searched from inception to March 12, 2024. Two investigators independently screened the literature and extracted data. The Prediction Model Risk of Bias Assessment Tool (PROBAST) checklist was employed to evaluate both the risk of bias and applicability. Result A total of 14 studies involving 23 models were included. These studies were mainly published between 2014 and 2023. The applicability of all studies was good. However, all studies exhibited a high risk of bias, primarily attributed to inappropriate data sources, insufficient sample size, irrational treatment of variables and missing data, and lack of model validation. The incidence of POP in patients undergoing esophageal cancer surgery ranged from 14.60% to 39.26%. The most frequently used predictors were smoking, age, chronic obstructive pulmonary disease(COPD), diabetes mellitus, and methods of thoracotomy. Inter-model discrimination ranged from 0.627 to 0.850, sensitivity ranged between 60.7% and 84.0%, and specificity ranged from 59.1% to 83.9%. Conclusion In all included studies, good discrimination was reported for risk prediction models for POP in patients undergoing esophageal cancer surgery, indicating stable model performance. However, according to the PROBAST checklist, all studies had a high risk of bias. Future studies should use the predictive model assessment tool to improve study design and develop new models with larger samples and multicenter external validation. Systematic review registration https://www.crd.york.ac.uk/prospero, identifier CRD42024527085.
Collapse
Affiliation(s)
- Yaxin Jiang
- Department of Healthcare-Associated Infection Management, The Second Affiliated Hospital of Fujian University of Traditional Chinese Medicine, Fuzhou, China
- School of Nursing, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Zimeng Li
- School of Nursing, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Weiting Jiang
- Department of Healthcare-Associated Infection Management, The Second Affiliated Hospital of Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Tingyu Wei
- School of Nursing, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Bizhen Chen
- Department of Healthcare-Associated Infection Management, The Second Affiliated Hospital of Fujian University of Traditional Chinese Medicine, Fuzhou, China
| |
Collapse
|
9
|
Tangedal S, Nielsen R, Aanerud M, Drengenes C, Husebø GR, Lehmann S, Knudsen KS, Hiemstra PS, Eagan TM. Lower airway microbiota in COPD and healthy controls. Thorax 2024:thorax-2023-220455. [PMID: 38331579 DOI: 10.1136/thorax-2023-220455] [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: 05/08/2023] [Accepted: 01/08/2024] [Indexed: 02/10/2024]
Abstract
BACKGROUND The lower airway microbiota in patients with chronic obstructive pulmonary disease (COPD) are likely altered compared with the microbiota in healthy individuals. Information on how the microbiota is affected by smoking, use of inhaled corticosteroids (ICS) and COPD severity is still scarce. METHODS In the MicroCOPD Study, participant characteristics were obtained through standardised questionnaires and clinical measurements at a single centre from 2012 to 2015. Protected bronchoalveolar lavage samples from 97 patients with COPD and 97 controls were paired-end sequenced with the Illumina MiSeq System. Data were analysed in QIIME 2 and R. RESULTS Alpha-diversity was lower in patients with COPD than controls (Pielou evenness: COPD=0.76, control=0.80, p=0.004; Shannon entropy: COPD=3.98, control=4.34, p=0.01). Beta-diversity differed with smoking only in the COPD cohort (weighted UniFrac: permutational analysis of variance R2=0.04, p=0.03). Nine genera were differentially abundant between COPD and controls. Genera enriched in COPD belonged to the Firmicutes phylum. Pack years were linked to differential abundance of taxa in controls only (ANCOM-BC (Analysis of Compositions of Microbiomes with Bias Correction) log-fold difference/q-values: Haemophilus -0.05/0.048; Lachnoanaerobaculum -0.04/0.03). Oribacterium was absent in smoking patients with COPD compared with non-smoking patients (ANCOM-BC log-fold difference/q-values: -1.46/0.03). We found no associations between the microbiota and COPD severity or ICS. CONCLUSION The lower airway microbiota is equal in richness in patients with COPD to controls, but less even. Genera from the Firmicutes phylum thrive particularly in COPD airways. Smoking has different effects on diversity and taxonomic abundance in patients with COPD compared with controls. COPD severity and ICS use were not linked to the lower airway microbiota.
Collapse
Affiliation(s)
- Solveig Tangedal
- Department of Thoracic Medicine, Haukeland University Hospital, Bergen, Norway
- Department of Clinical Science, Faculty of Medicine, University of Bergen, Bergen, Norway
| | - Rune Nielsen
- Department of Thoracic Medicine, Haukeland University Hospital, Bergen, Norway
- Department of Clinical Science, Faculty of Medicine, University of Bergen, Bergen, Norway
| | - Marianne Aanerud
- Department of Thoracic Medicine, Haukeland University Hospital, Bergen, Norway
- Department of Clinical Science, Faculty of Medicine, University of Bergen, Bergen, Norway
| | - Christine Drengenes
- Department of Clinical Science, Faculty of Medicine, University of Bergen, Bergen, Norway
| | - Gunnar R Husebø
- Department of Thoracic Medicine, Haukeland University Hospital, Bergen, Norway
| | - Sverre Lehmann
- Department of Thoracic Medicine, Haukeland University Hospital, Bergen, Norway
- Department of Clinical Science, Faculty of Medicine, University of Bergen, Bergen, Norway
| | - Kristel S Knudsen
- Department of Thoracic Medicine, Haukeland University Hospital, Bergen, Norway
| | - Pieter S Hiemstra
- Department of Pulmonology, Leiden University Medical Center, Leiden, The Netherlands
| | - Tomas Ml Eagan
- Department of Thoracic Medicine, Haukeland University Hospital, Bergen, Norway
- Department of Clinical Science, Faculty of Medicine, University of Bergen, Bergen, Norway
| |
Collapse
|
10
|
Shipman JG, Onyenwoke RU, Sivaraman V. Vaping-Dependent Pulmonary Inflammation Is Ca 2+ Mediated and Potentially Sex Specific. Int J Mol Sci 2024; 25:1785. [PMID: 38339063 PMCID: PMC10855597 DOI: 10.3390/ijms25031785] [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: 12/15/2023] [Revised: 01/23/2024] [Accepted: 01/25/2024] [Indexed: 02/12/2024] Open
Abstract
Here we use the SCIREQ InExpose system to simulate a biologically relevant vaping model in mice to investigate the role of calcium signaling in vape-dependent pulmonary disease as well as to investigate if there is a gender-based difference of disease. Male and female mice were vaped with JUUL Menthol (3% nicotine) using the SCIREQ InExpose system for 2 weeks. Additionally, 2-APB, a known calcium signaling inhibitor, was administered as a prophylactic for lung disease and damage caused by vaping. After 2 weeks, mice were exposed to lipopolysaccharide (LPS) to mimic a bacterial infection. Post-infection (24 h), mice were sacrificed, and bronchoalveolar lavage fluid (BALF) and lungs were taken. Vaping primed the lungs for worsened disease burden after microbial challenge (LPS) for both males and females, though females presented increased neutrophilia and inflammatory cytokines post-vape compared to males, which was assessed by flow cytometry, and cytokine and histopathological analysis. This increased inflammatory burden was controlled by calcium signaling inhibition, suggesting that calcium dysregulation may play a role in lung injury caused by vaping in a gender-dependent manner.
Collapse
Affiliation(s)
- Jeffrey G. Shipman
- Department of Biological & Biomedical Sciences, North Carolina Central University, Durham, NC 27707, USA; (J.G.S.); (R.U.O.)
| | - Rob U. Onyenwoke
- Department of Biological & Biomedical Sciences, North Carolina Central University, Durham, NC 27707, USA; (J.G.S.); (R.U.O.)
- Biomanufacturing Research Institute and Technology Enterprise (BRITE), North Carolina Central University, Durham, NC 27707, USA
| | - Vijay Sivaraman
- Department of Biological & Biomedical Sciences, North Carolina Central University, Durham, NC 27707, USA; (J.G.S.); (R.U.O.)
- The Julius L. Chambers Biomedical/Biotechnology Research Institute, North Carolina Central University, Durham, NC 27707, USA
| |
Collapse
|
11
|
Chen Y, Wu J, Li R, Kang W, Zhao A, Yin Y, Tong S, Yuan J, Li S. Individual and joint association of phenols, parabens, and phthalates with childhood lung function: Exploring the mediating role of peripheral immune responses. JOURNAL OF HAZARDOUS MATERIALS 2023; 454:131457. [PMID: 37099904 DOI: 10.1016/j.jhazmat.2023.131457] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 04/17/2023] [Accepted: 04/19/2023] [Indexed: 05/19/2023]
Abstract
The functioning of the respiratory system can be interfered with by exposure to mixtures of environmental chemicals, however, the evidence is still ambiguous. We evaluated the association of exposure to mixtures of 14 chemicals, including 2 phenols, 2 parabens, and 10 phthalates, with four major lung function metrics. Based on data from the National Health and Nutrition Examination Survey 2007-2012, this analysis was conducted among 1462 children aged 6-19 years. Linear regression, Bayesian kernel machine regression, quantile-based g-computation regression, and a generalized additive model were performed to estimate the associations. Mediation analyses were performed to investigate plausible biological pathways mediated by immune cells. Our results indicated that the phenols, parabens, and phthalates mixture was negatively related to lung function parameters. And BPA and PP were identified as important contributors to negative associations with FEV1, FVC, and PEF, with non-linear relationships observed between BPA and those outcomes. The most influential factor for a probable FEF25-75 % decline was MCNP. BPA, and MCNP had an interaction effect on FEF25-75 %. The association of PP with FVC and FEV1 has been postulated to be mediated by neutrophils and monocytes. The findings offer insights into the associations of chemical mixtures with respiratory health and the possible driving mechanism, which would be of significance in adding novel evidence of the role of peripheral immune responses, as well as calling for remediation actions to be prioritized during childhood.
Collapse
Affiliation(s)
- Yiting Chen
- School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jinhong Wu
- Department of Respiratory Medicine, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Rong Li
- School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wenhui Kang
- School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Anda Zhao
- Department of Nutrition, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yong Yin
- Department of Respiratory Medicine, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shilu Tong
- School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Department of Biostatistics, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China; School of Public Health, Institute of Environment and Population Health, Anhui Medical University, Hefei, China; School of Public Health and Social Work, Queensland University of Technology, Brisbane, Australia
| | - Jiajun Yuan
- Child Health Advocacy Institute, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shenghui Li
- School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China; MOE-Shanghai Key Laboratory of Children's Environmental Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
12
|
Reifenberg J, Gecili E, Pestian T, Andrinopoulou ER, Ryan PH, Brokamp C, Collaco JM, Szczesniak RD. Lung function and secondhand smoke exposure among children with cystic fibrosis: A Bayesian meta-analysis. J Cyst Fibros 2023; 22:694-701. [PMID: 37142525 PMCID: PMC10524940 DOI: 10.1016/j.jcf.2023.04.020] [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: 02/12/2023] [Revised: 04/14/2023] [Accepted: 04/24/2023] [Indexed: 05/06/2023]
Abstract
BACKGROUND Secondhand smoke exposure, an important environmental health factor in cystic fibrosis (CF), remains uniquely challenging to children with CF as they strive to maintain pulmonary function during early stages of growth and throughout adolescence. Despite various epidemiologic studies among CF populations, little has been done to coalesce estimates of the association between secondhand smoke exposure and lung function decline. METHODS A systematic review was performed using PRISMA guidelines. A Bayesian random-effects model was employed to estimate the association between secondhand smoke exposure and change in lung function (measured as FEV1% predicted). RESULTS Quantitative synthesis of study estimates indicated that second-hand smoke exposure corresponded to a significant drop in FEV1 (estimated decrease: -5.11% predicted; 95% CI: -7.20, -3.47). The estimate of between-study heterogeneity was 1.32% predicted (95% CI: 0.05, 4.26). There was moderate heterogeneity between the 6 analyzed studies that met review criteria (degree of heterogeneity: I2=61.9% [95% CI: 7.3-84.4%] and p = 0.022 from the frequentist method.) CONCLUSIONS: Our results quantify the impact at the pediatric population level and corroborate the assertion that secondhand smoke exposure negatively affects pulmonary function in children with CF. Findings highlight challenges and opportunities for future environmental health interventions in pediatric CF care.
Collapse
Affiliation(s)
| | - Emrah Gecili
- Division of Biostatistics & Epidemiology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Ave (MLC 5041), Cincinnati, OH 45229, United States; Department of Pediatrics, University of Cincinnati, Cincinnati, OH, United States
| | - Teresa Pestian
- Department of Anesthesia, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States
| | - Eleni-Rosalina Andrinopoulou
- Department of Biostatistics, Erasmus Medical Center, Rotterdam, the Netherlands; Department of Epidemiology, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Patrick H Ryan
- Division of Biostatistics & Epidemiology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Ave (MLC 5041), Cincinnati, OH 45229, United States; Department of Pediatrics, University of Cincinnati, Cincinnati, OH, United States
| | - Cole Brokamp
- Division of Biostatistics & Epidemiology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Ave (MLC 5041), Cincinnati, OH 45229, United States; Department of Pediatrics, University of Cincinnati, Cincinnati, OH, United States
| | - Joseph M Collaco
- Department of Pediatrics, Johns Hopkins University, Baltimore, MD, United States
| | - Rhonda D Szczesniak
- Division of Biostatistics & Epidemiology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Ave (MLC 5041), Cincinnati, OH 45229, United States; Department of Pediatrics, University of Cincinnati, Cincinnati, OH, United States; Division of Pulmonary Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States.
| |
Collapse
|
13
|
Cheng TY, Chang CC, Luo CS, Chen KY, Yeh YK, Zheng JQ, Wu SM. Targeting Lung-Gut Axis for Regulating Pollution Particle-Mediated Inflammation and Metabolic Disorders. Cells 2023; 12:901. [PMID: 36980242 PMCID: PMC10047528 DOI: 10.3390/cells12060901] [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: 12/27/2022] [Revised: 03/09/2023] [Accepted: 03/13/2023] [Indexed: 03/17/2023] Open
Abstract
Cigarette smoking (CS) or ambient particulate matter (PM) exposure is a risk factor for metabolic disorders, such as insulin resistance (IR), increased plasma triglycerides, hyperglycemia, and diabetes mellitus (DM); it can also cause gut microbiota dysbiosis. In smokers with metabolic disorders, CS cessation decreases the risks of serious pulmonary events, inflammation, and metabolic disorder. This review included recent studies examining the mechanisms underlying the effects of CS and PM on gut microbiota dysbiosis and metabolic disorder development; one of the potential mechanisms is the disruption of the lung-gut axis, leading to gut microbiota dysbiosis, intestinal dysfunction, systemic inflammation, and metabolic disease. Short-chain fatty acids (SCFAs) are the primary metabolites of gut bacteria, which are derived from the fermentation of dietary fibers. They activate G-protein-coupled receptor (GPCR) signaling, suppress histone deacetylase (HDAC) activity, and inhibit inflammation, facilitating the maintenance of gut health and biofunction. The aforementioned gut microbiota dysbiosis reduces SCFA levels. Treatment targeting SCFA/GPCR signaling may alleviate air pollution-associated inflammation and metabolic disorders, which involve lung-gut axis disruption.
Collapse
Affiliation(s)
- Tzu-Yu Cheng
- Division of Cardiovascular Surgery, Department of Surgery, Wan Fang Hospital, Taipei Medical University, Taipei 11696, Taiwan;
- Division of Cardiology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Chih-Cheng Chang
- Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City 23561, Taiwan; (C.-C.C.); (C.-S.L.); (K.-Y.C.); (Y.-K.Y.); (J.-Q.Z.)
- Division of Pulmonary Medicine, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
- TMU Research Center for Thoracic Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Ching-Shan Luo
- Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City 23561, Taiwan; (C.-C.C.); (C.-S.L.); (K.-Y.C.); (Y.-K.Y.); (J.-Q.Z.)
- Division of Pulmonary Medicine, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
- TMU Research Center for Thoracic Medicine, Taipei Medical University, Taipei 11031, Taiwan
- International Ph.D. Program in Cell Therapy and Regenerative Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Kuan-Yuan Chen
- Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City 23561, Taiwan; (C.-C.C.); (C.-S.L.); (K.-Y.C.); (Y.-K.Y.); (J.-Q.Z.)
- Division of Pulmonary Medicine, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
- TMU Research Center for Thoracic Medicine, Taipei Medical University, Taipei 11031, Taiwan
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Yun-Kai Yeh
- Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City 23561, Taiwan; (C.-C.C.); (C.-S.L.); (K.-Y.C.); (Y.-K.Y.); (J.-Q.Z.)
- TMU Research Center for Thoracic Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Jing-Quan Zheng
- Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City 23561, Taiwan; (C.-C.C.); (C.-S.L.); (K.-Y.C.); (Y.-K.Y.); (J.-Q.Z.)
- Division of Pulmonary Medicine, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
- TMU Research Center for Thoracic Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Sheng-Ming Wu
- Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City 23561, Taiwan; (C.-C.C.); (C.-S.L.); (K.-Y.C.); (Y.-K.Y.); (J.-Q.Z.)
- Division of Pulmonary Medicine, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
- TMU Research Center for Thoracic Medicine, Taipei Medical University, Taipei 11031, Taiwan
| |
Collapse
|
14
|
Increased serum cotinine and obesity negatively impact asthma exacerbations and hospitalizations: A cross-sectional analysis of NHANES. J Clin Transl Sci 2023; 7:e10. [PMID: 36755538 PMCID: PMC9879909 DOI: 10.1017/cts.2022.509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 11/17/2022] [Accepted: 11/18/2022] [Indexed: 12/12/2022] Open
Abstract
Background Asthma is the most common non-communicable chronic airway disease worldwide. Obesity and cigarette use independently increase asthma morbidity and mortality. Current literature suggests that obesity and smoking synergistically increase asthma-related wheezing. Objective To assess whether increased serum cotinine and obesity act synergistically to increase the likelihood of having an asthma exacerbation, emergency department (ED) visit, or hospitalization. Methods A cross-sectional analysis of the 2011-2015 iterations of NHANES database was performed. Patients aged 18 years or greater with asthma were included. Serum cotinine was utilized as an accurate measurement of cigarette use. Logistic regression models were constructed to determine whether elevated serum cotinine and obesity were associated with self-reported asthma exacerbations, asthma-specific ED usage, and hospitalizations for any reason in the past year. Odds ratios were adjusted for age, gender, race, and ethnicity. Interactions were assessed by multiplying the adjusted effect sizes for elevated cotinine and obesity. Results We identified 2179 (N = 32,839,290) patients with asthma, of which 32.2% were active smokers and 42.7% were obese. Patients with an elevated cotinine and asthma were significantly more likely to have had an asthma-related ED visit in the past year (adjusted odds ratio [AOR] 1.82; 95% CI 1.19-2.79), have a physician-prescribed asthma medication (AOR 2.04; 95% CI 1.11-3.74), and have a hospitalization for any reason (AOR 3.65; 95% CI 1.88-7.07) compared to those with low cotinine. Patients with asthma and obesity were more likely to have an asthma-related ED visit (AOR 1.67; 95% CI 1.06-2.62) or hospitalization for any reason in the past year compared to non-obese patients (AOR 2.76; 95% CI 1.69-4.5). However, a statistically significant interaction between obesity and cotinine was only identified in patients who currently have asthma compared to a previous asthma diagnosis (AOR 1.76; 95% CI 1.10-2.82). There were no synergistic interactions among ED usage or asthma exacerbations. Conclusion Nearly one-third of patients with asthma were current smokers, and almost half were obese. This study identified elevated serum cotinine, a metabolite of cigarette use, and obesity as key risk factors for asthma exacerbations, asthma-related ED visits, and hospitalizations for any reason. Elevated serum cotinine and obesity were not found to act synergistically in increasing asthma exacerbations or ED visits. However, the presence of both risk factors increased the risk of currently having asthma (compared to a previous diagnosis) by 76%. Serum cotinine may be useful in predicting asthma outcomes.
Collapse
|
15
|
Arreola-Ramírez JL, Vargas MH, Carbajal V, Alquicira-Mireles J, Montaño M, Ramos-Abraham C, Ortiz-Quintero B, Torres-Machorro AL, Rodríguez-Velasco A, Esquivel-Campos AL, Vásquez-Vásquez JA, Segura-Medina P. Mesenchymal stem cells attenuate the proinflammatory cytokine pattern in a guinea pig model of chronic cigarette smoke exposure. Cytokine 2023; 162:156104. [PMID: 36493630 DOI: 10.1016/j.cyto.2022.156104] [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: 07/12/2022] [Revised: 11/16/2022] [Accepted: 11/26/2022] [Indexed: 12/13/2022]
Abstract
AIMS Cigarette smoke often induces pulmonary and systemic inflammation. In animal models, mesenchymal stem cells (MSC) tend to ameliorate these effects. We aimed to explore the local and systemic expression of cytokines in guinea pigs chronically exposed to cigarette smoke, and their modifications by MSC. MAIN METHODS Concentrations of IL-1β, IL-6, IL-8, IL-12, TNF-α, INF-ɣ, TSG-6, MMP-9, TIMP-1, and/or TIMP-2 in serum and bronchoalveolar lavage (BALF) from animals exposed to tobacco smoke (20 cigarettes/day, 5 days/week for 10 weeks) were determined, and mRNA expression of some of them was measured in lung tissue. Intratracheal instillation of allogeneic bone marrow MSC (5x106 cells in 1 ml) was done at week 2. KEY FINDINGS After cigarette smoke, IL-6 and IFN-γ increased in serum and BALF, while IL-1β and IL-12 decreased in serum, and TSG-6 and TIMP-2 increased in BALF. IL-1β had a paradoxical increase in BALF. MSC had an almost null effect in unexposed animals. The intratracheal administration of MSC in guinea pigs exposed to cigarette smoke was associated with a statistically significant decrease of IL-12 and TSG-6 in serum, as well as a decrease of IL-1β and IFN-γ and an increase in TIMP-1 in BALF. Concerning mRNA expression in lung tissue, cigarette smoke did not modify the relative amount of the studied transcripts, but even so, MSC decreased the IL-12 mRNA and increased the TIMP-1 mRNA. SIGNIFICANCE A single intratracheal instillation of MSC reduces the pulmonary and systemic proinflammatory pattern induced by chronic exposure to cigarette smoke in guinea pigs. TRIAL REGISTRATION Not applicable.
Collapse
Affiliation(s)
- José Luis Arreola-Ramírez
- Departamento de Investigación en Hiperreactividad Bronquial, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Calzada de Tlalpan 4502, CP 14080, Mexico City, Mexico.
| | - Mario H Vargas
- Departamento de Investigación en Hiperreactividad Bronquial, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Calzada de Tlalpan 4502, CP 14080, Mexico City, Mexico
| | - Verónica Carbajal
- Departamento de Investigación en Hiperreactividad Bronquial, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Calzada de Tlalpan 4502, CP 14080, Mexico City, Mexico
| | - Jesús Alquicira-Mireles
- Departamento de Investigación en Hiperreactividad Bronquial, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Calzada de Tlalpan 4502, CP 14080, Mexico City, Mexico
| | - Martha Montaño
- Departamento de Investigación en Fibrosis Pulmonar, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Calzada de Tlalpan 4502, CP 14080, Mexico City, Mexico
| | - Carlos Ramos-Abraham
- Departamento de Investigación en Fibrosis Pulmonar, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Calzada de Tlalpan 4502, CP 14080, Mexico City, Mexico
| | - Blanca Ortiz-Quintero
- Departamento de Investigación en Bioquímica, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Calzada de Tlalpan 4502, CP 14080, Mexico City, Mexico
| | - Ana Lilia Torres-Machorro
- Departamento de Investigación en Fibrosis Pulmonar, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Calzada de Tlalpan 4502, CP 14080, Mexico City, Mexico
| | - Alicia Rodríguez-Velasco
- Servicio de Anatomía Patológica, Hospital de Pediatría, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Av. Cuauhtémoc 330, CP 06720, Mexico City, Mexico
| | - Ana Laura Esquivel-Campos
- Laboratorio de Investigación en Biología Experimental, Universidad Autónoma Metropolitana, Mexico City, Mexico
| | | | - Patricia Segura-Medina
- Departamento de Investigación en Hiperreactividad Bronquial, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Calzada de Tlalpan 4502, CP 14080, Mexico City, Mexico; Escuela de Medicina y Ciencias de la Salud, Tecnológico de Monterrey, Mexico City, Mexico
| |
Collapse
|
16
|
Serrano Gotarredona MP, Navarro Herrero S, Gómez Izquierdo L, Rodríguez Portal JA. Smoking-related interstitial lung disease. RADIOLOGIA 2022; 64 Suppl 3:277-289. [PMID: 36737166 DOI: 10.1016/j.rxeng.2022.10.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Accepted: 10/22/2022] [Indexed: 02/05/2023]
Abstract
Exposure to smoke is associated with the development of diseases of the airways and lung parenchyma. Apart from chronic obstructive pulmonary disease (COPD), in some individuals, tobacco smoke can also trigger mechanisms of interstitial damage that result in various pathological changes and pulmonary fibrosis. A causal relation has been established between tobacco smoke and a group of entities that includes respiratory bronchiolitis-associated interstitial lung disease (RB-ILD), desquamative interstitial pneumonia (DIP), Langerhans cell histiocytosis (LCH), and acute eosinophilic pneumonia (AEP). Smoking is considered a risk factor for idiopathic pulmonary fibrosis (IPF); however, the role and impact of smoking in the development of this differentiated clinical entity, which has also been called combined pulmonary fibrosis and emphysema (CPFE) as well as nonspecific interstitial pneumonia (NIP), remains to be determined. The definition of smoking-related interstitial fibrosis (SRIF) is relatively recent, with differentiated histological characteristics. The likely interconnection between the mechanisms involved in inflammation and pulmonary fibrosis in all these processes often results in an overlapping of clinical, radiological, and histological features in the same patient that can sometimes lead to radiological patterns of interstitial lung disease that are impossible to classify. For this reason, a combined approach to diagnosis is recommendable. This combined approach should be based on the joint interpretation of the histological and radiological findings while taking the clinical context into consideration. This paper aims to describe the high-resolution computed tomography (HRCT) findings in this group of disease entities in correlation with the clinical manifestations and histological changes underlying the radiological pattern.
Collapse
Affiliation(s)
- M P Serrano Gotarredona
- Unidad de Imagen Cardiotorácica, Servicio de Radiodiagnóstico, Hospital Universitario Virgen del Rocío, Sevilla, Spain.
| | - S Navarro Herrero
- Unidad de Imagen Cardiotorácica, Servicio de Radiodiagnóstico, Hospital Universitario Virgen del Rocío, Sevilla, Spain.
| | - L Gómez Izquierdo
- Servicio de Anatomía Patológica, Hospital Universitario Virgen del Rocío, Sevilla, Spain
| | - J A Rodríguez Portal
- Unidad de Patología Intersticial, Servicio de Neumología, Hospital Universitario Virgen del Rocío, Sevilla, Spain
| |
Collapse
|
17
|
White A, Wang Z, Wang X, King M, Guo C, Mantsounga C, Ayala A, Morrison AR, Choudhary G, Sellke F, Chambers E, Ware LB, Rounds S, Lu Q. NLRP3 inflammasome activation in cigarette smoke priming for Pseudomonas aeruginosa-induced acute lung injury. Redox Biol 2022; 57:102467. [PMID: 36175355 PMCID: PMC9618465 DOI: 10.1016/j.redox.2022.102467] [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: 10/01/2021] [Revised: 08/30/2022] [Accepted: 09/02/2022] [Indexed: 02/04/2023] Open
Abstract
It is increasingly recognized that cigarette smoke (CS) exposure increases the incidence and severity of acute respiratory distress syndrome (ARDS) in critical ill humans and animals. However, the mechanism(s) is not well understood. This study aims to investigate mechanism underlying the priming effect of CS on Pseudomonas aeruginosa-triggered acute lung injury, by using pre-clinic animal models and genetically modified mice. We demonstrated that CS impaired P. aeruginosa-induced mitophagy flux, promoted p62 accumulation, and exacerbated P. aeruginosa-triggered mitochondrial damage and NLRP3 inflammasome activation in alveolar macrophages; an effect associated with increased acute lung injury and mortality. Pharmacological inhibition of caspase-1, a component of inflammasome, attenuated CS primed P. aeruginosa-triggered acute lung injury and improved animal survival. Global or myeloid-specific knockout of IL-1β, a downstream component of inflammasome activation, also attenuated CS primed P. aeruginosa-triggered acute lung injury. Our results suggest that NLRP3 inflammasome activation is an important mechanism for CS primed P. aeruginosa-triggered acute lung injury. (total words: 155).
Collapse
Affiliation(s)
- Alexis White
- Vascular Research Laboratory, Providence Veterans Affairs Medical Center, Providence, RI, USA
| | - Zhengke Wang
- Vascular Research Laboratory, Providence Veterans Affairs Medical Center, Providence, RI, USA
| | - Xing Wang
- Vascular Research Laboratory, Providence Veterans Affairs Medical Center, Providence, RI, USA
| | - Michelle King
- Vascular Research Laboratory, Providence Veterans Affairs Medical Center, Providence, RI, USA
| | - Cynthia Guo
- Vascular Research Laboratory, Providence Veterans Affairs Medical Center, Providence, RI, USA
| | - Chris Mantsounga
- Vascular Research Laboratory, Providence Veterans Affairs Medical Center, Providence, RI, USA
| | - Alfred Ayala
- Department of Surgery, The Warren Alpert Medical School of Brown University and Lifespan-Rhode Island Hospital, Providence, RI, USA
| | - Alan R Morrison
- Vascular Research Laboratory, Providence Veterans Affairs Medical Center, Providence, RI, USA; Department of Medicine, The Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Gaurav Choudhary
- Vascular Research Laboratory, Providence Veterans Affairs Medical Center, Providence, RI, USA; Department of Medicine, The Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Frank Sellke
- Cardiothoracic Surgery, The Warren Alpert Medical School of Brown University and Lifespan-Rhode Island Hospital, Providence, RI, USA
| | - Eboni Chambers
- Vascular Research Laboratory, Providence Veterans Affairs Medical Center, Providence, RI, USA
| | - Lorraine B Ware
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Sharon Rounds
- Vascular Research Laboratory, Providence Veterans Affairs Medical Center, Providence, RI, USA; Department of Medicine, The Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Qing Lu
- Vascular Research Laboratory, Providence Veterans Affairs Medical Center, Providence, RI, USA; Department of Medicine, The Warren Alpert Medical School of Brown University, Providence, RI, USA.
| |
Collapse
|
18
|
Serrano Gotarredona M, Navarro Herrero S, Gómez Izquierdo L, Rodríguez Portal J. Enfermedades pulmonares intersticiales relacionadas con el tabaco. RADIOLOGIA 2022. [DOI: 10.1016/j.rx.2022.10.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
19
|
Bagale K, Kulkarni R. A Systematic Review of the Literature Examining the Effects of Cigarette Smoke and e-Cigarette Vapor on the Virulence of Human Pathogenic Bacteria. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:12518. [PMID: 36231813 PMCID: PMC9565164 DOI: 10.3390/ijerph191912518] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 09/22/2022] [Accepted: 09/28/2022] [Indexed: 06/16/2023]
Abstract
The bioactive chemicals in cigarette smoke (CS) and e-cigarette vapor (EV) may affect pathogenic bacteria in the nasopharyngeal microflora, which may have implications on the pathophysiology of respiratory infections in cigarette smokers and e-cigarette users. In this systematic review, we seek to synthesize the research evidence supporting this hypothesis. To address the central research question, "what is known from the published, peer-reviewed literature about the effects of cigarette smoke or e-cigarette vapor exposure on the physiology of human pathogenic bacteria?", we screened the PubMed®, Web of ScienceTM, and ScienceDirect databases for reports examining the virulence characteristics and gene expression in human pathogenic bacteria exposed to either CS or EV. The principal conclusion from our analysis is that exposure to either CS or EV induces the virulence of respiratory pathogenic bacteria in a strain-dependent manner, which may in turn facilitate respiratory infections in cigarette smokers and e-cigarette users. In addition, we present evidence that nicotine and reactive oxygen species are the main chemicals responsible for CS/EV-mediated alterations in bacterial physiology. We note limitations that this review does not examine reports describing the alterations in host respiratory physiology or nasopharyngeal dysbiosis caused by CS/EV exposure. Future research to determine whether CS/EV-mediated augmentation of bacterial virulence indeed plays a role in human respiratory tract infections is warranted.
Collapse
Affiliation(s)
| | - Ritwij Kulkarni
- Department of Biology, University of Louisiana at Lafayette, Lafayette, LA 70504, USA
| |
Collapse
|
20
|
Spagnolo P, Tonelli R, Samarelli AV, Castelli G, Cocconcelli E, Petrarulo S, Cerri S, Bernardinello N, Clini E, Saetta M, Balestro E. The role of immune response in the pathogenesis of idiopathic pulmonary fibrosis: far beyond the Th1/Th2 imbalance. Expert Opin Ther Targets 2022; 26:617-631. [PMID: 35983984 DOI: 10.1080/14728222.2022.2114897] [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: 11/04/2022]
Abstract
INTRODUCTION . Idiopathic pulmonary fibrosis (IPF) is a chronic disease of unknown origin characterized by progressive scarring of the lung leading to irreversible loss of function. Despite the availability of two drugs that are able to slow down disease progression, IPF remains a deadly disease. The pathogenesis of IPF is poorly understood, but a dysregulated wound healing response following recurrent alveolar epithelial injury is thought to be crucial. Areas covered. In the last few years, the role of the immune system in IPF pathobiology has been reconsidered; indeed, recent data suggest that a dysfunctional immune system may promote and unfavorable interplay with pro-fibrotic pathways thus acting as a cofactor in disease development and progression. In this article, we review and critically discuss the role of T cells in the pathogenesis and progression of IPF in the attempt to highlight ways in which further research in this area may enable the development of targeted immunomodulatory therapies for this dreadful disease. EXPERT OPINION A better understanding of T cells interactions has the potential to facilitate the development of immune modulators targeting multiple T cell-mediated pathways thus halting disease initiation and progression.
Collapse
Affiliation(s)
- Paolo Spagnolo
- Respiratory Disease Unit, Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Padova, Italy
| | - Roberto Tonelli
- Laboratory of Cell Therapies and Respiratory Medicine, Department of Medical and Surgical Sciences for Children & Adults University Hospital of Modena and Reggio Emilia, Modena, Italy.,University Hospital of Modena, Respiratory Diseases Unit, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Modena, Italy.,Clinical and Experimental Medicine PhD Program, University of Modena and Reggio Emilia, Modena, Italy
| | - Anna Valeria Samarelli
- Laboratory of Cell Therapies and Respiratory Medicine, Department of Medical and Surgical Sciences for Children & Adults University Hospital of Modena and Reggio Emilia, Modena, Italy.,University Hospital of Modena, Respiratory Diseases Unit, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Gioele Castelli
- Respiratory Disease Unit, Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Padova, Italy
| | - Elisabetta Cocconcelli
- Respiratory Disease Unit, Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Padova, Italy
| | - Simone Petrarulo
- Respiratory Disease Unit, Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Padova, Italy
| | - Stefania Cerri
- Laboratory of Cell Therapies and Respiratory Medicine, Department of Medical and Surgical Sciences for Children & Adults University Hospital of Modena and Reggio Emilia, Modena, Italy.,University Hospital of Modena, Respiratory Diseases Unit, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Nicol Bernardinello
- Respiratory Disease Unit, Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Padova, Italy
| | - Enrico Clini
- Laboratory of Cell Therapies and Respiratory Medicine, Department of Medical and Surgical Sciences for Children & Adults University Hospital of Modena and Reggio Emilia, Modena, Italy.,University Hospital of Modena, Respiratory Diseases Unit, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Marina Saetta
- Respiratory Disease Unit, Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Padova, Italy
| | - Elisabetta Balestro
- Respiratory Disease Unit, Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Padova, Italy
| |
Collapse
|
21
|
Wang X, Murugesan P, Zhang P, Xu S, Peng L, Wang C, Cai H. NADPH Oxidase Isoforms in COPD Patients and Acute Cigarette Smoke-Exposed Mice: Induction of Oxidative Stress and Lung Inflammation. Antioxidants (Basel) 2022; 11:antiox11081539. [PMID: 36009258 PMCID: PMC9405243 DOI: 10.3390/antiox11081539] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Revised: 07/25/2022] [Accepted: 07/26/2022] [Indexed: 11/16/2022] Open
Abstract
Cigarette smoke (CS) is a major risk factor for chronic obstructive pulmonary disease (COPD), which represents the third leading cause of death worldwide. CS induces reactive oxygen species (ROS) production, leading to pulmonary inflammation and remodeling. NADPH oxidases (NOXs) represent essential sources of ROS production in the cardiovascular system. Whether and how NOX isoforms are activated in COPD patients and in response to acute cigarette smoke (ACS) remains incompletely understood. In the present study, the expression of NOX isoforms was examined in the lungs of end-stage COPD patients. In addition, mice silenced of NOX1 or NOX4 expression using in vivo RNA interference (RNAi), and NOX2-deficient (NOX2−/y) mice, were exposed to ACS for 1 h using a standard TE-10B smoking machine. In lung sections isolated from COPD patients undergoing lung transplantation, protein expression of NOX1, NOX2, NOX4, or NOX5 was markedly upregulated compared to non-smoking donor controls. Likewise, ACS upregulated protein expression of NOX1, NOX2, and NOX4, production of ROS, inflammatory cell infiltration, and mRNA expression of proinflammatory cytokines TNF-α and KC in the mouse lung. In vivo RNAi knockdown of NOX1 or NOX4 decreased ACS induced ROS production, inflammatory cell influx, and the expression of TNF-α and KC, which were accompanied by inhibition of the NF-κB-COX-2 axis. Although ACS induced ROS production was reduced in the lungs of NOX2−/y mice, inflammatory cell influx and expression of NF-κB/COX-2 were increased. Taken together, our results demonstrate for the first time that NOX isoforms 1, 2, 4 and 5 all remain activated in end-stage COPD patients, while NOX1 and NOX4 mediate oxidative stress and inflammatory responses in response to acute cigarette smoke. Therefore, targeting different isoforms of NOX might be necessary to treat COPD at different stages of the disease, which represents novel mechanistic insights enabling improved management of the devastating disease.
Collapse
Affiliation(s)
- Xinjing Wang
- Department of Pulmonary and Critical Care Medicine, China-Japan Friendship Hospital, Capital Medical University, Beijing 100069, China
| | - Priya Murugesan
- Department of Anesthesiology, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Pan Zhang
- Department of Pulmonary and Critical Care Medicine, China-Japan Friendship Hospital, Capital Medical University, Beijing 100069, China
| | - Shiqing Xu
- Department of Pulmonary and Critical Care Medicine, China-Japan Friendship Hospital, Capital Medical University, Beijing 100069, China
| | - Liang Peng
- Department of Pulmonary and Critical Care Medicine, China-Japan Friendship Hospital, Capital Medical University, Beijing 100069, China
| | - Chen Wang
- Department of Pulmonary and Critical Care Medicine, China-Japan Friendship Hospital, Capital Medical University, Beijing 100069, China
- Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
- Correspondence: (C.W.); (H.C.)
| | - Hua Cai
- Department of Anesthesiology, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
- Correspondence: (C.W.); (H.C.)
| |
Collapse
|
22
|
Keskinidou C, Vassiliou AG, Dimopoulou I, Kotanidou A, Orfanos SE. Mechanistic Understanding of Lung Inflammation: Recent Advances and Emerging Techniques. J Inflamm Res 2022; 15:3501-3546. [PMID: 35734098 PMCID: PMC9207257 DOI: 10.2147/jir.s282695] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 05/04/2022] [Indexed: 12/12/2022] Open
Abstract
Acute respiratory distress syndrome (ARDS) is a life-threatening lung injury characterized by an acute inflammatory response in the lung parenchyma. Hence, it is considered as the most appropriate clinical syndrome to study pathogenic mechanisms of lung inflammation. ARDS is associated with increased morbidity and mortality in the intensive care unit (ICU), while no effective pharmacological treatment exists. It is very important therefore to fully characterize the underlying pathobiology and the related mechanisms, in order to develop novel therapeutic approaches. In vivo and in vitro models are important pre-clinical tools in biological and medical research in the mechanistic and pathological understanding of the majority of diseases. In this review, we will present data from selected experimental models of lung injury/acute lung inflammation, which have been based on clinical disorders that can lead to the development of ARDS and related inflammatory lung processes in humans, including ventilation-induced lung injury (VILI), sepsis, ischemia/reperfusion, smoke, acid aspiration, radiation, transfusion-related acute lung injury (TRALI), influenza, Streptococcus (S.) pneumoniae and coronaviruses infection. Data from the corresponding clinical conditions will also be presented. The mechanisms related to lung inflammation that will be covered are oxidative stress, neutrophil extracellular traps, mitogen-activated protein kinase (MAPK) pathways, surfactant, and water and ion channels. Finally, we will present a brief overview of emerging techniques in the field of omics research that have been applied to ARDS research, encompassing genomics, transcriptomics, proteomics, and metabolomics, which may recognize factors to help stratify ICU patients at risk, predict their prognosis, and possibly, serve as more specific therapeutic targets.
Collapse
Affiliation(s)
- Chrysi Keskinidou
- First Department of Critical Care Medicine and Pulmonary Services, School of Medicine, National and Kapodistrian University of Athens, "Evangelismos" Hospital, Athens, Greece
| | - Alice G Vassiliou
- First Department of Critical Care Medicine and Pulmonary Services, School of Medicine, National and Kapodistrian University of Athens, "Evangelismos" Hospital, Athens, Greece
| | - Ioanna Dimopoulou
- First Department of Critical Care Medicine and Pulmonary Services, School of Medicine, National and Kapodistrian University of Athens, "Evangelismos" Hospital, Athens, Greece
| | - Anastasia Kotanidou
- First Department of Critical Care Medicine and Pulmonary Services, School of Medicine, National and Kapodistrian University of Athens, "Evangelismos" Hospital, Athens, Greece
| | - Stylianos E Orfanos
- First Department of Critical Care Medicine and Pulmonary Services, School of Medicine, National and Kapodistrian University of Athens, "Evangelismos" Hospital, Athens, Greece
| |
Collapse
|
23
|
Wold LE, Tarran R, Crotty Alexander LE, Hamburg NM, Kheradmand F, St Helen G, Wu JC. Cardiopulmonary Consequences of Vaping in Adolescents: A Scientific Statement From the American Heart Association. Circ Res 2022; 131:e70-e82. [PMID: 35726609 DOI: 10.1161/res.0000000000000544] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Although the US Food and Drug Administration has not approved e-cigarettes as a cessation aid, industry has at times positioned their products in that way for adults trying to quit traditional cigarettes; however, their novelty and customizability have driven them into the hands of unintended users, particularly adolescents. Most new users of e-cigarette products have never smoked traditional cigarettes; therefore, understanding the respiratory and cardiovascular consequences of e-cigarette use has become of increasing interest to the research community. Most studies have been performed on adult e-cigarette users, but the majority of these study participants are either former traditional smokers or smokers who have used e-cigarettes to switch from traditional smoking. Therefore, the respiratory and cardiovascular consequences in this population are not attributable to e-cigarette use alone. Preclinical studies have been used to study the effects of naive e-cigarette use on various organ systems; however, almost all of these studies have used adult animals, which makes translation of health effects to adolescents problematic. Given that inhalation of any foreign substance can have effects on the respiratory and cardiovascular systems, a more holistic understanding of the pathways involved in toxicity could help to guide researchers to novel therapeutic treatment strategies. The goals of this scientific statement are to provide salient background information on the cardiopulmonary consequences of e-cigarette use (vaping) in adolescents, to guide therapeutic and preventive strategies and future research directions, and to inform public policymakers on the risks, both short and long term, of vaping.
Collapse
|
24
|
Moshensky A, Brand CS, Alhaddad H, Shin J, Masso-Silva JA, Advani I, Gunge D, Sharma A, Mehta S, Jahan A, Nilaad S, Olay J, Gu W, Simonson T, Almarghalani D, Pham J, Perera S, Park K, Al-Kolla R, Moon H, Das S, Byun MK, Shah Z, Sari Y, Heller Brown J, Crotty Alexander LE. Effects of mango and mint pod-based e-cigarette aerosol inhalation on inflammatory states of the brain, lung, heart, and colon in mice. eLife 2022; 11:e67621. [PMID: 35411847 PMCID: PMC9005188 DOI: 10.7554/elife.67621] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Accepted: 03/04/2022] [Indexed: 12/13/2022] Open
Abstract
While health effects of conventional tobacco are well defined, data on vaping devices, including one of the most popular e-cigarettes which have high nicotine levels, are less established. Prior acute e-cigarette studies have demonstrated inflammatory and cardiopulmonary physiology changes while chronic studies have demonstrated extra-pulmonary effects, including neurotransmitter alterations in reward pathways. In this study we investigated the impact of inhalation of aerosols produced from pod-based, flavored e-cigarettes (JUUL) aerosols three times daily for 3 months on inflammatory markers in the brain, lung, heart, and colon. JUUL aerosol exposure induced upregulation of cytokine and chemokine gene expression and increased HMGB1 and RAGE in the nucleus accumbens in the central nervous system. Inflammatory gene expression increased in the colon, while gene expression was more broadly altered by e-cigarette aerosol inhalation in the lung. Cardiopulmonary inflammatory responses to acute lung injury with lipopolysaccharide were exacerbated in the heart. Flavor-specific findings were detected across these studies. Our findings suggest that daily e-cigarette use may cause neuroinflammation, which may contribute to behavioral changes and mood disorders. In addition, e-cigarette use may cause gut inflammation, which has been tied to poor systemic health, and cardiac inflammation, which leads to cardiovascular disease.
Collapse
Affiliation(s)
- Alex Moshensky
- Pulmonary and Critical Care Section, VA San Diego Healthcare SystemLa JollaUnited States
- Division of Pulmonary, Critical Care and Sleep Medicine and Section of Physiology, Department of Medicine, University of California San Diego (UCSD)San DiegoUnited States
| | - Cameron S Brand
- Department of Pharmacology, University of California San Diego (UCSD)San DiegoUnited States
| | - Hasan Alhaddad
- Department of Pharmacology and Experimental Therapeutics, College of Pharmacy and Pharmaceutical Sciences, University of ToledoToledoUnited States
| | - John Shin
- Pulmonary and Critical Care Section, VA San Diego Healthcare SystemLa JollaUnited States
- Division of Pulmonary, Critical Care and Sleep Medicine and Section of Physiology, Department of Medicine, University of California San Diego (UCSD)San DiegoUnited States
| | - Jorge A Masso-Silva
- Pulmonary and Critical Care Section, VA San Diego Healthcare SystemLa JollaUnited States
- Division of Pulmonary, Critical Care and Sleep Medicine and Section of Physiology, Department of Medicine, University of California San Diego (UCSD)San DiegoUnited States
| | - Ira Advani
- Pulmonary and Critical Care Section, VA San Diego Healthcare SystemLa JollaUnited States
- Division of Pulmonary, Critical Care and Sleep Medicine and Section of Physiology, Department of Medicine, University of California San Diego (UCSD)San DiegoUnited States
| | - Deepti Gunge
- Pulmonary and Critical Care Section, VA San Diego Healthcare SystemLa JollaUnited States
- Division of Pulmonary, Critical Care and Sleep Medicine and Section of Physiology, Department of Medicine, University of California San Diego (UCSD)San DiegoUnited States
| | - Aditi Sharma
- Department of Pathology, University of California San Diego (UCSD)San DiegoUnited States
| | - Sagar Mehta
- Pulmonary and Critical Care Section, VA San Diego Healthcare SystemLa JollaUnited States
- Division of Pulmonary, Critical Care and Sleep Medicine and Section of Physiology, Department of Medicine, University of California San Diego (UCSD)San DiegoUnited States
| | - Arya Jahan
- Pulmonary and Critical Care Section, VA San Diego Healthcare SystemLa JollaUnited States
- Division of Pulmonary, Critical Care and Sleep Medicine and Section of Physiology, Department of Medicine, University of California San Diego (UCSD)San DiegoUnited States
| | - Sedtavut Nilaad
- Pulmonary and Critical Care Section, VA San Diego Healthcare SystemLa JollaUnited States
- Division of Pulmonary, Critical Care and Sleep Medicine and Section of Physiology, Department of Medicine, University of California San Diego (UCSD)San DiegoUnited States
| | - Jarod Olay
- Pulmonary and Critical Care Section, VA San Diego Healthcare SystemLa JollaUnited States
- Division of Pulmonary, Critical Care and Sleep Medicine and Section of Physiology, Department of Medicine, University of California San Diego (UCSD)San DiegoUnited States
| | - Wanjun Gu
- Division of Pulmonary, Critical Care and Sleep Medicine and Section of Physiology, Department of Medicine, University of California San Diego (UCSD)San DiegoUnited States
| | - Tatum Simonson
- Division of Pulmonary, Critical Care and Sleep Medicine and Section of Physiology, Department of Medicine, University of California San Diego (UCSD)San DiegoUnited States
| | - Daniyah Almarghalani
- Department of Pharmacology and Experimental Therapeutics, College of Pharmacy and Pharmaceutical Sciences, University of ToledoToledoUnited States
| | - Josephine Pham
- Pulmonary and Critical Care Section, VA San Diego Healthcare SystemLa JollaUnited States
- Division of Pulmonary, Critical Care and Sleep Medicine and Section of Physiology, Department of Medicine, University of California San Diego (UCSD)San DiegoUnited States
| | - Samantha Perera
- Pulmonary and Critical Care Section, VA San Diego Healthcare SystemLa JollaUnited States
- Division of Pulmonary, Critical Care and Sleep Medicine and Section of Physiology, Department of Medicine, University of California San Diego (UCSD)San DiegoUnited States
| | - Kenneth Park
- Pulmonary and Critical Care Section, VA San Diego Healthcare SystemLa JollaUnited States
- Division of Pulmonary, Critical Care and Sleep Medicine and Section of Physiology, Department of Medicine, University of California San Diego (UCSD)San DiegoUnited States
| | - Rita Al-Kolla
- Pulmonary and Critical Care Section, VA San Diego Healthcare SystemLa JollaUnited States
- Division of Pulmonary, Critical Care and Sleep Medicine and Section of Physiology, Department of Medicine, University of California San Diego (UCSD)San DiegoUnited States
| | - Hoyoung Moon
- Department of Pharmacology, University of California San Diego (UCSD)San DiegoUnited States
| | - Soumita Das
- Department of Pathology, University of California San Diego (UCSD)San DiegoUnited States
| | - Min Kwang Byun
- Division of Pulmonary, Critical Care and Sleep Medicine and Section of Physiology, Department of Medicine, University of California San Diego (UCSD)San DiegoUnited States
- Division of Pulmonology, Department of Internal Medicine, Gangnam Severance Hospital, Yonsei University College of MedicineSeoulRepublic of Korea
| | - Zahoor Shah
- Department of Medicinal and Biological Chemistry, College of Pharmacy and Pharmaceutical Sciences, University of ToledoToledoUnited States
| | - Youssef Sari
- Department of Pharmacology and Experimental Therapeutics, College of Pharmacy and Pharmaceutical Sciences, University of ToledoToledoUnited States
| | - Joan Heller Brown
- Department of Pharmacology, University of California San Diego (UCSD)San DiegoUnited States
| | - Laura E Crotty Alexander
- Pulmonary and Critical Care Section, VA San Diego Healthcare SystemLa JollaUnited States
- Division of Pulmonary, Critical Care and Sleep Medicine and Section of Physiology, Department of Medicine, University of California San Diego (UCSD)San DiegoUnited States
| |
Collapse
|
25
|
Cytotoxic and Inflammatory Effects of Electronic and Traditional Cigarettes on Oral Gingival Cells Using a Novel Automated Smoking Instrument: An In Vitro Study. TOXICS 2022; 10:toxics10040179. [PMID: 35448440 PMCID: PMC9032316 DOI: 10.3390/toxics10040179] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 03/31/2022] [Accepted: 04/04/2022] [Indexed: 02/04/2023]
Abstract
Information about the potential oral health effects of vaping from electronic cigarettes (e-cigs) is still sparse and inconsistent. The purpose of this study was to compare the safety and cytotoxicity of e-cig liquid aerosols versus traditional cigarette (t-cig) smoke on human epithelial oral cells. T-cig smoke and e-cig aerosols were generated by a newly developed automated smoking instrument in order to simulate realistic user puffing behaviors. Air−liquid interface transwell cell cultures were exposed to standardized puff topography (puff duration: 2 s, puff volume: 35 mL, puff frequency: 1 puff every 60 s) of reference t-cigs or commercially available e-cigs at different air dilutions. Cell viability, morphology, and death rate were evaluated with MTT and TUNEL assays. The inflammatory cytokine gene expression of inflammatory genes was assessed by quantitative RT-PCR. E-cigs and t-cigs indicated similar adverse effects by enhancing cytotoxicity and cell death in a dose-dependent manner. E-cig aerosol and t-cig smoke treatment expressed upregulation of inflammatory cytokines up to 3.0-fold (p < 0.05). These results indicate that e-cig smoking may contribute to oral tissue−cell damage and tissue inflammation. Our approach allows the production of e-cig aerosol and t-cig smoke in order to identify harmful effects in oral tissues in vitro.
Collapse
|
26
|
Gomez N, James V, Onion D, Fairclough LC. Extracellular vesicles and chronic obstructive pulmonary disease (COPD): a systematic review. Respir Res 2022; 23:82. [PMID: 35382831 PMCID: PMC8985325 DOI: 10.1186/s12931-022-01984-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 03/12/2022] [Indexed: 12/15/2022] Open
Abstract
Background Chronic Obstructive Pulmonary Disease (COPD) is a common inflammatory disease of the airways characterized by irreversible airflow limitation, ranking the third highest cause of death worldwide. Extracellular vesicles (EVs) are important intercellular communication mediators released by cells into their extracellular environment with the capacity to transfer biological signals. EVs involved in COPD hold great potential to understand disease pathogenesis and identify important biomarkers. This systematic review aims to examine all available research on EVs in the pathogenesis and diagnosis of COPD to identify existing knowledge and support further research within the field. Methods Publications were searched using PubMed and EMBASE with the search terms (Exosomes or extracellular vesicles or microvesicles or microparticles or ectosomes) AND (chronic obstructive pulmonary disease or COPD or emphysema or bronchitis). Results Initial search yielded 512 papers of which 142 were manually selected for review and 43 were eligible for analyses. The studies were divided into groups according to the role of EVs in pathogenesis, EV origin and cargo, their role in COPD exacerbations and their diagnostic utility. EVs were found to be involved in the mechanism of pathogenesis of COPD, derived from various cell types, as well as containing modified levels of miRNAs. EVs also varied according to the pathophysiological status of disease, therefore presenting a possible method for COPD diagnosis and progress monitoring. Conclusion The current findings show the limited but good quality research looking at the role of EVs in COPD, demonstrating the need for more studies to better define and provide further insight into the functional characteristics of EV in COPD pathogenesis. Supplementary Information The online version contains supplementary material available at 10.1186/s12931-022-01984-0.
Collapse
Affiliation(s)
- Nancy Gomez
- School of Life Sciences, The University of Nottingham, Life Sciences Building, Nottingham, NG7 2RD, UK
| | - Victoria James
- School of Veterinary Medicine and Science, The University of Nottingham, Nottingham, NG7 2UH, UK
| | - David Onion
- School of Life Sciences, The University of Nottingham, Life Sciences Building, Nottingham, NG7 2RD, UK
| | - Lucy C Fairclough
- School of Life Sciences, The University of Nottingham, Life Sciences Building, Nottingham, NG7 2RD, UK.
| |
Collapse
|
27
|
Safety and Feasibility of Lung Cancer Surgery under the COVID-19 Circumstance. Cancers (Basel) 2022; 14:cancers14051334. [PMID: 35267642 PMCID: PMC8909353 DOI: 10.3390/cancers14051334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Accepted: 02/25/2022] [Indexed: 11/29/2022] Open
Abstract
Simple Summary The global coronavirus disease 2019 (COVID-19) pandemic has drastically changed the current practice of medicine worldwide. As more clinical data is collected and processed, we are beginning to have an understanding of which patients are more at risk for severe complications of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infections. Preliminary data has shown that patients with lung cancer are disproportionally affected by the current COVID-19 pandemic. Furthermore, studies have shown that lung cancer patients are also significantly more likely to be admitted to the ICU and need mechanical ventilation. A specific subset of patients that are even more at risk for severe COVID-19 are those that require lung cancer surgery. To minimize the risk of SARS-CoV-2 infections in patients undergoing surgery, new treatment guidelines and preventive measures are necessary. In this review, we summarize the latest evidence regarding recommendations for patients undergoing lung cancer surgery in the COVID-19 era. Abstract The current coronavirus disease 2019 (COVID-19) pandemic has forced healthcare providers worldwide to adapt their practices. Our understanding of the effects of COVID-19 has increased exponentially since the beginning of the pandemic. Data from large-scale, international registries has provided more insight regarding risk factors for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infections and has allowed us to delineate specific subgroups of patients that have higher risks for severe complications. One particular subset of patients that have significantly higher risks of SARS-CoV-2 infection with higher morbidity and mortality rates are those that require surgical treatment for lung cancer. Earlier studies have shown that COVID-19 infections in patients that underwent lung cancer surgery is associated with higher rates of respiratory failure and mortality. However, deferral of cancer treatments is associated with increased mortality as well. This creates difficult situations in which healthcare providers are forced to weigh the benefits of surgical treatment against the possibility of SARS-CoV-2 infections. A number of oncological and surgical organizations have proposed treatment guidelines and recommendations for patients planned for lung cancer surgery. In this review, we summarize the latest data and recommendations for patients undergoing lung cancer surgery in the COVID-19 circumstance.
Collapse
|
28
|
Araújo NPDS, de Matos NA, Oliveira M, de Souza ABF, Castro TDF, Machado-Júnior PA, de Souza DMS, Talvani A, Cangussú SD, de Menezes RCA, Bezerra FS. Quercetin Improves Pulmonary Function and Prevents Emphysema Caused by Exposure to Cigarette Smoke in Male Mice. Antioxidants (Basel) 2022; 11:antiox11020181. [PMID: 35204064 PMCID: PMC8868486 DOI: 10.3390/antiox11020181] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 12/22/2021] [Accepted: 01/05/2022] [Indexed: 12/12/2022] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is the major cause of morbidity and mortality worldwide, and cigarette smoke is a key factor in the development of COPD. Thus, the development of effective therapies to prevent the advancement of COPD has become increasingly essential. We hypothesized that quercetin protects lungs in mice exposed to long-term cigarette smoke. Thirty-five C57BL/6 mice were exposed to cigarette smoke (12 cigarettes per day) for 60 days and pretreated with 10 mg/kg/day of quercetin via orogastric gavage. After the experimental protocol, the animals were euthanized and samples were collected for histopathological, antioxidant defense, oxidative stress and inflammatory analysis. The animals exposed to cigarette smoke showed an increase in respiratory rate and hematological parameters, cell influx into the airways, oxidative damage and inflammatory mediators, besides presenting with alterations in the pulmonary histoarchitecture. The animals receiving 10 mg/kg/day of quercetin that were exposed to cigarette smoke presented a reduction in cellular influx, less oxidative damage, reduction in cytokine levels, improvement in the histological pattern and improvement in pulmonary emphysema compared to the group that was only exposed to cigarette smoke. These results suggest that quercetin may be an agent in preventing pulmonary emphysema induced by cigarette smoke.
Collapse
Affiliation(s)
- Natália Pereira da Silva Araújo
- Laboratory of Experimental Pathophysiology, Department of Biological Sciences and Center of Research in Biological Sciences, Federal University of Ouro Preto (UFOP), Ouro Preto 35400-000, Brazil; (N.P.d.S.A.); (N.A.d.M.); (M.O.); (A.B.F.d.S.); (T.d.F.C.); (P.A.M.-J.); (S.D.C.)
| | - Natália Alves de Matos
- Laboratory of Experimental Pathophysiology, Department of Biological Sciences and Center of Research in Biological Sciences, Federal University of Ouro Preto (UFOP), Ouro Preto 35400-000, Brazil; (N.P.d.S.A.); (N.A.d.M.); (M.O.); (A.B.F.d.S.); (T.d.F.C.); (P.A.M.-J.); (S.D.C.)
| | - Michel Oliveira
- Laboratory of Experimental Pathophysiology, Department of Biological Sciences and Center of Research in Biological Sciences, Federal University of Ouro Preto (UFOP), Ouro Preto 35400-000, Brazil; (N.P.d.S.A.); (N.A.d.M.); (M.O.); (A.B.F.d.S.); (T.d.F.C.); (P.A.M.-J.); (S.D.C.)
| | - Ana Beatriz Farias de Souza
- Laboratory of Experimental Pathophysiology, Department of Biological Sciences and Center of Research in Biological Sciences, Federal University of Ouro Preto (UFOP), Ouro Preto 35400-000, Brazil; (N.P.d.S.A.); (N.A.d.M.); (M.O.); (A.B.F.d.S.); (T.d.F.C.); (P.A.M.-J.); (S.D.C.)
| | - Thalles de Freitas Castro
- Laboratory of Experimental Pathophysiology, Department of Biological Sciences and Center of Research in Biological Sciences, Federal University of Ouro Preto (UFOP), Ouro Preto 35400-000, Brazil; (N.P.d.S.A.); (N.A.d.M.); (M.O.); (A.B.F.d.S.); (T.d.F.C.); (P.A.M.-J.); (S.D.C.)
| | - Pedro Alves Machado-Júnior
- Laboratory of Experimental Pathophysiology, Department of Biological Sciences and Center of Research in Biological Sciences, Federal University of Ouro Preto (UFOP), Ouro Preto 35400-000, Brazil; (N.P.d.S.A.); (N.A.d.M.); (M.O.); (A.B.F.d.S.); (T.d.F.C.); (P.A.M.-J.); (S.D.C.)
| | - Débora Maria Soares de Souza
- Laboratory of Immunobiology of Inflammation, Department of Biological Sciences, Institute of Exact and Biological Sciences, Federal University of Ouro Preto (UFOP), Ouro Preto 35400-000, Brazil; (D.M.S.d.S.); (A.T.)
| | - André Talvani
- Laboratory of Immunobiology of Inflammation, Department of Biological Sciences, Institute of Exact and Biological Sciences, Federal University of Ouro Preto (UFOP), Ouro Preto 35400-000, Brazil; (D.M.S.d.S.); (A.T.)
| | - Sílvia Dantas Cangussú
- Laboratory of Experimental Pathophysiology, Department of Biological Sciences and Center of Research in Biological Sciences, Federal University of Ouro Preto (UFOP), Ouro Preto 35400-000, Brazil; (N.P.d.S.A.); (N.A.d.M.); (M.O.); (A.B.F.d.S.); (T.d.F.C.); (P.A.M.-J.); (S.D.C.)
| | - Rodrigo Cunha Alvim de Menezes
- Laboratory of Cardiovascular Physiology, Department of Biological Sciences and Center of Research in Biological Sciences, Federal University of Ouro Preto (UFOP), Ouro Preto 35400-000, Brazil;
| | - Frank Silva Bezerra
- Laboratory of Experimental Pathophysiology, Department of Biological Sciences and Center of Research in Biological Sciences, Federal University of Ouro Preto (UFOP), Ouro Preto 35400-000, Brazil; (N.P.d.S.A.); (N.A.d.M.); (M.O.); (A.B.F.d.S.); (T.d.F.C.); (P.A.M.-J.); (S.D.C.)
- Correspondence:
| |
Collapse
|
29
|
Dorna MS, Barbosa EMS, Callegari MA, Tanni SE, Chiuso-Minicucci F, Felix TF, Seneda AL, Correa CR, Fernandes AAH, Azevedo PS, Polegato BF, Rogero MM, Paiva SAR, Zornoff LAM, Reis PP, Minicucci MF. Orange Juice Attenuates Circulating miR-150-5p, miR-25-3p, and miR-451a in Healthy Smokers: A Randomized Crossover Study. Front Nutr 2022; 8:775515. [PMID: 35004810 PMCID: PMC8740272 DOI: 10.3389/fnut.2021.775515] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 12/09/2021] [Indexed: 12/20/2022] Open
Abstract
Introduction: Tobacco smoke is associated with oxidative and inflammatory pathways, increasing the risk of chronic-degenerative diseases. Our goal was to evaluate the effects of acute “Pera” and “Moro” orange juice consumption on inflammatory processes and oxidative stress in microRNA (miRNA) expression in plasma from healthy smokers. Methods: This was a randomized crossover study that included healthy smokers over 18 years old. Blood samples were collected before and 11 h after beverage ingestion. Participants were instructed to drink 400 mL of Pera orange juice (Citrus sinensis), Moro orange juice (Citrus sinensis L. Osbeck), or water. Each subject drank the beverages in a 3-way crossover study design. Inflammatory and oxidative stress biomarkers and circulating miRNA expression profiles were determined. The subjects maintained their usual tobacco exposure during the experiment. Results: We included 18 individuals (12 men and 6 women), with 37.0 ± 12.0 years old. All subjects received the 3 interventions. Increased expression of circulating miRNAs (miR-150-5p, miR-25-3p, and miR-451a) was verified after cigarette smoking, which were attenuated after intake of both types of orange juice. There was no difference regarding serum levels of TNF-α, IL-6, MMP-9, and C-reactive protein. Despite the increased activity of serum superoxide dismutase and glutathione peroxidase after “Pera” or “Moro” orange juice intake, respectively, no changes in lipid hydroperoxide levels were detected. Conclusion: Tobaccos smokers showed increased expression of miR-150-5p, miR-25-3p, and miR-451a was noted, and attenuated by orange juice intake. miRNAs were predicted to regulate 244 target genes with roles in oxidative stress, PI3K-Akt, and MAPK signaling, which are pathways frequently involved in smoking-related cardiovascular diseases and cancer.
Collapse
Affiliation(s)
- Mariana S Dorna
- Internal Medicine Department, Botucatu Medical School, São Paulo State University, UNESP, Botucatu, Brazil
| | - Elizabete M S Barbosa
- Internal Medicine Department, Botucatu Medical School, São Paulo State University, UNESP, Botucatu, Brazil
| | - Matheus A Callegari
- Internal Medicine Department, Botucatu Medical School, São Paulo State University, UNESP, Botucatu, Brazil
| | - Suzana E Tanni
- Internal Medicine Department, Botucatu Medical School, São Paulo State University, UNESP, Botucatu, Brazil
| | - Fernanda Chiuso-Minicucci
- Internal Medicine Department, Botucatu Medical School, São Paulo State University, UNESP, Botucatu, Brazil
| | - Tainara F Felix
- Experimental Research Unit, São Paulo State University, UNESP, Botucatu, Brazil
| | - Ana L Seneda
- Experimental Research Unit, São Paulo State University, UNESP, Botucatu, Brazil
| | - Camila R Correa
- Botucatu Medical School, São Paulo State University, UNESP, Botucatu, Brazil
| | - Ana A H Fernandes
- Chemistry and Biochemistry Department, Institute of Biosciences, São Paulo State University, UNESP, Botucatu, Brazil
| | - Paula S Azevedo
- Internal Medicine Department, Botucatu Medical School, São Paulo State University, UNESP, Botucatu, Brazil
| | - Bertha F Polegato
- Internal Medicine Department, Botucatu Medical School, São Paulo State University, UNESP, Botucatu, Brazil
| | - Marcelo M Rogero
- Department of Nutrition, School of Public Health, USP - University of São Paulo, São Paulo, Brazil
| | - Sergio A R Paiva
- Internal Medicine Department, Botucatu Medical School, São Paulo State University, UNESP, Botucatu, Brazil
| | - Leonardo A M Zornoff
- Internal Medicine Department, Botucatu Medical School, São Paulo State University, UNESP, Botucatu, Brazil
| | - Patricia P Reis
- Experimental Research Unit, São Paulo State University, UNESP, Botucatu, Brazil.,Department of Surgery and Orthopedics, São Paulo State University, UNESP, Botucatu, Brazil
| | - Marcos F Minicucci
- Internal Medicine Department, Botucatu Medical School, São Paulo State University, UNESP, Botucatu, Brazil
| |
Collapse
|
30
|
Wu X, Wang C, Li H, Meng H, Jie J, Fu M, Bai Y, Li G, Wei W, Feng Y, Li M, Guan X, He M, Zhang X, Guo H. Circulating white blood cells and lung function impairment: the observational studies and Mendelian randomization analysis. Ann Med 2021; 53:1118-1128. [PMID: 34259107 PMCID: PMC8280897 DOI: 10.1080/07853890.2021.1948603] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Accepted: 06/22/2021] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND Circulating white blood cell (WBC) counts have been related to lung function impairment, but causal relationship was not established. We aimed to evaluate independent effects and causal relationships of WBC subtypes with lung function. METHODS The 19,159 participants from NHANES 2011-2012 (n = 3570), coke-oven workers (COW, n = 1762) and Dongfeng-Tongji (DFTJ, n = 13,827) cohorts were included in the observational studies. The associations between circulating counts of WBC subtypes and prebronchodilator lung function were evaluated by linear regression models and LASSO regression was used to select effective WBC subtypes. Summary statistics for WBC-associated SNPs were extracted from literature, and Mendelian randomization (MR) analysis with inverse-variance weighted (IVW) method was applied to estimate the causal effects of total WBC and subtypes on lung function among 4012 subjects from COW (n = 1126) and DFTJ cohorts (n = 2886). RESULTS Total WBC counts were negatively associated with lung function among three populations and their pooled analysis indicated that per 1 × 109 cells/L increase in total WBC was associated with 36.13 (95% CI: 30.35, 41.91) mL and 25.23 (95% CI: 19.97, 30.50) mL decrease in FVC and FEV1, respectively. Independent associations with lung function were found for neutrophils, monocytes, eosinophils and basophils (all p < .05), except lymphocytes. Besides, IVW MR analysis showed that genetically predicted total WBC and neutrophil counts were associated with reduced FVC (p = .017 and .021, respectively) and FEV1 (p = .048 and .043, respectively). CONCLUSIONS WBC subtypes were independently associated with lower lung function except lymphocytes. Our findings suggest that circulating neutrophils may be causal factors in lung function impairment.KEY MESSAGESWhite blood cell (WBC) subtypes were negatively associated with lung function level except lymphocytes in the observational studies.Associations of WBC subtypes with lung function may be modified by sex and smoking.Mendelian randomization analysis shows that neutrophils may be causal factors in lung function impairment.
Collapse
Affiliation(s)
- Xiulong Wu
- Department of Occupational and Environmental Health, State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chenming Wang
- Department of Occupational and Environmental Health, State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hang Li
- Department of Occupational and Environmental Health, State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hua Meng
- Department of Occupational and Environmental Health, State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jiali Jie
- Department of Occupational and Environmental Health, State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ming Fu
- Department of Occupational and Environmental Health, State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yansen Bai
- Department of Occupational and Environmental Health, State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Guyanan Li
- Department of Occupational and Environmental Health, State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wei Wei
- Department of Occupational and Environmental Health, State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yue Feng
- Department of Occupational and Environmental Health, State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Mengying Li
- Department of Occupational and Environmental Health, State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xin Guan
- Department of Occupational and Environmental Health, State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Meian He
- Department of Occupational and Environmental Health, State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaomin Zhang
- Department of Occupational and Environmental Health, State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Huan Guo
- Department of Occupational and Environmental Health, State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
31
|
Shipman JG, Onyenwoke RU, Sivaraman V. Calcium-Dependent Pulmonary Inflammation and Pharmacological Interventions and Mediators. BIOLOGY 2021; 10:1053. [PMID: 34681152 PMCID: PMC8533358 DOI: 10.3390/biology10101053] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 10/11/2021] [Accepted: 10/14/2021] [Indexed: 12/17/2022]
Abstract
Pulmonary diseases present a significant burden worldwide and lead to severe morbidity and mortality. Lung inflammation caused by interactions with either viruses, bacteria or fungi is a prominent characteristic of many pulmonary diseases. Tobacco smoke and E-cig use ("vaping") are considered major risk factors in the development of pulmonary disease as well as worsening disease prognosis. However, at present, relatively little is known about the mechanistic actions by which smoking and vaping may worsen the disease. One theory suggests that long-term vaping leads to Ca2+ signaling dysregulation. Ca2+ is an important secondary messenger in signal transduction. Cellular Ca2+ concentrations are mediated by a complex series of pumps, channels, transporters and exchangers that are responsible for triggering various intracellular processes such as cell death, proliferation and secretion. In this review, we provide a detailed understating of the complex series of components that mediate Ca2+ signaling and how their dysfunction may result in pulmonary disease. Furthermore, we summarize the recent literature investigating the negative effects of smoking and vaping on pulmonary disease, cell toxicity and Ca2+ signaling. Finally, we summarize Ca2+-mediated pharmacological interventions that could potentially lead to novel treatments for pulmonary diseases.
Collapse
Affiliation(s)
- Jeffrey G. Shipman
- Department of Biological and Biomedical Sciences, College of Health and Sciences, North Carolina Central University, Durham, NC 27707, USA;
| | - Rob U. Onyenwoke
- Department of Pharmaceutical Sciences, North Carolina Central University, Durham, NC 27707, USA;
- Biomanufacturing Research Institute and Technology Enterprise (BRITE), North Carolina Central University, Durham, NC 27707, USA
| | - Vijay Sivaraman
- Department of Biological and Biomedical Sciences, College of Health and Sciences, North Carolina Central University, Durham, NC 27707, USA;
| |
Collapse
|
32
|
Lemos AEG, Silva GR, Gimba ERP, Matos ADR. Susceptibility of lung cancer patients to COVID-19: A review of the pandemic data from multiple nationalities. Thorac Cancer 2021; 12:2637-2647. [PMID: 34435733 PMCID: PMC8520793 DOI: 10.1111/1759-7714.14067] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 06/14/2021] [Accepted: 06/18/2021] [Indexed: 01/08/2023] Open
Abstract
Several studies have highlighted that cancer patients tend to be more susceptible to develop severe infection and to die from COVID-19. Certain medical conditions such as immunosuppression, presence of comorbidities, and underlying pulmonary damage are possible determinants of disease severity, especially in lung cancer patients. While recent studies have shown that lung cancer is one of the most prevalent tumor types among COVID-19 cancer patients, we still have an incomplete view of how data from several countries work as a whole. The aim of this review was to investigate COVID-19 prevalence in lung cancer patient cohorts and their probability to develop severe illness and death when compared to nonlung cancer patients from multiple nationalities, including countries that have been the epicenters of the pandemic. We also focus on some intrinsic lung cancer features that might influence COVID-19 outcomes. An integrative view of the susceptibility of lung cancer patients might be especially relevant to assist physicians in evaluating the risks of COVID-19 in these patients, and to foster better decisions on treatment delay.
Collapse
Affiliation(s)
- Ana Emília Goulart Lemos
- Department of Physiology and PharmacologyBiomedical Institute, Federal Fluminense University (UFF)NiteroiBrazil
- National School of Public Health Sergio Arouca, Department of Epidemiology and Quantitative Methods in HealthOswaldo Cruz Foundation (FIOCRUZ)Rio de JaneiroBrazil
- Cellular and Molecular Oncobiology Program, Research CentreNational Cancer Institute (INCA)Rio de JaneiroBrazil
| | - Gabriela Ribeiro Silva
- Department of Physiology and PharmacologyBiomedical Institute, Federal Fluminense University (UFF)NiteroiBrazil
- Cellular and Molecular Oncobiology Program, Research CentreNational Cancer Institute (INCA)Rio de JaneiroBrazil
| | - Etel Rodrigues Pereira Gimba
- Department of Physiology and PharmacologyBiomedical Institute, Federal Fluminense University (UFF)NiteroiBrazil
- Cellular and Molecular Oncobiology Program, Research CentreNational Cancer Institute (INCA)Rio de JaneiroBrazil
- Institute of Humanities and Health, Department of Natural SciencesFederal Fluminense University (UFF)Rio das OstrasBrazil
| | - Aline da Rocha Matos
- Oswaldo Cruz Institute, Respiratory and Measles Viruses Laboratory/SARS‐CoV‐2 Reference, Laboratory, MoHWorld Health Organization (WHO), FIOCRUZRio de JaneiroBrazil
| |
Collapse
|
33
|
Nikniaz L, Ghojazadeh M, Nateghian H, Nikniaz Z, Farhangi MA, Pourmanaf H. The interaction effect of aerobic exercise and vitamin D supplementation on inflammatory factors, anti-inflammatory proteins, and lung function in male smokers: a randomized controlled trial. BMC Sports Sci Med Rehabil 2021; 13:102. [PMID: 34461991 PMCID: PMC8406718 DOI: 10.1186/s13102-021-00333-w] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Accepted: 08/20/2021] [Indexed: 01/26/2023]
Abstract
Background This study aimed to investigate the interaction effect of aerobic exercise and vitamin D supplementation on inflammation (TNF-α, IL-6, CC16, SP-D, and CC16/SP-D ratio) and lung function (FEV1, FVC, and FEV1/FVC ratio) in male smokers.
Methods After applying inclusion criteria, a total of 40 healthy male smokers were recruited in this study. The participants were randomly divided into four groups as follows: Aerobic Exercise + vitamin D Supplementation (AE + VitD, n = 10), Aerobic Exercise (AE, n = 10), vitamin D Supplementation (VitD, n = 10), and Control (C, n = 10). The participants in the AE + VitD and AE groups performed aerobic exercise training (running) up to 50% of the maximum heart rate, three times a week for four weeks. Participants in AE + VitD and VitD groups received 6000 IU/w vitamin D3 for four weeks. The participants in control group did not receive any intervention. Serum tumor necrosis factor (TNF)-α, interleukin (IL)-6, Clara cell protein (CC16), surfactant protein (SP)-D, CC16/SP-D ratio, and lung function (FEV1, FVC, and FEV1/FVC ratio) were measured before and after four weeks of intervention. Results Serum levels of TNF-α, IL-6, and CC16 decreased significantly in AE + VitD, VitD, and AE groups after four weeks (P < 0.05). Serum SP-D level decreased significantly only in the AE + VitD group (P = 0.011). In addition, FEV1 and FVC increased significantly (P < 0.05) in AE + VitD and AE groups after four weeks of intervention. However, the interventions did not have a significant effect on CC16/SP-D ratio and FEV1/FVC ratio (P > 0.05). Furthermore, serum levels of 1,25-dihydroxyvitamin D increased significantly in AE + VitD and VitD groups (P < 0.05) after four weeks of intervention. However, except for TNF-α, between-group comparisons showed no significant differences in levels of IL-6, CC16, SP-D, CC16/SP-D ratio, FEV1, FVC, FEV1/FVC, and 1,25-dihydroxyvitamin D (P > 0.05). Conclusions The results of present study were that aerobic exercise combined with vitamin D supplementation can reduce serum inflammatory factors and anti-inflammatory proteins and improve lung function after four weeks of intervention. Further trials with larger sample size and longer duration are suggested to confirm these results. Trial registration Retrospectively registered. IRCT20180513039637N4. Registration date: 2020/10/20. URL: https://www.irct.ir/search/result?query=IRCT20180513039637N4
Collapse
Affiliation(s)
- Leila Nikniaz
- Tabriz Health Services Management Research Center, Health Management and Safety Promotion Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Morteza Ghojazadeh
- Research Center for Evidence-Based Medicine, Iranian EBM Centre: A Joanna Briggs Institute Affiliated Group, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hooman Nateghian
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Zeinab Nikniaz
- Liver and Gastrointestinal Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Hadi Pourmanaf
- Faculty of Physical Education and Sport Sciences, University of Tehran, Tehran, Iran.
| |
Collapse
|
34
|
Samarelli AV, Tonelli R, Marchioni A, Bruzzi G, Gozzi F, Andrisani D, Castaniere I, Manicardi L, Moretti A, Tabbì L, Cerri S, Beghè B, Dominici M, Clini E. Fibrotic Idiopathic Interstitial Lung Disease: The Molecular and Cellular Key Players. Int J Mol Sci 2021; 22:8952. [PMID: 34445658 PMCID: PMC8396471 DOI: 10.3390/ijms22168952] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 08/05/2021] [Accepted: 08/10/2021] [Indexed: 12/12/2022] Open
Abstract
Interstitial lung diseases (ILDs) that are known as diffuse parenchymal lung diseases (DPLDs) lead to the damage of alveolar epithelium and lung parenchyma, culminating in inflammation and widespread fibrosis. ILDs that account for more than 200 different pathologies can be divided into two groups: ILDs that have a known cause and those where the cause is unknown, classified as idiopathic interstitial pneumonia (IIP). IIPs include idiopathic pulmonary fibrosis (IPF), non-specific interstitial pneumonia (NSIP), cryptogenic organizing pneumonia (COP) known also as bronchiolitis obliterans organizing pneumonia (BOOP), acute interstitial pneumonia (AIP), desquamative interstitial pneumonia (DIP), respiratory bronchiolitis-associated interstitial lung disease (RB-ILD), and lymphocytic interstitial pneumonia (LIP). In this review, our aim is to describe the pathogenic mechanisms that lead to the onset and progression of the different IIPs, starting from IPF as the most studied, in order to find both the common and standalone molecular and cellular key players among them. Finally, a deeper molecular and cellular characterization of different interstitial lung diseases without a known cause would contribute to giving a more accurate diagnosis to the patients, which would translate to a more effective treatment decision.
Collapse
Affiliation(s)
- Anna Valeria Samarelli
- Laboratory of Cell Therapies and Respiratory Medicine, Department of Medical and Surgical Sciences for Children & Adults, University Hospital of Modena and Reggio Emilia, 41100 Modena, Italy; (A.V.S.); (R.T.); (A.M.); (G.B.); (F.G.); (D.A.); (I.C.); (L.M.); (A.M.); (S.C.); (B.B.); (M.D.)
- Respiratory Diseases Unit, Department of Medical and Surgical Sciences, University Hospital of Modena, University of Modena Reggio Emilia, 41100 Modena, Italy;
| | - Roberto Tonelli
- Laboratory of Cell Therapies and Respiratory Medicine, Department of Medical and Surgical Sciences for Children & Adults, University Hospital of Modena and Reggio Emilia, 41100 Modena, Italy; (A.V.S.); (R.T.); (A.M.); (G.B.); (F.G.); (D.A.); (I.C.); (L.M.); (A.M.); (S.C.); (B.B.); (M.D.)
- Respiratory Diseases Unit, Department of Medical and Surgical Sciences, University Hospital of Modena, University of Modena Reggio Emilia, 41100 Modena, Italy;
- Clinical and Experimental Medicine PhD Program, University of Modena Reggio Emilia, 41100 Modena, Italy
| | - Alessandro Marchioni
- Laboratory of Cell Therapies and Respiratory Medicine, Department of Medical and Surgical Sciences for Children & Adults, University Hospital of Modena and Reggio Emilia, 41100 Modena, Italy; (A.V.S.); (R.T.); (A.M.); (G.B.); (F.G.); (D.A.); (I.C.); (L.M.); (A.M.); (S.C.); (B.B.); (M.D.)
- Respiratory Diseases Unit, Department of Medical and Surgical Sciences, University Hospital of Modena, University of Modena Reggio Emilia, 41100 Modena, Italy;
| | - Giulia Bruzzi
- Laboratory of Cell Therapies and Respiratory Medicine, Department of Medical and Surgical Sciences for Children & Adults, University Hospital of Modena and Reggio Emilia, 41100 Modena, Italy; (A.V.S.); (R.T.); (A.M.); (G.B.); (F.G.); (D.A.); (I.C.); (L.M.); (A.M.); (S.C.); (B.B.); (M.D.)
- Respiratory Diseases Unit, Department of Medical and Surgical Sciences, University Hospital of Modena, University of Modena Reggio Emilia, 41100 Modena, Italy;
| | - Filippo Gozzi
- Laboratory of Cell Therapies and Respiratory Medicine, Department of Medical and Surgical Sciences for Children & Adults, University Hospital of Modena and Reggio Emilia, 41100 Modena, Italy; (A.V.S.); (R.T.); (A.M.); (G.B.); (F.G.); (D.A.); (I.C.); (L.M.); (A.M.); (S.C.); (B.B.); (M.D.)
- Respiratory Diseases Unit, Department of Medical and Surgical Sciences, University Hospital of Modena, University of Modena Reggio Emilia, 41100 Modena, Italy;
- Clinical and Experimental Medicine PhD Program, University of Modena Reggio Emilia, 41100 Modena, Italy
| | - Dario Andrisani
- Laboratory of Cell Therapies and Respiratory Medicine, Department of Medical and Surgical Sciences for Children & Adults, University Hospital of Modena and Reggio Emilia, 41100 Modena, Italy; (A.V.S.); (R.T.); (A.M.); (G.B.); (F.G.); (D.A.); (I.C.); (L.M.); (A.M.); (S.C.); (B.B.); (M.D.)
- Respiratory Diseases Unit, Department of Medical and Surgical Sciences, University Hospital of Modena, University of Modena Reggio Emilia, 41100 Modena, Italy;
- Clinical and Experimental Medicine PhD Program, University of Modena Reggio Emilia, 41100 Modena, Italy
| | - Ivana Castaniere
- Laboratory of Cell Therapies and Respiratory Medicine, Department of Medical and Surgical Sciences for Children & Adults, University Hospital of Modena and Reggio Emilia, 41100 Modena, Italy; (A.V.S.); (R.T.); (A.M.); (G.B.); (F.G.); (D.A.); (I.C.); (L.M.); (A.M.); (S.C.); (B.B.); (M.D.)
- Respiratory Diseases Unit, Department of Medical and Surgical Sciences, University Hospital of Modena, University of Modena Reggio Emilia, 41100 Modena, Italy;
- Clinical and Experimental Medicine PhD Program, University of Modena Reggio Emilia, 41100 Modena, Italy
| | - Linda Manicardi
- Laboratory of Cell Therapies and Respiratory Medicine, Department of Medical and Surgical Sciences for Children & Adults, University Hospital of Modena and Reggio Emilia, 41100 Modena, Italy; (A.V.S.); (R.T.); (A.M.); (G.B.); (F.G.); (D.A.); (I.C.); (L.M.); (A.M.); (S.C.); (B.B.); (M.D.)
- Respiratory Diseases Unit, Department of Medical and Surgical Sciences, University Hospital of Modena, University of Modena Reggio Emilia, 41100 Modena, Italy;
| | - Antonio Moretti
- Laboratory of Cell Therapies and Respiratory Medicine, Department of Medical and Surgical Sciences for Children & Adults, University Hospital of Modena and Reggio Emilia, 41100 Modena, Italy; (A.V.S.); (R.T.); (A.M.); (G.B.); (F.G.); (D.A.); (I.C.); (L.M.); (A.M.); (S.C.); (B.B.); (M.D.)
- Respiratory Diseases Unit, Department of Medical and Surgical Sciences, University Hospital of Modena, University of Modena Reggio Emilia, 41100 Modena, Italy;
| | - Luca Tabbì
- Respiratory Diseases Unit, Department of Medical and Surgical Sciences, University Hospital of Modena, University of Modena Reggio Emilia, 41100 Modena, Italy;
| | - Stefania Cerri
- Laboratory of Cell Therapies and Respiratory Medicine, Department of Medical and Surgical Sciences for Children & Adults, University Hospital of Modena and Reggio Emilia, 41100 Modena, Italy; (A.V.S.); (R.T.); (A.M.); (G.B.); (F.G.); (D.A.); (I.C.); (L.M.); (A.M.); (S.C.); (B.B.); (M.D.)
- Respiratory Diseases Unit, Department of Medical and Surgical Sciences, University Hospital of Modena, University of Modena Reggio Emilia, 41100 Modena, Italy;
| | - Bianca Beghè
- Laboratory of Cell Therapies and Respiratory Medicine, Department of Medical and Surgical Sciences for Children & Adults, University Hospital of Modena and Reggio Emilia, 41100 Modena, Italy; (A.V.S.); (R.T.); (A.M.); (G.B.); (F.G.); (D.A.); (I.C.); (L.M.); (A.M.); (S.C.); (B.B.); (M.D.)
- Respiratory Diseases Unit, Department of Medical and Surgical Sciences, University Hospital of Modena, University of Modena Reggio Emilia, 41100 Modena, Italy;
| | - Massimo Dominici
- Laboratory of Cell Therapies and Respiratory Medicine, Department of Medical and Surgical Sciences for Children & Adults, University Hospital of Modena and Reggio Emilia, 41100 Modena, Italy; (A.V.S.); (R.T.); (A.M.); (G.B.); (F.G.); (D.A.); (I.C.); (L.M.); (A.M.); (S.C.); (B.B.); (M.D.)
- Oncology Unit, University Hospital of Modena, University of Modena and Reggio Emilia, 41100 Modena, Italy
| | - Enrico Clini
- Laboratory of Cell Therapies and Respiratory Medicine, Department of Medical and Surgical Sciences for Children & Adults, University Hospital of Modena and Reggio Emilia, 41100 Modena, Italy; (A.V.S.); (R.T.); (A.M.); (G.B.); (F.G.); (D.A.); (I.C.); (L.M.); (A.M.); (S.C.); (B.B.); (M.D.)
- Respiratory Diseases Unit, Department of Medical and Surgical Sciences, University Hospital of Modena, University of Modena Reggio Emilia, 41100 Modena, Italy;
| |
Collapse
|
35
|
Bhat TA, Kalathil SG, Leigh N, Muthumalage T, Rahman I, Goniewicz ML, Thanavala YM. Acute Effects of Heated Tobacco Product (IQOS) Aerosol Inhalation on Lung Tissue Damage and Inflammatory Changes in the Lungs. Nicotine Tob Res 2021; 23:1160-1167. [PMID: 33346355 PMCID: PMC8186425 DOI: 10.1093/ntr/ntaa267] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Accepted: 12/15/2020] [Indexed: 12/19/2022]
Abstract
INTRODUCTION Emerging heated tobacco products (HTPs) were designed to reduce exposure to toxicants from cigarette smoke (CS) by avoiding burning tobacco and instead heating tobacco. We studied the effects of short-term inhalation of aerosols emitted from HTP called IQOS, on lung damage and immune-cell recruitment to the lungs in mice. METHODS Numerous markers of lung damage and inflammation including albumin and lung immune-cell infiltrates, proinflammatory cytokines, and chemokines were quantified in lungs and bronchoalveolar (BAL) fluid from IQOS, CS, or air-exposed (negative control) mice. RESULTS Importantly, as a surrogate marker of lung epithelial-cell damage, we detected significantly increased levels of albumin in the BAL fluid of both HTP- and CS-exposed mice compared with negative controls. Total numbers of leukocytes infiltrating the lungs were equivalent following both IQOS aerosols and CS inhalation and significantly increased compared with air-exposed controls. We also observed significantly increased numbers of CD4+IL-17A+ T cells, a marker of a T-cell immune response, in both groups compared with air controls; however, numbers were the highest following CS exposure. Finally, the numbers of CD4+RORγt+ T cells, an inflammatory T-cell subtype expressing the transcription factor that is essential for promoting differentiation into proinflammatory Th17 cells, were significantly augmented in both groups compared with air-exposed controls. Levels of several cytokines in BAL were significantly elevated, reflecting a proinflammatory milieu. CONCLUSIONS Our study demonstrates that short-term inhalation of aerosols from IQOS generates damage and proinflammatory changes in the lung that are substantially similar to that elicited by CS exposure. IMPLICATIONS Exposure of mice to IQOS, one of the candidate modified-risk tobacco products, induces inflammatory immune-cell accumulation in the lungs and augments the levels of proinflammatory cytokines and chemokines in the BAL fluid. Such an exacerbated pulmonary proinflammatory microenvironment is associated with lung epithelial-cell damage in IQOS-exposed mice, suggesting a potential association with the impairment of lung function.
Collapse
Affiliation(s)
- Tariq A Bhat
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY
| | - Suresh G Kalathil
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY
| | - Noel Leigh
- Department of Health Behavior, Roswell Park Comprehensive Cancer Center, Buffalo, NY
| | - Thivanka Muthumalage
- Department of Environmental Medicine, University of Rochester Medical Center, School of Medicine and Dentistry, Rochester, NY
| | - Irfan Rahman
- Department of Environmental Medicine, University of Rochester Medical Center, School of Medicine and Dentistry, Rochester, NY
| | - Maciej L Goniewicz
- Department of Health Behavior, Roswell Park Comprehensive Cancer Center, Buffalo, NY
| | - Yasmin M Thanavala
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY
| |
Collapse
|
36
|
Abstract
Cigarette smoke (CS) is likely the most common preventable cause of human morbidity and mortality worldwide. Consequently, inexpensive interventional strategies for preventing CS-related diseases would positively impact health systems. Inhaled CS is a powerful inflammatory stimulus and produces a shift in the normal balance between antioxidants and oxidants, inducing oxidative stress in both the respiratory system and throughout the body. This enduring and systemic pro-oxidative state within the body is reflected by increased levels of oxidative stress and inflammation biomarkers seen in smokers. Smokers might benefit from consuming antioxidant supplements, or a diet rich in fruit and vegetables, which can reduce the CS-related oxidative stress. This review provides an overview of the plasma profile of antioxidants observable in smokers and examines the heterogeneous literature to elucidate and discuss the effectiveness of interventional strategies based on antioxidant supplements or an antioxidant-rich diet to improve the health of smokers. An antioxidant-rich diet can provide an easy-to-implement and cost-effective preventative strategy to reduce the risk of CS-related diseases, thus being one of the simplest ways for smokers to stay in good health for as long as possible. The health benefits attributable to the intake of antioxidants have been observed predominantly when these have been consumed within their natural food matrices in an optimal antioxidant-rich diet, while these preventive effects are rarely achieved with the intake of individual antioxidants, even at high doses.
Collapse
|
37
|
Suryadinata RV, Wirjatmadi B, Lorensia A. The time pattern of selenomethionine administration in preventing free radicals due to exposure to electric cigarette smoke. J Public Health Res 2021; 10. [PMID: 33855416 PMCID: PMC8129744 DOI: 10.4081/jphr.2021.2232] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Accepted: 03/29/2021] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND Most people believe that electric cigarettes have no negative effect on health, which causes them to use it more. However, exposure to the smoke from these cigarettes is bad for the health and causes cell damage. Antioxidants play an important role in preventing cell damage, and they can be obtained through the oral administration of selenomethionine. DESIGN AND METHODS This study used an experimental method and a post-test control group design. Male Wistar rats, which were exposed to cigarette smoke were given selenomethionine orally and then tested for the presence of free radicals. The measurement of lung tissue damage was conducted by assessing the level of malondialdehyde in the blood and immunohistochemistry (IHC) of the lung tissue. RESULT The study showed that differences in the time of administration of selenomethionine affect the levels of malondialdehyde in the blood and expression of malondialdehyde in the lung tissue (p<0.05). Consequently, the two groups showed a strong (r=0.861) and significant (p=0.000) relationship with each other. CONCLUSION The early application of selenomethionine can prevent increased levels of malondialdehyde in the blood and lung tissue due to exposure to e-cigarette smoke.
Collapse
Affiliation(s)
| | | | - Amelia Lorensia
- Faculty of Pharmacy, Universitas Surabaya (UBAYA), Surabaya.
| |
Collapse
|
38
|
Peprah E, Armstrong-Hough M, Cook SH, Mukasa B, Taylor JY, Xu H, Chang L, Gyamfi J, Ryan N, Ojo T, Snyder A, Iwelunmor J, Ezechi O, Iyegbe C, O’Reilly P, Pascal Kengne A. An Emerging Syndemic of Smoking and Cardiopulmonary Diseases in People Living with HIV in Africa. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:3111. [PMID: 33803504 PMCID: PMC8003038 DOI: 10.3390/ijerph18063111] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 03/11/2021] [Accepted: 03/14/2021] [Indexed: 11/29/2022]
Abstract
BACKGROUND African countries have the highest number of people living with HIV (PWH). The continent is home to 12% of the global population, but accounts for 71% of PWH globally. Antiretroviral therapy has played an important role in the reduction of the morbidity and mortality rates for HIV, which necessitates increased surveillance of the threats from pernicious risks to which PWH who live longer remain exposed. This includes cardiopulmonary comorbidities, which pose significant public health and economic challenges. A significant contributor to the cardiopulmonary comorbidities is tobacco smoking. Indeed, globally, PWH have a 2-4-fold higher utilization of tobacco compared to the general population, leading to endothelial dysfunction and atherogenesis that result in cardiopulmonary diseases, such as chronic obstructive pulmonary disease and coronary artery disease. In the context of PWH, we discuss (1) the current trends in cigarette smoking and (2) the lack of geographically relevant data on the cardiopulmonary conditions associated with smoking; we then review (3) the current evidence on chronic inflammation induced by smoking and the potential pathways for cardiopulmonary disease and (4) the multifactorial nature of the syndemic of smoking, HIV, and cardiopulmonary diseases. This commentary calls for a major, multi-setting cohort study using a syndemics framework to assess cardiopulmonary disease outcomes among PWH who smoke. CONCLUSION We call for a parallel program of implementation research to promote the adoption of evidence-based interventions, which could improve health outcomes for PWH with cardiopulmonary diseases and address the health inequities experienced by PWH in African countries.
Collapse
Affiliation(s)
- Emmanuel Peprah
- School of Global Public Health, New York University, New York, NY 10012, USA; (M.A.-H.); (S.H.C.); (J.G.); (N.R.); (T.O.); (A.S.)
| | - Mari Armstrong-Hough
- School of Global Public Health, New York University, New York, NY 10012, USA; (M.A.-H.); (S.H.C.); (J.G.); (N.R.); (T.O.); (A.S.)
| | - Stephanie H. Cook
- School of Global Public Health, New York University, New York, NY 10012, USA; (M.A.-H.); (S.H.C.); (J.G.); (N.R.); (T.O.); (A.S.)
| | | | | | - Huichun Xu
- Division of Endocrinology, Diabetes and Nutrition, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA;
| | - Linda Chang
- Department of Neurology, University of Maryland School of Medicine, Baltimore, MD 21201, USA;
| | - Joyce Gyamfi
- School of Global Public Health, New York University, New York, NY 10012, USA; (M.A.-H.); (S.H.C.); (J.G.); (N.R.); (T.O.); (A.S.)
| | - Nessa Ryan
- School of Global Public Health, New York University, New York, NY 10012, USA; (M.A.-H.); (S.H.C.); (J.G.); (N.R.); (T.O.); (A.S.)
| | - Temitope Ojo
- School of Global Public Health, New York University, New York, NY 10012, USA; (M.A.-H.); (S.H.C.); (J.G.); (N.R.); (T.O.); (A.S.)
| | - Anya Snyder
- School of Global Public Health, New York University, New York, NY 10012, USA; (M.A.-H.); (S.H.C.); (J.G.); (N.R.); (T.O.); (A.S.)
| | - Juliet Iwelunmor
- College for Public Health and Social Justice, Saint Louis University, St. Louis, MO 63103, USA;
| | - Oliver Ezechi
- Nigerian Institute of Medical Research, Lagos, Nigeria;
| | - Conrad Iyegbe
- Icahn School of Medicine at Mount Sinai, Mount Sinai Hospital, New York, NY 10029, USA; (C.I.); (P.O.); (A.P.K.)
| | - Paul O’Reilly
- Icahn School of Medicine at Mount Sinai, Mount Sinai Hospital, New York, NY 10029, USA; (C.I.); (P.O.); (A.P.K.)
| | - Andre Pascal Kengne
- Icahn School of Medicine at Mount Sinai, Mount Sinai Hospital, New York, NY 10029, USA; (C.I.); (P.O.); (A.P.K.)
- South African Medical Research Council, Cape Town, South Africa
| |
Collapse
|
39
|
Orlandi RR, Kingdom TT, Smith TL, Bleier B, DeConde A, Luong AU, Poetker DM, Soler Z, Welch KC, Wise SK, Adappa N, Alt JA, Anselmo-Lima WT, Bachert C, Baroody FM, Batra PS, Bernal-Sprekelsen M, Beswick D, Bhattacharyya N, Chandra RK, Chang EH, Chiu A, Chowdhury N, Citardi MJ, Cohen NA, Conley DB, DelGaudio J, Desrosiers M, Douglas R, Eloy JA, Fokkens WJ, Gray ST, Gudis DA, Hamilos DL, Han JK, Harvey R, Hellings P, Holbrook EH, Hopkins C, Hwang P, Javer AR, Jiang RS, Kennedy D, Kern R, Laidlaw T, Lal D, Lane A, Lee HM, Lee JT, Levy JM, Lin SY, Lund V, McMains KC, Metson R, Mullol J, Naclerio R, Oakley G, Otori N, Palmer JN, Parikh SR, Passali D, Patel Z, Peters A, Philpott C, Psaltis AJ, Ramakrishnan VR, Ramanathan M, Roh HJ, Rudmik L, Sacks R, Schlosser RJ, Sedaghat AR, Senior BA, Sindwani R, Smith K, Snidvongs K, Stewart M, Suh JD, Tan BK, Turner JH, van Drunen CM, Voegels R, Wang DY, Woodworth BA, Wormald PJ, Wright ED, Yan C, Zhang L, Zhou B. International consensus statement on allergy and rhinology: rhinosinusitis 2021. Int Forum Allergy Rhinol 2021; 11:213-739. [PMID: 33236525 DOI: 10.1002/alr.22741] [Citation(s) in RCA: 490] [Impact Index Per Article: 122.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Accepted: 11/09/2020] [Indexed: 02/06/2023]
Abstract
I. EXECUTIVE SUMMARY BACKGROUND: The 5 years since the publication of the first International Consensus Statement on Allergy and Rhinology: Rhinosinusitis (ICAR-RS) has witnessed foundational progress in our understanding and treatment of rhinologic disease. These advances are reflected within the more than 40 new topics covered within the ICAR-RS-2021 as well as updates to the original 140 topics. This executive summary consolidates the evidence-based findings of the document. METHODS ICAR-RS presents over 180 topics in the forms of evidence-based reviews with recommendations (EBRRs), evidence-based reviews, and literature reviews. The highest grade structured recommendations of the EBRR sections are summarized in this executive summary. RESULTS ICAR-RS-2021 covers 22 topics regarding the medical management of RS, which are grade A/B and are presented in the executive summary. Additionally, 4 topics regarding the surgical management of RS are grade A/B and are presented in the executive summary. Finally, a comprehensive evidence-based management algorithm is provided. CONCLUSION This ICAR-RS-2021 executive summary provides a compilation of the evidence-based recommendations for medical and surgical treatment of the most common forms of RS.
Collapse
Affiliation(s)
| | | | | | | | | | - Amber U Luong
- University of Texas Medical School at Houston, Houston, TX
| | | | - Zachary Soler
- Medical University of South Carolina, Charleston, SC
| | - Kevin C Welch
- Feinberg School of Medicine, Northwestern University, Chicago, IL
| | | | | | | | | | - Claus Bachert
- Ghent University, Ghent, Belgium.,Karolinska Institute, Stockholm, Sweden.,Sun Yatsen University, Gangzhou, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - David A Gudis
- Columbia University Irving Medical Center, New York, NY
| | - Daniel L Hamilos
- Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | | | - Richard Harvey
- University of New South Wales and Macquarie University, Sydney, New South Wales, Australia
| | | | | | | | | | - Amin R Javer
- University of British Columbia, Vancouver, British Columbia, Canada
| | | | | | | | | | | | | | | | | | | | | | - Valerie Lund
- Royal National Throat Nose and Ear Hospital, UCLH, London, UK
| | - Kevin C McMains
- Uniformed Services University of Health Sciences, San Antonio, TX
| | | | - Joaquim Mullol
- IDIBAPS Hospital Clinic, University of Barcelona, Barcelona, Spain
| | | | | | | | | | | | | | | | | | | | - Alkis J Psaltis
- University of Adelaide, Adelaide, South Australia, Australia
| | | | | | | | - Luke Rudmik
- University of Calgary, Calgary, Alberta, Canada
| | - Raymond Sacks
- University of New South Wales, Sydney, New South Wales, Australia
| | | | | | | | | | | | | | | | | | | | | | | | | | - De Yun Wang
- National University of Singapore, Singapore, Singapore
| | | | | | | | - Carol Yan
- University of California San Diego, La Jolla, CA
| | - Luo Zhang
- Capital Medical University, Beijing, China
| | - Bing Zhou
- Capital Medical University, Beijing, China
| |
Collapse
|
40
|
Sokar SS, Afify EH, Osman EY. Dexamethasone and losartan combination treatment protected cigarette smoke-induced COPD in rats. Hum Exp Toxicol 2021; 40:284-296. [PMID: 32812458 DOI: 10.1177/0960327120950012] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Chronic Obstructive Pulmonary Disease (COPD) is a dangerous prevalent smoking-related disease characterized by abnormal inflammation and oxidative stress and expected to be the third cause of death in the world next decade. Corticosteroids have low effects in decreasing numbers of inflammatory mediators specifically in long-term use. Our study designed to investigate the possible protective effects of combined dexamethasone (Dex) (2mg/kg) and losartan (Los) (30mg/kg angiotensin receptor blocker, it possesses antioxidant and anti-inflammatory properties in lung injury in mice) against cigarette -smoke (CS) induced COPD in rats compared with dexamethasone and losartan. Male Sprague Dawley rats (N = 40) divided into five groups (n = 8): control group, CS group, Dex group, Los group, and Dex +Los group. COPD induced in rats by CS exposure twice daily for 10 weeks. After the specified treatment period, bronchoalveolar lavage fluid (BALF) and lung tissue were collected for measurement of SOD, NO, MDA, ICAM-, MMP-9, CRP, NF-κB and histopathology scoring. Our results indicated that Los+Dex significantly prevent CS-induced COPD emphysema, congested alveoli, and elevation of lung injury parameters in BALF. They also showed a significant decrease in MDA, ICAM-1, MMP-9, CRP, and NF-κB and a significant increase in SOD and NO. In conclusion, adding Los to Dex potentiating their activity in inhibition the progression of COPD based on its activity on oxidative stress, inflammation, and NF-κB protein expression.
Collapse
Affiliation(s)
- Samia S Sokar
- Professor of Pharmacology and Toxicology, 68904Faculty of Pharmacy, Tanta University, Egypt
| | | | - Enass Y Osman
- Department of Pharmacology and Toxicology, 68904Faculty of Pharmacy, Tanta University, Egypt
| |
Collapse
|
41
|
Wang L, Sun Y, Yuan Y, Mei Q, Yuan X. Clinical challenges in cancer patients with COVID-19: Aging, immunosuppression, and comorbidities. Aging (Albany NY) 2020; 12:24462-24474. [PMID: 33232275 PMCID: PMC7762454 DOI: 10.18632/aging.104205] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Accepted: 09/06/2020] [Indexed: 01/08/2023]
Abstract
COVID-19 caused by severe acute respiratory syndrome coronavirus 2 has developed into a global pandemic. COVID-19 poses a huge threat to health care, and the shortage of medical resources caused by COVID-19 brought serious secondary disasters to elderly cancer patients who are particularly dependent on medical resources. The clinical challenges of cancer management, including aging, immunosuppression, and comorbidities, make cancer patients more vulnerable to COVID-19 with different clinical manifestations, disease severity, and outcomes. The review comprehensively analyzed the characteristics of the cancer patients under the pandemic and concluded that cancer patients were more susceptible to COVID-19, and also concluded that they were more likely to develop poor outcomes and the severe form of the disease. Three basic management strategies have been proposed to protect susceptible elderly cancer patients, find reliable indicators to monitor the course of disease, and implement effective prevention measures.
Collapse
Affiliation(s)
- Lu Wang
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yinan Sun
- Department of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Ye Yuan
- Department of Gastroenterology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Qi Mei
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xianglin Yuan
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| |
Collapse
|
42
|
Jiang Y, Liao H, Zhang X, Cao S, Hu X, Yang Z, Fang Y, Wang H. IL-33 synergistically promotes the proliferation of lung cancer cells in vitro by inducing antibacterial peptide LL-37 and proinflammatory cytokines in macrophages. Immunobiology 2020; 225:152025. [PMID: 33190003 DOI: 10.1016/j.imbio.2020.152025] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 08/30/2020] [Accepted: 10/18/2020] [Indexed: 02/07/2023]
Abstract
Lung cancer is the primary cause of cancer-related deaths, and the persistent inflammation is inextricably linked with the lung cancer tumorigenesis. Pro-inflammatory cytokine interleukin-33 (IL-33) is able to serve as a potent modulator of cancer. Mounting evidence indicates IL-33 has significant effect on lung cancer progression by regulating host immune response, but the current opinions about the function and mechanism of IL-33 in lung cancer are still controversial. Meanwhile, antibacterial peptide LL-37 also exerts a momentous effect on immune responses to lung cancer. LL-37 is regarded as versatile, including antimicrobial activities, chemotaxis and immunoregulation. However, the immunomodulatory mechanism of IL-33 and LL-37 in lung cancer remains thoroughly not defined. Here, we determined the secretion of LL-37 was up-regulated in lung cancer serum samples. Similarly, the expression of CRAMP was enhancive in macrophages after co-cultured with lung cancer cells. Moreover, we expounded that IL-33 could up-regulate LL-37 secretion in macrophages, resulting in the massive releases of IL-6 and IL-1β. Additionally, LL-37 cooperated with IL-33 to increase the phosphorylation of p38 MAPK and NF-κB p65 pathways, and augmented IL-6 and IL-1β secretion, which resulting in the proliferation of lung cancer cells in vitro. In conclusion, our study identified that IL-33 aggravated the inflammation of lung cancer by increasing LL-37 expression in macrophages, thereby promoting lung cancer cell proliferation in vitro. It is contributed to our present understanding of the immunomodulatory relationship between pro-inflammatory cytokines and antibacterial peptides in the tumor immune response, and offer a novel perspective for controlling the progress of lung cancer.
Collapse
Affiliation(s)
- Yinting Jiang
- Key Laboratory of Diagnostic Medicine Designated by the Ministry of Education, Chongqing Medical University, Chongqing 400016, China; School of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Hongyi Liao
- Key Laboratory of Diagnostic Medicine Designated by the Ministry of Education, Chongqing Medical University, Chongqing 400016, China; Department of Clinical Laboratory Medicine, Yongchuan Hospital, Chongqing Medical University, Chongqing, China
| | - Xuemei Zhang
- Key Laboratory of Diagnostic Medicine Designated by the Ministry of Education, Chongqing Medical University, Chongqing 400016, China; School of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Sijia Cao
- Key Laboratory of Diagnostic Medicine Designated by the Ministry of Education, Chongqing Medical University, Chongqing 400016, China; School of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Xuexue Hu
- Key Laboratory of Diagnostic Medicine Designated by the Ministry of Education, Chongqing Medical University, Chongqing 400016, China; School of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Zihan Yang
- Key Laboratory of Diagnostic Medicine Designated by the Ministry of Education, Chongqing Medical University, Chongqing 400016, China; School of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Yuting Fang
- Key Laboratory of Diagnostic Medicine Designated by the Ministry of Education, Chongqing Medical University, Chongqing 400016, China; School of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Hong Wang
- Key Laboratory of Diagnostic Medicine Designated by the Ministry of Education, Chongqing Medical University, Chongqing 400016, China; School of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China.
| |
Collapse
|
43
|
Tsai M, Byun MK, Shin J, Crotty Alexander LE. Effects of e-cigarettes and vaping devices on cardiac and pulmonary physiology. J Physiol 2020; 598:5039-5062. [PMID: 32975834 PMCID: PMC10506663 DOI: 10.1113/jp279754] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Accepted: 08/21/2020] [Indexed: 12/11/2022] Open
Abstract
E-cigarette aerosols are exceedingly different from conventional tobacco smoke, containing dozens of chemicals not found in cigarette smoke. It is highly likely that chronic use of e-cigarettes will induce pathological changes in both the heart and lungs. Here we review human and animal studies published to date and summarize the cardiopulmonary physiological changes caused by vaping. In terms of cardiac physiology, acute exposure to e-cigarette aerosols in human subjects led to increased blood pressure and heart rate, similar to traditional cigarettes. Chronic exposure to e-cigarette aerosols using animal models caused increased arterial stiffness, vascular endothelial changes, increased angiogenesis, cardiorenal fibrosis and increased atherosclerotic plaque formation. Pulmonary physiology is also affected by e-cigarette aerosol inhalation, with increased airway reactivity, airway obstruction, inflammation and emphysema. Research thus far demonstrates that the heart and lung undergo numerous changes in response to e-cigarette use, and disease development will depend on how those changes combine with both environmental and genetic factors. E-cigarettes have been advertised as a healthy alternative to cigarette smoking, and users are under the impression that vaping of e-cigarettes is harmless, but these claims that e-cigarettes are safer and healthier are not based on evidence. Data from both humans and animal models are consistent in demonstrating that vaping of e-cigarettes causes health effects both similar to and disparate from those of cigarette smoking. Further work is needed to define the long-term cardiopulmonary effects of e-cigarette use in humans.
Collapse
Affiliation(s)
- MuChun Tsai
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, The Ohio State University, Columbus, OH, USA
| | - Min Kwang Byun
- Pulmonary and Critical Care Section, VA San Diego Healthcare System, La Jolla, CA, USA
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of California San Diego (UCSD), La Jolla, CA, USA
- Division of Pulmonology, Department of Internal Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, South Korea
| | - John Shin
- Pulmonary and Critical Care Section, VA San Diego Healthcare System, La Jolla, CA, USA
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of California San Diego (UCSD), La Jolla, CA, USA
| | - Laura E Crotty Alexander
- Pulmonary and Critical Care Section, VA San Diego Healthcare System, La Jolla, CA, USA
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of California San Diego (UCSD), La Jolla, CA, USA
| |
Collapse
|
44
|
Brózman O, Novák J, Bauer AK, Babica P. Airborne PAHs inhibit gap junctional intercellular communication and activate MAPKs in human bronchial epithelial cell line. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2020; 79:103422. [PMID: 32492535 PMCID: PMC7486243 DOI: 10.1016/j.etap.2020.103422] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 05/08/2020] [Accepted: 05/20/2020] [Indexed: 06/11/2023]
Abstract
Inhalation exposures to polycyclic aromatic hydrocarbons (PAHs) have been associated with various adverse health effects, including chronic lung diseases and cancer. Using human bronchial epithelial cell line HBE1, we investigated the effects of structurally different PAHs on tissue homeostatic processes, namely gap junctional intercellular communication (GJIC) and MAPKs activity. Rapid (<1 h) and sustained (up to 24 h) inhibition of GJIC was induced by low/middle molecular weight (MW) PAHs, particularly by those with a bay- or bay-like region (1- and 9-methylanthracene, fluoranthene), but also by fluorene and pyrene. In contrast, linear low MW (anthracene, 2-methylanthracene) or higher MW (chrysene) PAHs did not affect GJIC. Fluoranthene, 1- and 9-methylanthracene induced strong and sustained activation of MAPK ERK1/2, whereas MAPK p38 was activated rather nonspecifically by all tested PAHs. Low/middle MW PAHs can disrupt tissue homeostasis in human airway epithelium via structure-dependent nongenotoxic mechanisms, which can contribute to their human health hazards.
Collapse
Affiliation(s)
- Ondřej Brózman
- RECETOX, Faculty of Science, Masaryk University, Brno 62500, Czech Republic.
| | - Jiří Novák
- RECETOX, Faculty of Science, Masaryk University, Brno 62500, Czech Republic.
| | - Alison K Bauer
- Department of Environmental and Occupational Health, University of Colorado, Anschutz Medical Center, Aurora, Colorado 80045, USA.
| | - Pavel Babica
- RECETOX, Faculty of Science, Masaryk University, Brno 62500, Czech Republic.
| |
Collapse
|
45
|
Casey AM, Muise ED, Crotty Alexander LE. Vaping and e-cigarette use. Mysterious lung manifestations and an epidemic. Curr Opin Immunol 2020; 66:143-150. [PMID: 33186869 PMCID: PMC7755270 DOI: 10.1016/j.coi.2020.10.003] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Accepted: 10/04/2020] [Indexed: 12/13/2022]
Abstract
Electronic (e)-cigarette use and the practice of vaping has rapidly expanded both in adult smokers and previously nicotine naïve youths. Research has focused on harm reduction in adults using e-cigarettes to stop or reduce traditional cigarette use, but the short and long-term safety of these products has not been established. Vaping has more recently been associated with a growing list of pulmonary complications with the most urgent being the e-cigarette or vaping product use-associated lung injury (EVALI) epidemic. This review details the inhalant toxicology of vaping products, the described lung diseases associated with vaping with a focus on EVALI, and the predicted long-term consequences of e-cigarette use, including increased asthma severity.
Collapse
Affiliation(s)
- Alicia M Casey
- Division of Pulmonary Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA 02215, United States
| | - Eleanor D Muise
- Division of Pulmonary Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA 02215, United States
| | - Laura E Crotty Alexander
- Pulmonary Critical Care Section, VA San Diego Healthcare System, La Jolla, CA 92161, United States; Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of California San Diego (UCSD), La Jolla, CA 92093, United States.
| |
Collapse
|
46
|
Lechasseur A, Huppé CA, Talbot M, Routhier J, Aubin S, Beaulieu MJ, Duchaine C, Marsolais D, Morissette MC. Exposure to nicotine-free and flavor-free e-cigarette vapors modifies the pulmonary response to tobacco cigarette smoke in female mice. Am J Physiol Lung Cell Mol Physiol 2020; 319:L717-L727. [PMID: 32845704 DOI: 10.1152/ajplung.00037.2020] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Most of electronic cigarette (e-cigarette) users are also smoking tobacco cigarettes. Because of the relative novelty of this habit, very little is known on the impact of vaping on pulmonary health, even less on the potential interactions of dual e-cigarette and tobacco cigarette use. Therefore, we used well-established mouse models to investigate the impact of dual exposure to e-cigarette vapors and tobacco cigarette smoke on lung homeostasis. Groups of female BALB/c mice were exposed to room air, tobacco smoke only, nicotine-free flavor-free e-cigarette vapors only or both tobacco smoke and e-cigarette vapors. Moreover, since tobacco smoke and electronic cigarette vapors both affect circadian processes in the lungs, groups of mice were euthanized at two different time points during the day. We found that dual-exposed mice had altered lung circadian gene expression compared with mice exposed to tobacco smoke alone. Dual-exposed mice also had different frequencies of dendritic cells, macrophages, and neutrophils in the lung tissue compared with mice exposed to tobacco smoke alone, an observation also valid for B-lymphocytes and CD4+ and CD8+ T lymphocytes. Exposure to e-cigarette vapors also impacted the levels of immunoglobulins in the bronchoalveolar lavage and serum. Finally, e-cigarette and dual exposures increased airway resistance compared with mice exposed to room air or tobacco smoke alone, respectively. Taken together, these data suggest that e-cigarette vapors, even without nicotine or flavors, could affect how the lungs react to tobacco cigarette smoke exposure in dual users, potentially altering the pathological course triggered by smoking.
Collapse
Affiliation(s)
- Ariane Lechasseur
- Quebec Heart and Lung Institute-Université Laval, Quebec, Quebec, Canada.,Faculty of Medicine, Université Laval, Quebec, Quebec, Canada
| | - Carol-Ann Huppé
- Quebec Heart and Lung Institute-Université Laval, Quebec, Quebec, Canada.,Faculty of Medicine, Université Laval, Quebec, Quebec, Canada
| | - Maude Talbot
- Quebec Heart and Lung Institute-Université Laval, Quebec, Quebec, Canada.,Faculty of Medicine, Université Laval, Quebec, Quebec, Canada
| | - Joanie Routhier
- Quebec Heart and Lung Institute-Université Laval, Quebec, Quebec, Canada
| | - Sophie Aubin
- Quebec Heart and Lung Institute-Université Laval, Quebec, Quebec, Canada
| | | | - Caroline Duchaine
- Quebec Heart and Lung Institute-Université Laval, Quebec, Quebec, Canada.,Departement of Biochemistry, Microbiology and Bioinformatics, Université Laval, Quebec, Quebec, Canada
| | - David Marsolais
- Quebec Heart and Lung Institute-Université Laval, Quebec, Quebec, Canada.,Department of Medicine, Université Laval, Quebec, Quebec, Canada
| | - Mathieu C Morissette
- Quebec Heart and Lung Institute-Université Laval, Quebec, Quebec, Canada.,Department of Medicine, Université Laval, Quebec, Quebec, Canada
| |
Collapse
|
47
|
Pinto DP, Coutinho DDS, Carvalho KIMD, Ferrero MR, Silva LVD, Silveira GPE, Silva DMD, Araújo JFG, Silva ACA, Pereira HM, Fonseca LBD, Faria RX, Souza MVND, Silva ETD, Santos-Filho OA, Costa JCSD, Amendoeira FC, Martins MA. Pharmacological profiling of JME-173, a novel mexiletine derivative combining dual anti-inflammatory/anti-spasmodic functions and limited action in Na + channels. Eur J Pharmacol 2020; 885:173367. [PMID: 32750364 DOI: 10.1016/j.ejphar.2020.173367] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 06/30/2020] [Accepted: 07/09/2020] [Indexed: 12/14/2022]
Abstract
Existing evidence suggests that the local anaesthetic mexiletine can be beneficial for patients with asthma. However, caution is required since anaesthesia of the airways inhibits protective bronchodilator neuronal reflexes, limiting applications in conditions of hyperirritable airways. Here, we describe the synthesis of a new series of mexiletine analogues, which were screened for reduced activity in Na+ channels and improved smooth muscle relaxant effects, that were evaluated using the patch-clamp technique and an isolated tracheal organ bath, respectively. JME-173 (1-(4-bromo-3,5-dimethylphenoxy)propan-2-amine) was the most effective among the four mexiletine analogues investigated. JME-173 was then studied in vivo using a murine model of lung inflammation induced by cigarette smoke (CS) and in vitro using neutrophil chemotaxis and mast cell degranulation assays. Finally, the JME-173 pharmacokinetic profile was assessed using HPLC-MS/MS bioanalytical method. JME-173 directly inhibited IL-8 (CXCL8)- and FMLP-induced human neutrophil chemotaxis and allergen-induced mast cell degranulation. After oral administration 1 h before CS exposure, JME-173 (50 mg/kg) strongly reduced the increased number of macrophages and neutrophils recovered in the bronchoalveolar effluent without altering lymphocyte counts. Pharmacokinetic experiments of JME-173 (10 mg/kg, orally) showed values of maximum concentration (Cmax), maximum time (Tmax), area under the blood concentration-time curve (AUC0-t) and area under the blood concentration-time curve from 0-Inf (AUC0-inf) of 163.3 ± 38.3 ng/mL, 1.2 ± 0.3 h, 729.4 ± 118.3 ng*h/ml and 868.9 ± 117.1 ng*h/ml (means ± S.E.M.), respectively. Collectively, these findings suggest that JME-173 has the potential to be an effective oral treatment for diseases associated with bronchoconstriction and inflammation.
Collapse
Affiliation(s)
- Douglas Pereira Pinto
- Laboratory of Pharmacokinetics, Vice Presidency of Research and Innovation in Health - Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | - Diego de Sá Coutinho
- Laboratory of Inflammation, Instituto Oswaldo Cruz - Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | | | - Maximiliano R Ferrero
- Laboratory of Inflammation, Instituto Oswaldo Cruz - Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | - Letícia Vallim da Silva
- Laboratory of Pharmacokinetics, Vice Presidency of Research and Innovation in Health - Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | | | - Diego Medeiros da Silva
- Laboratory of Pharmacokinetics, Vice Presidency of Research and Innovation in Health - Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | - João Felipe Garcia Araújo
- Laboratory of Pharmacokinetics, Vice Presidency of Research and Innovation in Health - Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | - Aline C A Silva
- Laboratory of Pharmacokinetics, Vice Presidency of Research and Innovation in Health - Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | - Heliana Martins Pereira
- Laboratory of Pharmacokinetics, Vice Presidency of Research and Innovation in Health - Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | - Laís Bastos da Fonseca
- Laboratory of Pharmacokinetics, Vice Presidency of Research and Innovation in Health - Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | - Robson Xavier Faria
- Laboratory of Toxoplasmosis and Other Protozoans, Oswaldo Cruz Institute, Brazil
| | - Marcus Vinicius Nora de Souza
- Laboratory of Organic Synthesis, Institute of Technology in Drugs, Farmanguinhos - Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | - Emerson Teixeira da Silva
- Laboratory of Organic Synthesis, Institute of Technology in Drugs, Farmanguinhos - Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | - Osvaldo Andrade Santos-Filho
- Laboratory of Molecular Modeling and Computational Structural Biology, Instituto de Pesquisas de Produtos Naturais - Universidade Federal Do Rio de Janeiro, Rio de Janeiro, Brazil
| | | | - Fábio Coelho Amendoeira
- Department of Pharmacology and Toxicology, National Institute of Quality Control in Health - Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | - Marco Aurélio Martins
- Laboratory of Inflammation, Instituto Oswaldo Cruz - Fundação Oswaldo Cruz, Rio de Janeiro, Brazil.
| |
Collapse
|
48
|
Association Between Inflammatory Pathways and Phenotypes of Pulmonary Dysfunction Using Cluster Analysis in Persons Living With HIV and HIV-Uninfected Individuals. J Acquir Immune Defic Syndr 2020; 83:189-196. [PMID: 31929407 DOI: 10.1097/qai.0000000000002234] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND Persons living with HIV (PLWH) are at risk of developing different phenotypes of chronic lung disease, including chronic obstructive pulmonary disease. Mechanisms underlying these phenotypes are unclear. OBJECTIVE To identify clusters of peripheral inflammatory mediators associated with pulmonary function to determine inflammatory pathways and phenotypes of chronic obstructive pulmonary disease in PLWH and HIV-uninfected individuals. METHODS Study participants were PLWH and HIV-uninfected individuals enrolled in the Pittsburgh HIV Lung Cohort. Pulmonary function tests were performed for all participants. Chest computed tomographic scans were performed in a subset of PLWH. Plasma levels of 19 inflammatory mediators were measured by Luminex or ELISA. Clusters were identified based on the expression pattern of inflammatory mediators in PLWH and HIV-uninfected individuals, and the relationships among clinical parameters were evaluated within clusters by using cluster and network analyses. RESULTS In PLWH, we identified a distinct cluster with higher levels of Th1, Th2, and Th17 inflammatory mediators with increased complexity of these mediators and inferred presence of pathogenic Th17 cell types. Individuals in this cluster had worse airway obstruction and more radiographic emphysema. In HIV-uninfected individuals, a cluster with high-grade systemic inflammation also had worse diffusing capacity for carbon monoxide. CONCLUSIONS Inflammatory pathways associated with pulmonary dysfunction in PLWH suggest multifaceted immune dysregulation involved in different phenotypes of pulmonary dysfunction with a potential specific contribution of the Th17 pathway to airway obstruction in PLWH. Identification of these associations may help in development of treatments that could alter the course of the disease.
Collapse
|
49
|
Kuderer NM, Choueiri TK, Shah DP, Shyr Y, Rubinstein SM, Rivera DR, Shete S, Hsu CY, Desai A, de Lima Lopes G, Grivas P, Painter CA, Peters S, Thompson MA, Bakouny Z, Batist G, Bekaii-Saab T, Bilen MA, Bouganim N, Larroya MB, Castellano D, Del Prete SA, Doroshow DB, Egan PC, Elkrief A, Farmakiotis D, Flora D, Galsky MD, Glover MJ, Griffiths EA, Gulati AP, Gupta S, Hafez N, Halfdanarson TR, Hawley JE, Hsu E, Kasi A, Khaki AR, Lemmon CA, Lewis C, Logan B, Masters T, McKay RR, Mesa RA, Morgans AK, Mulcahy MF, Panagiotou OA, Peddi P, Pennell NA, Reynolds K, Rosen LR, Rosovsky R, Salazar M, Schmidt A, Shah SA, Shaya JA, Steinharter J, Stockerl-Goldstein KE, Subbiah S, Vinh DC, Wehbe FH, Weissmann LB, Wu JTY, Wulff-Burchfield E, Xie Z, Yeh A, Yu PP, Zhou AY, Zubiri L, Mishra S, Lyman GH, Rini BI, Warner JL. Clinical impact of COVID-19 on patients with cancer (CCC19): a cohort study. Lancet 2020; 395:1907-1918. [PMID: 32473681 PMCID: PMC7255743 DOI: 10.1016/s0140-6736(20)31187-9] [Citation(s) in RCA: 1273] [Impact Index Per Article: 254.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 05/07/2020] [Accepted: 05/14/2020] [Indexed: 02/06/2023]
Abstract
BACKGROUND Data on patients with COVID-19 who have cancer are lacking. Here we characterise the outcomes of a cohort of patients with cancer and COVID-19 and identify potential prognostic factors for mortality and severe illness. METHODS In this cohort study, we collected de-identified data on patients with active or previous malignancy, aged 18 years and older, with confirmed severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection from the USA, Canada, and Spain from the COVID-19 and Cancer Consortium (CCC19) database for whom baseline data were added between March 17 and April 16, 2020. We collected data on baseline clinical conditions, medications, cancer diagnosis and treatment, and COVID-19 disease course. The primary endpoint was all-cause mortality within 30 days of diagnosis of COVID-19. We assessed the association between the outcome and potential prognostic variables using logistic regression analyses, partially adjusted for age, sex, smoking status, and obesity. This study is registered with ClinicalTrials.gov, NCT04354701, and is ongoing. FINDINGS Of 1035 records entered into the CCC19 database during the study period, 928 patients met inclusion criteria for our analysis. Median age was 66 years (IQR 57-76), 279 (30%) were aged 75 years or older, and 468 (50%) patients were male. The most prevalent malignancies were breast (191 [21%]) and prostate (152 [16%]). 366 (39%) patients were on active anticancer treatment, and 396 (43%) had active (measurable) cancer. At analysis (May 7, 2020), 121 (13%) patients had died. In logistic regression analysis, independent factors associated with increased 30-day mortality, after partial adjustment, were: increased age (per 10 years; partially adjusted odds ratio 1·84, 95% CI 1·53-2·21), male sex (1·63, 1·07-2·48), smoking status (former smoker vs never smoked: 1·60, 1·03-2·47), number of comorbidities (two vs none: 4·50, 1·33-15·28), Eastern Cooperative Oncology Group performance status of 2 or higher (status of 2 vs 0 or 1: 3·89, 2·11-7·18), active cancer (progressing vs remission: 5·20, 2·77-9·77), and receipt of azithromycin plus hydroxychloroquine (vs treatment with neither: 2·93, 1·79-4·79; confounding by indication cannot be excluded). Compared with residence in the US-Northeast, residence in Canada (0·24, 0·07-0·84) or the US-Midwest (0·50, 0·28-0·90) were associated with decreased 30-day all-cause mortality. Race and ethnicity, obesity status, cancer type, type of anticancer therapy, and recent surgery were not associated with mortality. INTERPRETATION Among patients with cancer and COVID-19, 30-day all-cause mortality was high and associated with general risk factors and risk factors unique to patients with cancer. Longer follow-up is needed to better understand the effect of COVID-19 on outcomes in patients with cancer, including the ability to continue specific cancer treatments. FUNDING American Cancer Society, National Institutes of Health, and Hope Foundation for Cancer Research.
Collapse
Affiliation(s)
| | | | - Dimpy P Shah
- Mays Cancer Center, UT Health San Antonio MD Anderson Cancer Center, San Antonio, TX, USA
| | - Yu Shyr
- Vanderbilt-Ingram Cancer Center at Vanderbilt University Medical Center, Nashville, TN, USA
| | - Samuel M Rubinstein
- Vanderbilt-Ingram Cancer Center at Vanderbilt University Medical Center, Nashville, TN, USA
| | - Donna R Rivera
- Division of Cancer Control and Population Sciences, National Cancer Institute, Rockville, MD, USA
| | | | - Chih-Yuan Hsu
- Vanderbilt-Ingram Cancer Center at Vanderbilt University Medical Center, Nashville, TN, USA
| | | | | | - Petros Grivas
- Fred Hutchinson Cancer Research Center, Seattle, WA, USA; University of Washington, Seattle, WA, USA
| | | | | | | | | | - Gerald Batist
- Segal Cancer Centre, Jewish General Hospital, McGill University, Montreal, QC, Canada
| | | | - Mehmet A Bilen
- Winship Cancer Institute of Emory University, Atlanta, GA, USA
| | | | | | | | | | - Deborah B Doroshow
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Pamela C Egan
- The Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Arielle Elkrief
- Segal Cancer Centre, Jewish General Hospital, McGill University, Montreal, QC, Canada
| | | | | | - Matthew D Galsky
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | | | | | | | - Navid Hafez
- Smilow Cancer Hospital at Yale New Haven, New Haven, CT, USA
| | | | - Jessica E Hawley
- Herbert Irving Comprehensive Cancer Center at Columbia University, New York, NY, USA
| | - Emily Hsu
- University of Connecticut, Farmington, CT, USA; Hartford Health Care, Hartford, CT, USA
| | - Anup Kasi
- University of Kansas Medical Center, Kansas City, KS, USA
| | - Ali R Khaki
- Fred Hutchinson Cancer Research Center, Seattle, WA, USA; University of Washington, Seattle, WA, USA
| | | | - Colleen Lewis
- Winship Cancer Institute of Emory University, Atlanta, GA, USA
| | | | - Tyler Masters
- Smilow Cancer Hospital at Yale New Haven, New Haven, CT, USA
| | - Rana R McKay
- Moores Cancer Center, University of California San Diego, La Jolla, CA, USA
| | - Ruben A Mesa
- Mays Cancer Center, UT Health San Antonio MD Anderson Cancer Center, San Antonio, TX, USA
| | - Alicia K Morgans
- The Robert H Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL, USA
| | - Mary F Mulcahy
- The Robert H Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL, USA
| | | | | | | | - Kerry Reynolds
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Lane R Rosen
- Willis-Knighton Cancer Center, Shreveport, LA, USA
| | - Rachel Rosovsky
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Mary Salazar
- Mays Cancer Center, UT Health San Antonio MD Anderson Cancer Center, San Antonio, TX, USA
| | | | | | - Justin A Shaya
- Moores Cancer Center, University of California San Diego, La Jolla, CA, USA
| | | | | | - Suki Subbiah
- Stanley S Scott Cancer Center, LSU Health, New Orleans, LA, USA
| | - Donald C Vinh
- McGill University Health Centre, Montreal, QC, Canada
| | - Firas H Wehbe
- The Robert H Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL, USA
| | | | | | | | - Zhuoer Xie
- Mayo Clinic Cancer Center, Rochester, MN, USA
| | - Albert Yeh
- Fred Hutchinson Cancer Research Center, Seattle, WA, USA; University of Washington, Seattle, WA, USA
| | | | - Alice Y Zhou
- Siteman Cancer Center, Washington University School of Medicine, St Louis, MO, USA
| | - Leyre Zubiri
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Sanjay Mishra
- Vanderbilt-Ingram Cancer Center at Vanderbilt University Medical Center, Nashville, TN, USA
| | - Gary H Lyman
- Fred Hutchinson Cancer Research Center, Seattle, WA, USA; University of Washington, Seattle, WA, USA
| | - Brian I Rini
- Vanderbilt-Ingram Cancer Center at Vanderbilt University Medical Center, Nashville, TN, USA
| | - Jeremy L Warner
- Vanderbilt-Ingram Cancer Center at Vanderbilt University Medical Center, Nashville, TN, USA.
| |
Collapse
|
50
|
Yang L, Wen M, Liu X, Wang K, Wang Y. Feikang granules ameliorate pulmonary inflammation in the rat model of chronic obstructive pulmonary disease via TLR2/4-mediated NF-κB pathway. BMC Complement Med Ther 2020; 20:170. [PMID: 32493287 PMCID: PMC7268508 DOI: 10.1186/s12906-020-02964-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Accepted: 05/19/2020] [Indexed: 12/21/2022] Open
Abstract
Purpose Several reports have shown that traditional Chinese medicine could be an alternative therapeutic approach for COPD patients, but the mechanism remains unknown. The present study aimed to examine the effects of Feikang granules in a COPD model rat and investigate the possible mechanisms via Toll-like receptor (TLR)/ nuclear factor kappa B (NF-κB) signaling. Methods The COPD model rats were treated with Feikang granules, dexamethasone, or normal saline. The pulmonary function; lung tissue histology; levels of inflammatory cytokines; mRNA levels of TNFα, IL-6, TLR4, and TLR2; and protein levels of TLR4, TLR2, p-IκB, IκB and P65 in lung tissues were evaluated. Results The present study confirmed that the pro-inflammatory cytokines, TNF-α, IL-1β, IL-6, and IL-17 levels were elevated and the pulmonary function and morphology are altered in COPD model rats. The TLR2 and TLR4 -mediated NF-kB signaling pathway plays a role in the mechanism of action. Feikang granules, a type of Chinese herbal medicine, significantly reduced LPS induced inflammatory cytokines release from lung tissue and alveolar macrophage in a dose-dependent manner. These medical herbs also prevented TLR2/4 and IκB downregulation and reversed the p-IκB and NF-κB p65 upregulation of the lung tissue in the COPD rats. Feikang granules were also found to protect against pulmonary dysfunction and pathological changes in the COPD rats. Conclusion The Chinese herbal medicine formula Feikang granules prevent pulmonary inflammation and improve pulmonary function, suggesting that Feikang granules may be an effective treatment for chronic pulmonary diseases, such as COPD.
Collapse
Affiliation(s)
- Liuliu Yang
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, No. 16 Airport Road, Baiyun District, Guangzhou, 510405, Guangdong, China.
| | - Minyong Wen
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, No. 16 Airport Road, Baiyun District, Guangzhou, 510405, Guangdong, China
| | - Xiaohong Liu
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, No. 16 Airport Road, Baiyun District, Guangzhou, 510405, Guangdong, China
| | - Kai Wang
- Guangzhou University of Chinese Medicine, Guangzhou, 510006, Guangdong, China
| | - Yong Wang
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, No. 16 Airport Road, Baiyun District, Guangzhou, 510405, Guangdong, China
| |
Collapse
|