1
|
Pleiss KL, Mosley DD, Bauer CD, Bailey KL, Ochoa CA, Knoell DL, Wyatt TA. Comparative effects of e-cigarette and conventional cigarette smoke on in vitro bronchial epithelial cell responses. Toxicol Lett 2025; 407:32-40. [PMID: 40101882 PMCID: PMC12011527 DOI: 10.1016/j.toxlet.2025.03.003] [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: 09/09/2024] [Revised: 02/21/2025] [Accepted: 03/08/2025] [Indexed: 03/20/2025]
Abstract
Because of cigarette smoking, chronic lung diseases are the third leading cause of death in the United States. Electronic cigarettes (e-cig) were originally marketed as harm reduction devices for cigarette smokers due to low success rates with traditional smoking cessation methods. While several studies show that cigarette smoke causes damage to the lungs, comparative research assessing the injury profile of e-cig to traditional cigarettes is still limited. Comparative lung injury studies are crucial in determining the validity of e-cig as a harm reduction device for chronic smokers and can be used to assess the quality of alternate nicotine delivery options to reduce the morbidity and mortality caused by cigarettes. We hypothesize that exposure to JUUL to e-cig vapor produces decreased in vitro markers of lung injury in comparison to cigarette smoke extract at equivalent and higher nicotine concentrations to that from CSE. We compared the extent of injury to airway epithelial tissue from cigarettes and e-cig using various assays of cellular function, including ciliary beat frequency (CBF), wound closure, barrier function, cytokine release, and kinase activity. Cells were treated with various concentrations of Virginia Tobacco-flavored JUUL™ vapor extract (JVE) and cigarette smoke extract (CSE) either normalized for nicotine concentration or equivalent % dilutions from a 100 % stock extract. CSE stimulated cilia in the short term, but slowed cilia after several hours of exposure, whereas cells treated with JVE showed no significant changes in CBF. CSE slowed wound repair, but nicotine-equivalent doses of JVE did not significantly slow wound repair. CSE increased epithelial cell monolayer permeability and interleukin release in a concentration-dependent manner, but these were not observed with JVE treatment. Kinase activity assays revealed activation translocation of protein kinase C (PKC) activity in cells treated with CSE, but no such change in PKC activity was observed in JVE groups. The results of these in vitro assays suggest that at nicotine-equivalent doses, JVE from Virginia Tobacco-flavored JUUL does not impact the airway epithelium in the same manner as CSE. The lack of evidence for in vitro tissue injury in this study caused by JUUL™ vapor extract is not a justification for the harm posed by nicotine addiction, particularly at the high levels of nicotine contained in these products which are several times the legal limit of many countries.
Collapse
Affiliation(s)
- K L Pleiss
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, College of Medicine, University of Nebraska Medical Center, Omaha, NE, United States
| | - D D Mosley
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, College of Medicine, University of Nebraska Medical Center, Omaha, NE, United States
| | - C D Bauer
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, College of Medicine, University of Nebraska Medical Center, Omaha, NE, United States
| | - K L Bailey
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, College of Medicine, University of Nebraska Medical Center, Omaha, NE, United States; Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, NE, United States
| | - C A Ochoa
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, College of Medicine, University of Nebraska Medical Center, Omaha, NE, United States
| | - D L Knoell
- Department of Pharmacy Practice and Science, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE, United States
| | - T A Wyatt
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, College of Medicine, University of Nebraska Medical Center, Omaha, NE, United States; Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, NE, United States; Department of Environmental, Agricultural & Occupational Health, College of Public Health, University of Nebraska Medical Center, Omaha, NE, United States.
| |
Collapse
|
2
|
Rached RA, Shakya AK, Fulgheri F, Aroffu M, Castangia I, García-Villén F, Ferraro M, Fernàndez-Busquets X, Pedraz JL, Louka N, Maroun RG, Manconi M, Manca ML. Resveratrol and grape pomace extract incorporated in modified phospholipid vesicles: A potential strategy to mitigate cigarette smoke-induced oxidative stress. Free Radic Biol Med 2025; 230:151-162. [PMID: 39952333 DOI: 10.1016/j.freeradbiomed.2025.02.009] [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: 11/26/2024] [Revised: 02/05/2025] [Accepted: 02/08/2025] [Indexed: 02/17/2025]
Abstract
In this study, the extraction process of grape pomace from the Lebanese autochthonous cultivar Asswad Karech was enhanced through the selection of specific parameters, yielding an antioxidant extract (20 mg/mL) that was co-loaded with resveratrol (5 mg/mL) into phospholipid vesicles containing penetration enhancers (PEVs). Propylene glycol (PG) was incorporated as a penetration enhancer at concentrations of 10, 20, and 30 % to obtain 10 PG-PEVs, 20 PG-PEVs, and 30 PG-PEVs. Vesicle preparation was achieved through direct sonication, yielding unilamellar and bilamellar vesicles with an average size of ∼205; 234 nm, a monodisperse distribution (polydispersity index <0.3), and a negative surface charge (∼-54;-56 mV). The formulations containing 30 % propylene glycol exhibited long-term stability, maintaining a consistent mean diameter over 12 months at room temperature (25 °C). Upon nebulization using the Next Generation Impactor, the vesicular dispersions successfully reached the deepest stages of the impactor, mimicking deposition in the lower respiratory airways. Biocompatibility studies on A549 and CuFi-1 cell lines demonstrated that the vesicles co-loaded with grape extract and resveratrol effectively counteracted apoptosis induced by hydrogen peroxide. Furthermore, when 16HBE bronchial epithelial cells were exposed to cigarette smoke extract, vesicles containing 30 % propylene glycol inhibited reactive oxygen species (ROS) generation. These findings highlight the potential of phospholipid vesicles co-loaded with grape pomace extract and resveratrol, particularly those formulated with 30 % propylene glycol, for pulmonary administration to mitigate oxidative damage associated with cigarette smoke exposure and related respiratory diseases.
Collapse
Affiliation(s)
- Rita Abi Rached
- Department of Life and Environmental Sciences, University of Cagliari, University Campus, S.P. Monserrato-Sestu Km 0.700, 09042, Monserrato, CA, Italy; Centre d'Analyses et de Recherche, Unité de Recherche TVA, Laboratoire CTA, Faculté des Sciences, Université Saint-Joseph de Beyrouth, B.P. 17-5208 Riad El Solh, Beirut, 1104 2020, Lebanon.
| | - Ashok K Shakya
- Pharmacological & Diagnostic Research Center, Faculty of Pharmacy, Al-Ahliyya Amman University, Amman, 19328, Jordan.
| | - Federica Fulgheri
- Department of Life and Environmental Sciences, University of Cagliari, University Campus, S.P. Monserrato-Sestu Km 0.700, 09042, Monserrato, CA, Italy.
| | - Matteo Aroffu
- Department of Life and Environmental Sciences, University of Cagliari, University Campus, S.P. Monserrato-Sestu Km 0.700, 09042, Monserrato, CA, Italy.
| | - Ines Castangia
- Department of Life and Environmental Sciences, University of Cagliari, University Campus, S.P. Monserrato-Sestu Km 0.700, 09042, Monserrato, CA, Italy.
| | - Fátima García-Villén
- Department of Pharmacy and Pharmaceutical Technology, School of Pharmacy, University of Granada, Campus of Cartuja, 18071, Granada, Spain.
| | - Maria Ferraro
- Istituto per la Ricerca e l'Innovazione Biomedica (IRIB), CNR, Via Ugo La Malfa 153, 90146, Palermo, Italy.
| | - Xavier Fernàndez-Busquets
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, Baldiri Reixac 10-12, 08028, Barcelona, Spain; Barcelona Institute for Global Health (ISGlobal), Hospital Clínic-Universitat de Barcelona, Rosselló 149-153, 08036, Barcelona, Spain.
| | - Jose Luis Pedraz
- NanoBioCel Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country (UPV/EHU), Biomedical Research Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), BioAraba, NanoBioCel research Group, Vitoria-Gasteiz, Spain.
| | - Nicolas Louka
- Centre d'Analyses et de Recherche, Unité de Recherche TVA, Laboratoire CTA, Faculté des Sciences, Université Saint-Joseph de Beyrouth, B.P. 17-5208 Riad El Solh, Beirut, 1104 2020, Lebanon.
| | - Richard G Maroun
- Centre d'Analyses et de Recherche, Unité de Recherche TVA, Laboratoire CTA, Faculté des Sciences, Université Saint-Joseph de Beyrouth, B.P. 17-5208 Riad El Solh, Beirut, 1104 2020, Lebanon.
| | - Maria Manconi
- Department of Life and Environmental Sciences, University of Cagliari, University Campus, S.P. Monserrato-Sestu Km 0.700, 09042, Monserrato, CA, Italy.
| | - Maria Letizia Manca
- Department of Life and Environmental Sciences, University of Cagliari, University Campus, S.P. Monserrato-Sestu Km 0.700, 09042, Monserrato, CA, Italy.
| |
Collapse
|
3
|
Qiu Y, Hu G. Lung-on-a-chip: From design principles to disease applications. BIOMICROFLUIDICS 2025; 19:021501. [PMID: 40161998 PMCID: PMC11954643 DOI: 10.1063/5.0257908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Accepted: 02/12/2025] [Indexed: 04/02/2025]
Abstract
To address the growing need for accurate lung models, particularly in light of respiratory diseases, lung cancer, and the COVID-19 pandemic, lung-on-a-chip technology is emerging as a powerful alternative. Lung-on-a-chip devices utilize microfluidics to create three-dimensional models that closely mimic key physiological features of the human lung, such as the air-liquid interface, mechanical forces associated with respiration, and fluid dynamics. This review provides a comprehensive overview of the fundamental components of lung-on-a-chip systems, the diverse fabrication methods used to construct these complex models, and a summary of their wide range of applications in disease modeling and aerosol deposition studies. Despite existing challenges, lung-on-a-chip models hold immense potential for advancing personalized medicine, drug development, and disease prevention, offering a transformative approach to respiratory health research.
Collapse
Affiliation(s)
- Yan Qiu
- Department of Engineering Mechanics, State Key Laboratory of Fluid Power and Mechatronic Systems, Zhejiang University, Hangzhou 310027, China
| | - Guoqing Hu
- Department of Engineering Mechanics, State Key Laboratory of Fluid Power and Mechatronic Systems, Zhejiang University, Hangzhou 310027, China
| |
Collapse
|
4
|
Lin Y, Lin X, Ren C, Song L, Gu C. Association of pan-immune inflammation value and lung health in adults. BMC Pulm Med 2025; 25:18. [PMID: 39810110 PMCID: PMC11734563 DOI: 10.1186/s12890-025-03493-4] [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: 10/17/2024] [Accepted: 01/09/2025] [Indexed: 01/16/2025] Open
Abstract
BACKGROUND Lung health is intricately linked with inflammation. The pan-immune-inflammation value (PIV) emerges as a promising biomarker, offering reflection into systemic inflammatory states and assisting in the prognosis of diverse diseases. This research aims to explore the associations between PIV and respiratory symptoms, respiratory diseases and lung function. METHODS The study was a cross-sectional population study from the National Health and Nutrition Examination Survey (NHANES). Restricted cubic spline (RCS) models were conducted to explore the relationships between PIV and respiratory health outcomes, while weighted linear regression models and weighted logistic regression models were the ones used for regression analysis. Trend tests probed the evolving relationship among PIV quartiles and outcomes. The study incorporated subgroup analysis and interaction tests to examine associations within specific subpopulations. RESULTS From the cohort of 6,263 participants, a distinct negative correlation was identified between PIV and lung health. Subsequent to confounding factors, a 100-unit increment in PIV was linked to a 2% increase in the incidence of cough and phlegm (OR, 95% CI: 1.02, 1.00 to 1.05; 1.02, 1.00 to 1.04). Additionally, higher PIV was associated with reductions in FEV1 (MD, 95% CI: -5.37, -9.10 to -1.64) and FVC (MD, 95% CI: -5.75, -10.34 to -1.15). Categorizing PIV into quartiles revealed an ascending trend: A significantly higher risk of cough/phlegm/wheeze was found in participants in the second/third/fourth PIV quartile compared to those in the first PIV quartile (all p for trend < 0.05). Moreover, lung function indicators (FEV1, FEV1%, FVC, FVC%, FEV1/FVC) declined significantly in the fourth quartile (all p for trend < 0.05). Besides, a nonlinear relationship between PIV and outcomes was evident. Subgroup analysis revealed variations in these associations stratified by gender, age, smoking and drinking status, as well as certain disease history. CONCLUSIONS The study highlighted the potential connections between PIV and respiratory symptoms, respiratory diseases and lung function. Monitoring PIV level could provide valuable insights into the inflammatory status and may inform clinical approaches for managing respiratory health.
Collapse
Affiliation(s)
- Ya Lin
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Xiao Lin
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Chufan Ren
- Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Lanlan Song
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Chao Gu
- Department of Respiratory Medicine, The First Hospital of Jiaxing (Affiliated Hospital of Jiaxing University), 1882 South Zhonghuan Road, Jiaxing, Zhejiang, 314000, China.
| |
Collapse
|
5
|
Wu L, Zhang E, Tu Y, Chen Y, Wang C, Ren Y, Fang B. Inherent immunity and adaptive immunity: Mechanism and role in AECOPD. Innate Immun 2025; 31:17534259251322612. [PMID: 40017227 PMCID: PMC11869301 DOI: 10.1177/17534259251322612] [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: 04/20/2024] [Revised: 12/20/2024] [Accepted: 02/04/2025] [Indexed: 03/01/2025] Open
Abstract
Acute exacerbation of chronic obstructive pulmonary disease (AECOPD) is the leading cause of hospitalization and mortality in COPD patients. The occurrence of antibiotic resistance and the progression of non-infectious diseases contribute to poor patient outcomes. Thus, a comprehensive understanding of the mechanisms underlying AECOPD is essential for effective prevention. It is widely acknowledged that the immune system plays a fundamental role in pathogen clearance and the development of inflammation. Immune dysregulation, either due to deficiency or hyperactivity, has been implicated in AECOPD pathogenesis. Therefore, the purpose of this review is to investigate the possible mechanisms underlying dysregulated immune function and disease progression in COPD patients, specifically focusing on the innate and adaptive immune responses. The ultimate aim is to provide new insights for clinical prevention and treatment strategies targeting AECOPD.
Collapse
Affiliation(s)
- Linguangjin Wu
- Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Emergency Department, Shanghai, China
| | - Erxin Zhang
- Department of Gastroenterology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yadan Tu
- Chongqing Hospital of Traditional Chinese Medicine, Classic Department of Traditional Chinese Medicine, Chongqing, China
| | - Yong Chen
- Chongqing Hospital of Traditional Chinese Medicine, Classic Department of Traditional Chinese Medicine, Chongqing, China
| | - Chenghu Wang
- Chongqing Hospital of Traditional Chinese Medicine, Classic Department of Traditional Chinese Medicine, Chongqing, China
| | - Yi Ren
- Chongqing Hospital of Traditional Chinese Medicine, Classic Department of Traditional Chinese Medicine, Chongqing, China
| | - Bangjiang Fang
- Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Emergency Department, Shanghai, China
| |
Collapse
|
6
|
Hirano M, Iwata K, Yamada Y, Shinoda Y, Yamazaki M, Hino S, Ikeda A, Shimizu A, Otsuka S, Nakagawa H, Watanabe Y. AlveoMPU: Bridging the Gap in Lung Model Interactions Using a Novel Alveolar Bilayer Film. Polymers (Basel) 2024; 16:1486. [PMID: 38891433 PMCID: PMC11174738 DOI: 10.3390/polym16111486] [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: 04/16/2024] [Revised: 05/20/2024] [Accepted: 05/21/2024] [Indexed: 06/21/2024] Open
Abstract
The alveoli, critical sites for gas exchange in the lungs, comprise alveolar epithelial cells and pulmonary capillary endothelial cells. Traditional experimental models rely on porous polyethylene terephthalate or polycarbonate membranes, which restrict direct cell-to-cell contact. To address this limitation, we developed AlveoMPU, a new foam-based mortar-like polyurethane-formed alveolar model that facilitates direct cell-cell interactions. AlveoMPU features a unique anisotropic mortar-shaped configuration with larger pores at the top and smaller pores at the bottom, allowing the alveolar epithelial cells to gradually extend toward the bottom. The underside of the film is remarkably thin, enabling seeded pulmonary microvascular endothelial cells to interact with alveolar epithelial cells. Using AlveoMPU, it is possible to construct a bilayer structure mimicking the alveoli, potentially serving as a model that accurately simulates the actual alveoli. This innovative model can be utilized as a drug-screening tool for measuring transepithelial electrical resistance, assessing substance permeability, observing cytokine secretion during inflammation, and evaluating drug efficacy and pharmacokinetics.
Collapse
Affiliation(s)
- Minoru Hirano
- Frontier Research Management Office, Toyota Central R&D Labs., Inc., 41-1 Yokomichi, Nagakute 480-1192, Aichi, Japan; (Y.Y.); (Y.W.)
| | - Kosuke Iwata
- Organic Device Development Department, Material Development Division, Toyoda Gosei Co., Ltd., 1-1 Higashitakasuka, Futatsudera, Ama 490-1207, Aichi, Japan; (K.I.); (M.Y.); (S.H.); (A.I.); (A.S.); (S.O.); (H.N.)
| | - Yuri Yamada
- Frontier Research Management Office, Toyota Central R&D Labs., Inc., 41-1 Yokomichi, Nagakute 480-1192, Aichi, Japan; (Y.Y.); (Y.W.)
| | - Yasuhiko Shinoda
- Organic Device Development Department, Material Development Division, Toyoda Gosei Co., Ltd., 1-1 Higashitakasuka, Futatsudera, Ama 490-1207, Aichi, Japan; (K.I.); (M.Y.); (S.H.); (A.I.); (A.S.); (S.O.); (H.N.)
| | - Masateru Yamazaki
- Organic Device Development Department, Material Development Division, Toyoda Gosei Co., Ltd., 1-1 Higashitakasuka, Futatsudera, Ama 490-1207, Aichi, Japan; (K.I.); (M.Y.); (S.H.); (A.I.); (A.S.); (S.O.); (H.N.)
| | - Sayaka Hino
- Organic Device Development Department, Material Development Division, Toyoda Gosei Co., Ltd., 1-1 Higashitakasuka, Futatsudera, Ama 490-1207, Aichi, Japan; (K.I.); (M.Y.); (S.H.); (A.I.); (A.S.); (S.O.); (H.N.)
| | - Aya Ikeda
- Organic Device Development Department, Material Development Division, Toyoda Gosei Co., Ltd., 1-1 Higashitakasuka, Futatsudera, Ama 490-1207, Aichi, Japan; (K.I.); (M.Y.); (S.H.); (A.I.); (A.S.); (S.O.); (H.N.)
| | - Akiko Shimizu
- Organic Device Development Department, Material Development Division, Toyoda Gosei Co., Ltd., 1-1 Higashitakasuka, Futatsudera, Ama 490-1207, Aichi, Japan; (K.I.); (M.Y.); (S.H.); (A.I.); (A.S.); (S.O.); (H.N.)
| | - Shuhei Otsuka
- Organic Device Development Department, Material Development Division, Toyoda Gosei Co., Ltd., 1-1 Higashitakasuka, Futatsudera, Ama 490-1207, Aichi, Japan; (K.I.); (M.Y.); (S.H.); (A.I.); (A.S.); (S.O.); (H.N.)
| | - Hiroyuki Nakagawa
- Organic Device Development Department, Material Development Division, Toyoda Gosei Co., Ltd., 1-1 Higashitakasuka, Futatsudera, Ama 490-1207, Aichi, Japan; (K.I.); (M.Y.); (S.H.); (A.I.); (A.S.); (S.O.); (H.N.)
| | - Yoshihide Watanabe
- Frontier Research Management Office, Toyota Central R&D Labs., Inc., 41-1 Yokomichi, Nagakute 480-1192, Aichi, Japan; (Y.Y.); (Y.W.)
| |
Collapse
|
7
|
Zhu XF, Hu YQ, Dai ZC, Li XJ, Zhang J. Associations between trans fatty acids and systemic immune-inflammation index: a cross-sectional study. Lipids Health Dis 2024; 23:122. [PMID: 38678208 PMCID: PMC11055356 DOI: 10.1186/s12944-024-02109-w] [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: 01/22/2024] [Accepted: 04/15/2024] [Indexed: 04/29/2024] Open
Abstract
BACKGROUND Previous studies have demonstrated that trans fatty acids (TFAs) intake was linked to an increased risk of chronic diseases. As a novel systemic inflammatory biomarker, the clinical value and efficacy of the systemic immune-inflammation index (SII) have been widely explored. However, the association between TFAs and SII is still unclear. Therefore, the study aims to investigate the connection between TFAs and SII in US adults. METHODS The study retrieved data from the National Health and Nutrition Examination Survey (NHANES) for the years 1999-2000 and 2009-2010. Following the exclusion of ineligible participants, the study encompassed a total of 3047 individuals. The research employed a multivariate linear regression model to investigate the connection between circulating TFAs and SII. Furthermore, the restricted cubic spline (RCS) model was utilized to evaluate the potential nonlinear association. Subgroup analysis was also conducted to investigate the latent interactive factors. RESULTS In this investigation, participants exhibited a mean age of 47.40 years, with 53.91% of them being female. Utilizing a multivariate linear regression model, the independent positive associations between the log2-transformed palmitelaidic acid, the log2 transformed-vaccenic acid, the log2-transformed elaidic acid, the log2-transformed linolelaidic acid, and the log2-transformed-total sum of TFAs with the SII (all P < 0.05) were noted. In the RCS analysis, no nonlinear relationship was observed between the log2-transformed palmitelaidic acid, the log2 transformed-vaccenic acid, the log2-transformed elaidic acid, the log2-transformed linolelaidic acid, the log2-transformed-total sum of TFAs and the SII (all P for nonlinear > 0.05). For the stratified analysis, the relationship between the circulating TFAs and the SII differed by the obesity status and the smoking status. CONCLUSIONS A positive association was investigated between three types of TFA, the sum of TFAs, and the SII in the US population. Additional rigorously designed studies are needed to verify the results and explore the potential mechanism.
Collapse
Affiliation(s)
- Xiao-Feng Zhu
- Department of Clinical Medicine, The Nanshan College of Guangzhou Medical University, Guangzhou, 511436, China
| | - Yu-Qi Hu
- Department of Clinical Medicine, The Third Clinical School of Guangzhou Medical University, Guangzhou, 511436, China
| | - Zhi-Cheng Dai
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 201600, China
| | - Xiu-Juan Li
- Department of Clinical Medicine, The Third Clinical School of Guangzhou Medical University, Guangzhou, 511436, China
| | - Jing Zhang
- Second Department of Infectious Disease, Shanghai Fifth People's Hospital, Fudan University, Shanghai, 201100, China.
| |
Collapse
|
8
|
Wei K, Li Y, Du B, Wu J. Differences in Airway Remodeling and Emphysematous Lesions between Rats Exposed to Smoke from New-Type and Conventional Tobacco Varieties. Antioxidants (Basel) 2024; 13:511. [PMID: 38790616 PMCID: PMC11117731 DOI: 10.3390/antiox13050511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 04/17/2024] [Accepted: 04/22/2024] [Indexed: 05/26/2024] Open
Abstract
Genes from Perilla frutescens and Ocimum basilicum were introduced into N. tabacum L. var. HHY via distant hybridization, and the new-type tobacco varieties "Zisu" and "Luole" were developed, with noticeable differences in chemical composition. Smoking is the leading cause of chronic obstructive pulmonary disease (COPD), and its pathogenesis is complex. In the present study, 48 male Sprague-Dawley (SD) rats were randomly divided into four groups, namely, the control, "HHY", "Zisu" and "Luole", and then exposed to fresh air/cigarette smoke (CS) for 30 days and 60 days. The COPD model was constructed, and their health hazards were compared and evaluated. CS from different tobacco varieties influenced rats in varying degrees at the tissue, cell and molecular levels. The rats in the "HHY" group showed obvious symptoms, such as cough and dyspnea, which were less severe in the "Zisu" and "Luole" groups. Pathological and morphological analyses, including scores, MLI, MAN, WAt/Pbm and WAm/Pbm, showed that "Zisu" and "Luole" caused less damage to the airways and lung parenchyma than "HHY". Significant increases in the numbers of total leukocytes and neutrophils in the BALF were found in "HHY" compared to those in "Zisu" and "Luole". Moreover, they caused less oxidative stress and apoptosis in lung tissues, as reflected by indicators such as ROS, MDA, T-AOC, GSH, the apoptotic index and the ratio of Bcl-2 to Bax. "Zisu" and "Luole" even altered the ratios of MMP-9/TIMP-1 and IFN-γ/IL-4 in lung tissues to a lesser degree. These differences between CS-exposed rats may be closely related to the altered expression of Nrf2, p38 MAPK and p-p38 MAPK. Changes in chemical composition via introducing genes from some medicinal plants may be an attractive strategy for tobacco harm reduction.
Collapse
Affiliation(s)
- Keqiang Wei
- School of Life Science, Shanxi University, Taiyuan 030006, China
| | | | | | | |
Collapse
|
9
|
Ma Q, Shen Y, Guo W, Feng K, Huang T, Cai Y. Machine Learning Reveals Impacts of Smoking on Gene Profiles of Different Cell Types in Lung. Life (Basel) 2024; 14:502. [PMID: 38672772 PMCID: PMC11051039 DOI: 10.3390/life14040502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 04/03/2024] [Accepted: 04/10/2024] [Indexed: 04/28/2024] Open
Abstract
Smoking significantly elevates the risk of lung diseases such as chronic obstructive pulmonary disease (COPD) and lung cancer. This risk is attributed to the harmful chemicals in tobacco smoke that damage lung tissue and impair lung function. Current research on the impact of smoking on gene expression in specific lung cells is limited. This study addresses this gap by analyzing gene expression profiles at the single-cell level from 43,539 lung endothelial cells, 234,349 lung epithelial cells, 189,843 lung immune cells, and 16,031 lung stromal cells using advanced machine learning techniques. The data, categorized by different lung cell types, were classified into three smoking states: active smoker, former smoker, and never smoker. Each cell sample encompassed 28,024 feature genes. Employing an incremental feature selection method within a computational framework, several specific genes have been identified as potential markers of smoking status in different lung cell types. These include B2M, EEF1A1, and TPT1 in lung endothelial cells; FTL and MT-ATP8 in lung epithelial cells; HLA-B and HLA-C in lung immune cells; and HSP90B1 and LCN2 in lung stroma cells. Additionally, this study developed quantitative rules for representing the gene expression patterns related to smoking. This research highlights the potential of machine learning in oncology, enhancing our molecular understanding of smoking's harm and laying the groundwork for future mechanism-based studies.
Collapse
Affiliation(s)
- Qinglan Ma
- School of Life Sciences, Shanghai University, Shanghai 200444, China;
| | - Yulong Shen
- Department of Radiotherapy, Strategic Support Force Medical Center, Beijing 100101, China;
| | - Wei Guo
- Key Laboratory of Stem Cell Biology, Shanghai Jiao Tong University School of Medicine (SJTUSM) & Shanghai Institutes for Biological Sciences (SIBS), Chinese Academy of Sciences (CAS), Shanghai 200030, China;
| | - Kaiyan Feng
- Department of Computer Science, Guangdong AIB Polytechnic College, Guangzhou 510507, China;
| | - Tao Huang
- Bio-Med Big Data Center, CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Yudong Cai
- School of Life Sciences, Shanghai University, Shanghai 200444, China;
| |
Collapse
|
10
|
Kim J, Cho Y, Oh GJ, Park HB, Yang MJ, Park CM, Kim YH, Choi KC, Go RE, Kim MS. Repeated intratracheal instillation of whole-cigarette smoke condensate to assess lung damage in a rat model. ENVIRONMENTAL TOXICOLOGY 2024; 39:2304-2315. [PMID: 38148711 DOI: 10.1002/tox.24113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 08/29/2023] [Accepted: 12/11/2023] [Indexed: 12/28/2023]
Abstract
Cigarette smoke induces an inflammatory response in the lungs by recruiting inflammatory cells, leading to lung diseases such as lung cancer, chronic obstructive pulmonary disease, and pulmonary fibrosis. Existing inhalation exposure methods for assessing the adverse effects of cigarette smoke require expensive equipment and are labor-intensive. Therefore, we attempted to develop a novel method to assess these adverse effects using intratracheal instillation (ITI) of whole cigarette smoke condensate (WCSC). The WCSC (0, 5, 10, or 20 mg/mL) was administered by ITI once daily for 6 or 12 days using an automatic video instillator. Repeated WCSC ITI increased the lung weight, and monocyte chemoattractant protein-1 (MCP-1), neutrophil, and lymphocyte levels within bronchoalveolar lavage fluid compared to the control. In the histopathological analysis of the lung tissue, a mild inflammatory response was observed in the 6 and 12 days 20 mg/mL WCSC exposure groups. The genome-wide RNA-seq expression patterns revealed that inflammatory and immune response-related genes, such as the chemokine signaling pathway, Th1/Th2 cell differentiation, and cytokine-cytokine receptor interaction, were employed following WCSC exposure. In addition, MCP-1 was time-dependent and increased in the 10 mg/mL exposure group compared to the control group. These results suggested that the WCSC might induce the potential pulmonary inflammatory response. Furthermore, we proposed that ITI may be a rapid and effective method of evaluating the adverse effects of WCSC within a short exposure period (less than 2 weeks), and it can be used to evaluate cigarette inhalation toxicity studies as an alternative method.
Collapse
Affiliation(s)
- Jinhee Kim
- Inhalation Toxicology Research Group, Korea Institute of Toxicology (KIT), Jeongeup, Jeonbuk, Republic of Korea
| | - Yoon Cho
- Inhalation Toxicology Research Group, Korea Institute of Toxicology (KIT), Jeongeup, Jeonbuk, Republic of Korea
| | - Gi-Jun Oh
- Inhalation Toxicology Research Group, Korea Institute of Toxicology (KIT), Jeongeup, Jeonbuk, Republic of Korea
| | - Hae-Bin Park
- Inhalation Toxicology Research Group, Korea Institute of Toxicology (KIT), Jeongeup, Jeonbuk, Republic of Korea
| | - Mi Jin Yang
- Inhalation Toxicology Research Group, Korea Institute of Toxicology (KIT), Jeongeup, Jeonbuk, Republic of Korea
| | - Chul-Min Park
- Inhalation Toxicology Research Group, Korea Institute of Toxicology (KIT), Jeongeup, Jeonbuk, Republic of Korea
| | - Yong-Hyun Kim
- Inhalation Toxicology Research Group, Korea Institute of Toxicology (KIT), Jeongeup, Jeonbuk, Republic of Korea
- Department of Environment & Energy, Jeonbuk National University, Jeonju, Jeonbuk, Republic of Korea
| | - Kyung-Chul Choi
- Laboratory of Biochemistry and Immunology, College of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea
| | - Ryeo-Eun Go
- Laboratory of Biochemistry and Immunology, College of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea
| | - Min-Seok Kim
- Inhalation Toxicology Research Group, Korea Institute of Toxicology (KIT), Jeongeup, Jeonbuk, Republic of Korea
| |
Collapse
|
11
|
Hussain N, Ikram N, Khan KUR, Hussain L, Alqahtani AM, Alqahtani T, Hussain M, Suliman M, Alshahrani MY, Sitohy B. Cichorium intybus L. significantly alleviates cigarette smoke-induced acute lung injury by lowering NF-κB pathway activation and inflammatory mediators. Heliyon 2023; 9:e22055. [PMID: 38045213 PMCID: PMC10692792 DOI: 10.1016/j.heliyon.2023.e22055] [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: 05/06/2023] [Revised: 11/02/2023] [Accepted: 11/02/2023] [Indexed: 12/05/2023] Open
Abstract
Background Cigarette smoke (CS) is one of the primary causes of acute lung injury (ALI) via provoking pulmonary inflammation and oxidative stress. Despite substantial studies, no effective treatment for ALI is presently available. Purpose New prospective treatment options for ALI are required. Thus, this project was designed to investigate the in vivo and in vitro protective effects of 70 % methanolic-aqueous crude extract of whole plant of Cichorium intybus (Ci.Mce) against CS-induced ALI. Study design /methods: Initially, male Swiss albino mice were subjected to whole-body CS exposure for 10 continuous days to prepare CS-induced ALI models. Normal saline (10 mL/kg), Ci.Mce (100, 200, 300 mg/kg), and Dexamethasone (1 mg/kg) were orally administered to respective animal groups 1 h prior to CS-exposure. 24 hrs after the last CS-exposure, BALF and lungs were harvested to study the key characteristics of ALI. Next, HPLC analysis was done to explore the phytoconstituents. Results Ci.Mce exhibited significant reductions in lung macrophage and neutrophil infiltration, lung weight coefficient, and albumin exudation. Additionally, it effectively ameliorated lung histopathological alterations and hypoxemia. Notably, Ci.Mce exerted inhibitory effects on the excessive generation of IL-6, IL-1β, and KC in both CS-induced ALI murine models and CSE-stimulated RAW 264.7 macrophages. Noteworthy benefits included the attenuation of oxidative stress induced by CS, evidenced by decreased levels of MDA, TOS, and MPO, alongside enhanced TAC production. Furthermore, Ci.Mce demonstrated a marked reduction in CS-induced NF-κB expression, both in vivo and in vitro. Conclusion Consequently, Cichorium intybus could be a therapeutic option for CS-induced ALI due to its ability to suppress inflammatory reactions, mitigate oxidative stress, and quell NF-κB p65 activation.
Collapse
Affiliation(s)
- Nadia Hussain
- Department of Pharmaceutical Sciences, College of Pharmacy, Al Ain University, Al Ain, 64141, United Arab Emirates
| | - Nadia Ikram
- Faculty of Pharmacy, The Islamia University of Bahawalpur, Bahawalpur, 63100, Pakistan
| | - Kashif ur Rehman Khan
- Faculty of Pharmacy, The Islamia University of Bahawalpur, Bahawalpur, 63100, Pakistan
| | - Liaqat Hussain
- Department of Pharmacology, Government College University, Faisalabad, 38,000, Pakistan
| | - Ali M. Alqahtani
- Department of Pharmacology, College of Pharmacy, King Khalid University, Abha, 62529, Saudi Arabia
| | - Taha Alqahtani
- Department of Pharmacology, College of Pharmacy, King Khalid University, Abha, 62529, Saudi Arabia
| | - Musaddique Hussain
- Faculty of Pharmacy, The Islamia University of Bahawalpur, Bahawalpur, 63100, Pakistan
| | - Muath Suliman
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, P.O. Box 61413, Abha, 9088, Saudi Arabia
| | - Mohammad Y. Alshahrani
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, P.O. Box 61413, Abha, 9088, Saudi Arabia
| | - Basel Sitohy
- Department of Clinical Microbiology, Infection and Immunology, Umeå University, SE-90185, Umeå, Sweden
- Department of Radiation Sciences, Oncology, Umeå University, SE-90185, Umeå, Sweden
| |
Collapse
|
12
|
Upadhyay P, Wu CW, Pham A, Zeki AA, Royer CM, Kodavanti UP, Takeuchi M, Bayram H, Pinkerton KE. Animal models and mechanisms of tobacco smoke-induced chronic obstructive pulmonary disease (COPD). JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART B, CRITICAL REVIEWS 2023; 26:275-305. [PMID: 37183431 PMCID: PMC10718174 DOI: 10.1080/10937404.2023.2208886] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2023]
Abstract
Chronic obstructive pulmonary disease (COPD) is the third leading cause of death worldwide, and its global health burden is increasing. COPD is characterized by emphysema, mucus hypersecretion, and persistent lung inflammation, and clinically by chronic airflow obstruction and symptoms of dyspnea, cough, and fatigue in patients. A cluster of pathologies including chronic bronchitis, emphysema, asthma, and cardiovascular disease in the form of hypertension and atherosclerosis variably coexist in COPD patients. Underlying causes for COPD include primarily tobacco use but may also be driven by exposure to air pollutants, biomass burning, and workplace related fumes and chemicals. While no single animal model might mimic all features of human COPD, a wide variety of published models have collectively helped to improve our understanding of disease processes involved in the genesis and persistence of COPD. In this review, the pathogenesis and associated risk factors of COPD are examined in different mammalian models of the disease. Each animal model included in this review is exclusively created by tobacco smoke (TS) exposure. As animal models continue to aid in defining the pathobiological mechanisms of and possible novel therapeutic interventions for COPD, the advantages and disadvantages of each animal model are discussed.
Collapse
Affiliation(s)
- Priya Upadhyay
- Center for Health and the Environment, University of California, Davis, Davis, CA 95616 USA
| | - Ching-Wen Wu
- Center for Health and the Environment, University of California, Davis, Davis, CA 95616 USA
| | - Alexa Pham
- Center for Health and the Environment, University of California, Davis, Davis, CA 95616 USA
| | - Amir A. Zeki
- Department of Internal Medicine; Division of Pulmonary, Critical Care, and Sleep Medicine, Center for Comparative Respiratory Biology and Medicine, School of Medicine; University of California, Davis, School of Medicine; U.C. Davis Lung Center; Davis, CA USA
| | - Christopher M. Royer
- California National Primate Research Center, University of California, Davis, Davis, CA 95616 USA
| | - Urmila P. Kodavanti
- Public Health and Integrated Toxicology Division, Center for Public Health and Environmental Assessment, Office of Research and Development, U.S. Environmental Protection Agency, Research Triangle Park, NC 27711, USA
| | - Minoru Takeuchi
- Department of Animal Medical Science, Kyoto Sangyo University, Kyoto, Japan
| | - Hasan Bayram
- Koc University Research Center for Translational Medicine (KUTTAM), School of Medicine, Istanbul, Turkey
| | - Kent E. Pinkerton
- Center for Health and the Environment, University of California, Davis, Davis, CA 95616 USA
| |
Collapse
|
13
|
Mycroft K, Paplińska-Goryca M, Proboszcz M, Nejman-Gryz P, Krenke R, Górska K. Blood and Sputum Eosinophils of COPD Patients Are Differently Polarized than in Asthma. Cells 2023; 12:1631. [PMID: 37371101 DOI: 10.3390/cells12121631] [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: 04/28/2023] [Revised: 06/07/2023] [Accepted: 06/12/2023] [Indexed: 06/29/2023] Open
Abstract
Different eosinophil subpopulations have been identified in asthma and other eosinophilic disorders. However, there is a paucity of data on eosinophil subpopulations in patients with chronic obstructive pulmonary disease (COPD). The aim of this study was to compare eosinophil phenotypes in blood and induced sputum in patients with COPD, asthma and controls. Stable patients with mild-to-moderate COPD (n = 15) and asthma (n = 14) with documented blood eosinophilia ≥100 cells/µL in the year prior to the study and the control group (n = 11) were included to the study. The blood and sputum eosinophil phenotypes were analyzed by flow cytometry. IL-5, IL-13, CCL5 and eotaxin-3 levels were measured in the induced sputum. The marker expression on blood eosinophils was similar among control, asthma and COPD groups. The expressions of CD125, CD193, CD14 and CD62L were higher on blood than on sputum eosinophils in all three groups. We found increased levels of CD193+ and CD66b+ sputum eosinophils from COPD patients, and an elevated level of CD11b+ sputum eosinophils in asthma compared to COPD patients. The results of our study suggest that the profile of marker expression on COPD sputum eosinophils differed from other groups, suggesting a distinct phenotype of eosinophils of COPD patients than in asthma or healthy subjects.
Collapse
Affiliation(s)
- Katarzyna Mycroft
- Department of Internal Medicine, Pulmonary Diseases and Allergy, Medical University of Warsaw, 02-097 Warsaw, Poland
| | - Magdalena Paplińska-Goryca
- Department of Internal Medicine, Pulmonary Diseases and Allergy, Medical University of Warsaw, 02-097 Warsaw, Poland
| | - Małgorzata Proboszcz
- Department of Internal Medicine, Pulmonary Diseases and Allergy, Medical University of Warsaw, 02-097 Warsaw, Poland
| | - Patrycja Nejman-Gryz
- Department of Internal Medicine, Pulmonary Diseases and Allergy, Medical University of Warsaw, 02-097 Warsaw, Poland
| | - Rafał Krenke
- Department of Internal Medicine, Pulmonary Diseases and Allergy, Medical University of Warsaw, 02-097 Warsaw, Poland
| | - Katarzyna Górska
- Department of Internal Medicine, Pulmonary Diseases and Allergy, Medical University of Warsaw, 02-097 Warsaw, Poland
| |
Collapse
|
14
|
Salehi Z, Motlagh Ghoochani BFN, Hasani Nourian Y, Jamalkandi SA, Ghanei M. The controversial effect of smoking and nicotine in SARS-CoV-2 infection. ALLERGY, ASTHMA, AND CLINICAL IMMUNOLOGY : OFFICIAL JOURNAL OF THE CANADIAN SOCIETY OF ALLERGY AND CLINICAL IMMUNOLOGY 2023; 19:49. [PMID: 37264452 PMCID: PMC10234254 DOI: 10.1186/s13223-023-00797-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Accepted: 04/18/2023] [Indexed: 06/03/2023]
Abstract
The effects of nicotine and cigarette smoke in many diseases, notably COVID-19 infection, are being debated more frequently. The current basic data for COVID-19 is increasing and indicating the higher risk of COVID-19 infections in smokers due to the overexpression of corresponding host receptors to viral entry. However, current multi-national epidemiological reports indicate a lower incidence of COVID-19 disease in smokers. Current data indicates that smokers are more susceptible to some diseases and more protective of some other. Interestingly, nicotine is also reported to play a dual role, being both inflammatory and anti-inflammatory. In the present study, we tried to investigate the effect of pure nicotine on various cells involved in COVID-19 infection. We followed an organ-based systematic approach to decipher the effect of nicotine in damaged organs corresponding to COVID-19 pathogenesis (12 related diseases). Considering that the effects of nicotine and cigarette smoke are different from each other, it is necessary to be careful in generalizing the effects of nicotine and cigarette to each other in the conducted researches. The generalization and the undifferentiation of nicotine from smoke is a significant bias. Moreover, different doses of nicotine stimulate different effects (dose-dependent response). In addition to further assessing the role of nicotine in COVID-19 infection and any other cases, a clever assessment of underlying diseases should also be considered to achieve a guideline for health providers and a personalized approach to treatment.
Collapse
Affiliation(s)
- Zahra Salehi
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Chemical Injuries Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | | | - Yazdan Hasani Nourian
- Chemical Injuries Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Sadegh Azimzadeh Jamalkandi
- Chemical Injuries Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| | - Mostafa Ghanei
- Chemical Injuries Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
15
|
Zhang F, Qin S, Xia F, Mao C, Li L. Case report: Streptococcus pneumoniae pneumonia characterized by diffuse centrilobular nodules in both lungs. Front Med (Lausanne) 2023; 9:1007160. [PMID: 36703900 PMCID: PMC9871572 DOI: 10.3389/fmed.2022.1007160] [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: 07/30/2022] [Accepted: 12/20/2022] [Indexed: 01/12/2023] Open
Abstract
Background Streptococcus pneumoniae (S. pneumoniae) is the most common pathogen in community-acquired pneumonia (CAP) and takes the form of lobar pneumonia as typical computed tomography (CT) findings. Various patterns of radiological manifestation have also been reported in patients with S. pneumoniae pneumonia; however, the appearance of diffuse centrilobular nodules in both lungs is rarely reported. Case presentation We report the case of a patient with a history of chronic lymphocytic leukemia (CLL) for 9 years who presented with new-onset fever, cough, excess sputum, and shortness of breath for 1 week. He was given intravenous antibacterial (cephalosporin) treatment for 4 days, but his condition did not improve and dyspnea became more serious. The chest CT indicated diffuse centrilobular nodules in both lungs at admission. Patient's bronchoalveolar (BAL) fluid was sent for metagenomic next-generation sequencing, which only supported a diagnosis of S. pneumoniae infection. His condition improved gradually after antimicrobial treatment (moxifloxacin) and a follow-up CT showed that the diffuse centrilobular nodules in both lungs were absorbed completely. Conclusion This case highlights a rare CT presentation of S. pneumoniae pneumonia that should alert clinicians, so as to avoid taking unnecessary treatment measures.
Collapse
|
16
|
Park MJ, Won JH, Kim DK. Thrombin Induced Apoptosis through Calcium-Mediated Activation of Cytosolic Phospholipase A 2 in Intestinal Myofibroblasts. Biomol Ther (Seoul) 2023; 31:59-67. [PMID: 36052603 PMCID: PMC9810453 DOI: 10.4062/biomolther.2022.043] [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: 03/25/2022] [Revised: 06/06/2022] [Accepted: 07/06/2022] [Indexed: 01/13/2023] Open
Abstract
Thrombin is a serine protease that participates in a variety of biological signaling through protease-activated receptors. Intestinal myofibroblasts play central roles in maintaining intestinal homeostasis. In this study, we found that thrombin-induced apoptosis is mediated by the calcium-mediated activation of cytosolic phospholipase A2 in the CCD-18Co cell. Thrombin reduced cell viability by inducing apoptosis and proteinase-activated receptor-1 antagonist attenuated thrombin-induced cell death. Endogenous ceramide did not affect the cell viability itself, but a ceramide-mediated pathway was involved in thrombin-induced cell death. Thrombin increased intracellular calcium levels and cytosolic phospholipase A2 activity. The ceramide synthase inhibitor Fumonisin B1, intracellular calcium chelator BAPTA-AM, and cytosolic phospholipase A2 inhibitor AACOCF3 inhibited thrombin-induced cell death. Thrombin stimulated arachidonic acid release and reactive oxygen species generation, which was blocked by AACOCF3, BAPTA-AM, and the antioxidant reagent Trolox. Taken together, thrombin triggered apoptosis through calcium-mediated activation of cytosolic phospholipase A2 in intestinal myofibroblasts.
Collapse
Affiliation(s)
- Mi Ja Park
- Department of Environmental & Health Chemistry, College of Pharmacy, Chung-Ang University, Seoul 06974, Republic of Korea
| | - Jong Hoon Won
- Department of Environmental & Health Chemistry, College of Pharmacy, Chung-Ang University, Seoul 06974, Republic of Korea
| | - Dae Kyong Kim
- Department of Environmental & Health Chemistry, College of Pharmacy, Chung-Ang University, Seoul 06974, Republic of Korea,Corresponding Author E-mail: , Tel: +82-31-724-2611, Fax: +82-31-724-2612
| |
Collapse
|
17
|
Pei Y, Zhang J, Qu J, Rao Y, Li D, Gai X, Chen Y, Liang Y, Sun Y. Complement component 3 protects human bronchial epithelial cells from cigarette smoke-induced oxidative stress and prevents incessant apoptosis. Front Immunol 2022; 13:1035930. [PMID: 36605203 PMCID: PMC9807617 DOI: 10.3389/fimmu.2022.1035930] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Accepted: 12/06/2022] [Indexed: 12/24/2022] Open
Abstract
The complement component 3 (C3) is a pivotal element of the complement system and plays an important role in innate immunity. A previous study showed that intracellular C3 was upregulated in airway epithelial cells (AECs) from individuals with end-stage chronic obstructive pulmonary disease (COPD). Accumulating evidence has shown that cigarette smoke extract (CSE) induces oxidative stress and apoptosis in AECs. Therefore, we investigated whether C3 modulated cigarette smoke-induced oxidative stress and apoptosis in AECs and participated in the pathogenesis of COPD. We found increased C3 expression, together with increased oxidative stress and apoptosis, in a cigarette smoke-induced mouse model of COPD and in AECs from patients with COPD. Different concentrations of CSEinduced C3 expression in 16HBE cells in vitro. Interestingly, C3 knockdown (KD) exacerbated oxidative stress and apoptosis in 16HBE cells exposed to CSE. Furthermore, C3 exerted its pro-survival effects through JNK inhibition, while exogenous C3 partially rescued CSE-induced cell death and oxidative stress in C3 KD cells. These data indicate that locally produced C3 is an important pro-survival molecule in AECs under cigarette smoke exposure, revealing a potentially novel mechanism in the pathogenesis of COPD.
Collapse
Affiliation(s)
| | - Jing Zhang
- *Correspondence: Jing Zhang, ; Yongchang Sun,
| | | | | | | | | | | | | | | |
Collapse
|
18
|
Resistance training prevents damage to the mitochondrial function of the skeletal muscle of rats exposed to secondary cigarette smoke. Life Sci 2022; 309:121017. [PMID: 36183779 DOI: 10.1016/j.lfs.2022.121017] [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: 08/01/2022] [Revised: 09/17/2022] [Accepted: 09/26/2022] [Indexed: 11/30/2022]
Abstract
AIM To analyze the consumption of oxygen and to quantify the mitochondrial respiratory chain proteins (OXPHOS) in the gastrocnemius muscle of rats exposed to cigarette smoke and/or RT practitioners. MAIN METHODS Wistar rats were divided into groups: Control (C), Smoker (S), Exercise (E) and Exercise Smoker (ES). Groups F and ES were exposed to the smoke of 4 cigarettes for 30 min, 2× a day, 5× a week, for 16 weeks. Groups E and ES performed four climbs with progressive load, 1× per day, 5× per week, for 16 weeks. The gastrocnemius muscle was collected for analysis of OXPHOS content and oxygen consumption. Groups S (vs. C) and ES (vs. C and E) showed lower body weight gain when observing the evolution curve. KEY FINDINGS The S rats showed a reduction in the NDUFB8 proteins of complex 1, SDHB of complex 2, MTC01 of complex 4 and ATP5A of complex 5 (ATP Synthase) compared to Group C. Additionally, S rats also showed increased consumption of O2 in Basal, Leak, Complex I and I/II combined measures compared to the other groups, suggesting that the activity of the mitochondria of these animals increased in terms of coupling and uncoupling parameters. SIGNIFICANCE Our data suggest that exposure to cigarette smoke for 16 weeks is capable of causing impairment of mitochondrial function with reduced expression of respiratory chain proteins in skeletal muscle. However, the RT was effective in preventing impairment of mitochondrial function in the skeletal muscle of rats exposed to secondary cigarette smoke.
Collapse
|
19
|
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
|
20
|
Wu X, Hussain M, Syed SK, Saadullah M, Alqahtani AM, Alqahtani T, Aldahish A, Fatima M, Shaukat S, Hussain L, Jamil Q, Mukhtar I, Khan KUR, Zeng LH. Verapamil attenuates oxidative stress and inflammatory responses in cigarette smoke (CS)-induced murine models of acute lung injury and CSE-stimulated RAW 264.7 macrophages via inhibiting the NF-κB pathway. Biomed Pharmacother 2022; 149:112783. [PMID: 35299124 DOI: 10.1016/j.biopha.2022.112783] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 02/21/2022] [Accepted: 02/28/2022] [Indexed: 01/09/2023] Open
Abstract
Acute lung injury (ALI) and acute respiratory distress syndrome (ARDS), severe form of ALI, are characterized by overwhelming of lung inflammation, and no treatment is currently available to treat ALI/ARDS. Cigarette smoke (CS) is one of the prime causes to induce ALI/ARDS via oxidative stress. Despite extensive research, no appropriate therapy is currently available to treat ALI/ARDS. Hence, new potential approaches are needed to treat ALI/ARDS. Consequently, this project was designed to explore the protective effects of verapamil against CS-induced ALI by in vivo and in vitro method. In vivo data obtained from respiratory mechanics, pulmonary morphometric analyses and lung histopathology revealed that verapamil dose-dependently and strikingly decreased the lung weight coefficient, attenuated the albumin exudation into lungs, minimized the infiltration of macrophages and neutrophils into lungs, reduced the pro-inflammatory cytokines (tumour necrosis factor-α (TNF-α), interleukin-6 (IL-6) and keratinocyte chemoattractant (KC)) production, and improved the hypoxemia and lung histopathological changes. Similarly, verapamil also reduced the production of TNF-α, IL-6 and KC from cigarette smoke extract (CSE)-stimulated RAW 264.7 macrophage. Importantly, verapamil dose-dependently and remarkably suppressed the CS-induced oxidative stress via not only reducing the myeloperoxidase (MPO) activity of lungs, total oxidative stress (TOS) and malondialdehyde (MDA) content in the lungs and supernatant of RAW 264.7 macrophage but also improving total antioxidant capacity (TAC) and superoxide dismutase (SOD) production. Finally, verapamil strikingly decreased the NF-κB expression both in in vivo and in vitro models. Hence, verapamil has positive therapeutic effects against CS-induced ALI via suppressing uncontrolled inflammatory response, oxidative stress and NF-κB p65 signaling.
Collapse
Affiliation(s)
- Ximei Wu
- Department of Pharmacology, Zhejiang University City College, 51 Huzhou Street, Hangzhou 310015, China.
| | - Musaddique Hussain
- Department of Pharmacology, Faculty of Pharmacy, The Islamia University of Bahawalpur, Bahawalpur 63100, Pakistan.
| | - Shahzada Khurram Syed
- Department of Basic Medical Sciences, School of Health Sciences, University of Management and Technology Lahore, 54000, Pakistan
| | - Malik Saadullah
- Department of Pharmaceutical Chemistry, Government College University, Faisalabad 38000, Pakistan
| | - Ali M Alqahtani
- Department of Pharmacology, College of Pharmacy, King Khalid University, Abha 62529, Saudi Arabia
| | - Taha Alqahtani
- Department of Pharmacology, College of Pharmacy, King Khalid University, Abha 62529, Saudi Arabia
| | - Afaf Aldahish
- Department of Pharmacology, College of Pharmacy, King Khalid University, Abha 62529, Saudi Arabia
| | - Mobeen Fatima
- Department of Pharmacology, Faculty of Pharmacy, The Islamia University of Bahawalpur, Bahawalpur 63100, Pakistan
| | - Saira Shaukat
- Department of Pharmacology, Faculty of Pharmacy, The Islamia University of Bahawalpur, Bahawalpur 63100, Pakistan
| | - Liaqat Hussain
- Department of Pharmacology, Government College University, Faisalabad 38000, Pakistan
| | - Qurratulain Jamil
- Department of Pharmacy Practice, Faculty of Pharmacy, The Islamia University of Bahawalpur, Pakistan
| | - Imran Mukhtar
- Department of Pharmacology, Faculty of Pharmacy, The Islamia University of Bahawalpur, Bahawalpur 63100, Pakistan; Sir Sadiq Muhammad Khan Abassi post Graduate Medical College, The Islamia University of Bahawalpur, Pakistan
| | - Kashif-Ur-Rehman Khan
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, The Islamia University of Bahawalpur, Bahawalpur 63100, Pakistan
| | - Ling-Hui Zeng
- Department of Pharmacology, Zhejiang University City College, 51 Huzhou Street, Hangzhou 310015, China
| |
Collapse
|
21
|
Kim Y, Kim J, Mo Y, Park DE, Lee HS, Jung JW, Kang HR. Cigarette smoke extract contributes to the inception and aggravation of asthmatic inflammation by stimulating innate immunity. ALLERGY ASTHMA & RESPIRATORY DISEASE 2022. [DOI: 10.4168/aard.2022.10.3.145] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Affiliation(s)
- Yujin Kim
- Department of Translational Medicine, Seoul National University College of Medicine, Seoul, Korea
- Institute of Allergy and Clinical Immunology, Seoul National University Medical Research Center, Seoul, Korea
| | - Jeonghyeon Kim
- Department of Translational Medicine, Seoul National University College of Medicine, Seoul, Korea
- Institute of Allergy and Clinical Immunology, Seoul National University Medical Research Center, Seoul, Korea
| | - Yosep Mo
- Department of Translational Medicine, Seoul National University College of Medicine, Seoul, Korea
- Institute of Allergy and Clinical Immunology, Seoul National University Medical Research Center, Seoul, Korea
| | - Da Eun Park
- Institute of Allergy and Clinical Immunology, Seoul National University Medical Research Center, Seoul, Korea
| | - Hyun-Seung Lee
- Institute of Allergy and Clinical Immunology, Seoul National University Medical Research Center, Seoul, Korea
| | - Jae-Woo Jung
- Department of Internal Medicine, Chung-Ang University Hospital, Seoul, Korea
| | - Hye-Ryun Kang
- Department of Translational Medicine, Seoul National University College of Medicine, Seoul, Korea
- Institute of Allergy and Clinical Immunology, Seoul National University Medical Research Center, Seoul, Korea
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
| |
Collapse
|
22
|
Aloufi N, Alluli A, Eidelman DH, Baglole CJ. Aberrant Post-Transcriptional Regulation of Protein Expression in the Development of Chronic Obstructive Pulmonary Disease. Int J Mol Sci 2021; 22:ijms222111963. [PMID: 34769392 PMCID: PMC8584689 DOI: 10.3390/ijms222111963] [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] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 10/25/2021] [Accepted: 10/28/2021] [Indexed: 02/07/2023] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is an incurable and prevalent respiratory disorder that is characterized by chronic inflammation and emphysema. COPD is primarily caused by cigarette smoke (CS). CS alters numerous cellular processes, including the post-transcriptional regulation of mRNAs. The identification of RNA-binding proteins (RBPs), microRNAs (miRNAs), and long non-coding RNAs (lncRNAs) as main factors engaged in the regulation of RNA biology opens the door to understanding their role in coordinating physiological cellular processes. Dysregulation of post-transcriptional regulation by foreign particles in CS may lead to the development of diseases such as COPD. Here we review current knowledge about post-transcriptional events that may be involved in the pathogenesis of COPD.
Collapse
Affiliation(s)
- Noof Aloufi
- Department of Pathology, McGill University, Montreal, QC H3A 2B4, Canada; (N.A.); (A.A.)
- Department of Medical Laboratory Technology, Applied Medical Science, Taibah University, Universities Road, Medina P.O. Box 344, Saudi Arabia
| | - Aeshah Alluli
- Department of Pathology, McGill University, Montreal, QC H3A 2B4, Canada; (N.A.); (A.A.)
| | - David H. Eidelman
- Department of Medicine, McGill University, Montreal, QC H4A 3J1, Canada;
| | - Carolyn J. Baglole
- Department of Pathology, McGill University, Montreal, QC H3A 2B4, Canada; (N.A.); (A.A.)
- Department of Medicine, McGill University, Montreal, QC H4A 3J1, Canada;
- Department of Pharmacology and Therapeutics, McGill University, Montreal, QC H3G 1Y6, Canada
- Correspondence:
| |
Collapse
|
23
|
Feng Q, Yu YZ, Meng QH. Blocking tumor necrosis factor-α delays progression of chronic obstructive pulmonary disease in rats through inhibiting MAPK signaling pathway and activating SOCS3/TRAF1. Exp Ther Med 2021; 22:1311. [PMID: 34630665 PMCID: PMC8461615 DOI: 10.3892/etm.2021.10746] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 01/25/2021] [Indexed: 01/04/2023] Open
Abstract
The present study was conducted in order to study the detailed molecular mechanism of tumor necrosis factor (TNF)-α in chronic obstructive pulmonary disease (COPD). The rats were treated with cigarette smoke (CS) and lipopolysaccharide (LPS) to establish the COPD model. Next, the changes in lung injury in COPD rats with TNF-α knockdown was tested. Meanwhile, the regulation of TNF-α on MAPK pathway and its downstream molecules (SOCS3/TRAF1) was determined by western blotting. On this basis, the activation of MAPK and inhibition of SOCS3/TRAF1 was also examined. Subsequently, the lung function was tested with the plethysmograph, the cells of bronchoalveolar lavage fluid was counted and classified. Furthermore, lung tissue sections were stained with hematoxylin and eosin to verify whether the treatment of MAPK pathway and downstream molecules affected the effect of TNF-α knockdown on COPD. The present study showed that TNF-α knockdown could alleviate the decrease in the function and inflammatory injury of the lungs of rats with COPD. Western blot analysis verified that TNF-α knockdown could inhibit the activation of MAPK pathway and increase the expression of SOCS3/TRAF1. The following experimental results showed that the relief of lung injury caused by TNF-α knockdown could be deteriorated by activating MAPK pathway. It was also found that the symptom of COPD was decreased following transfection with sh-TNF-α but worsened by SOCS3/TRAF1 knockdown. Overall, TNF-α knockdown inhibited the activation of MAPK pathway and increased the expression of SOCS3/TRAF1, thus delaying the process of COPD.
Collapse
Affiliation(s)
- Qiong Feng
- Department of Pulmonary and Critical Care Medicine, The Affiliated Hospital of Jianghan University, Wuhan, Hubei 430015, P.R. China
| | - Yan-Zi Yu
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Jianghan University, Wuhan, Hubei 430015, P.R. China
| | - Qing-Hua Meng
- Department of Pulmonary and Critical Care Medicine, The Affiliated Hospital of Jianghan University, Wuhan, Hubei 430015, P.R. China
| |
Collapse
|
24
|
Shin YJ, Kim SH, Park CM, Kim HY, Kim IH, Yang MJ, Lee K, Kim MS. Exposure to cigarette smoke exacerbates polyhexamethylene guanidine-induced lung fibrosis in mice. J Toxicol Sci 2021; 46:487-497. [PMID: 34602533 DOI: 10.2131/jts.46.487] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Cigarette smoke (CS) is the leading cause of chronic pulmonary diseases, including lung cancer, chronic obstructive pulmonary disease, and pulmonary fibrosis. In this study, we aimed to investigate the effects of repeated CS exposure on polyhexamethylene guanidine (PHMG)-induced pulmonary fibrosis in mice. A single intratracheal instillation of 0.6 mg/kg PHMG enhanced the immune response of mice by increasing the number of total and specific inflammatory cell types in the bronchoalveolar lavage fluid. It induced histopathological changes such as granulomatous inflammation/fibrosis and macrophage infiltration in the lungs. These responses were upregulated upon exposure to a combination of PHMG and CS. In contrast, a 4-hr/day exposure to 300 mg/m3 CS alone for 2 weeks by nose-only inhalation resulted in minimal inflammation in the mouse lung. Furthermore, PHMG administration increased the expression of fibrogenic mediators, especially in the pulmonary tissues of the PHMG + CS group compared with that in the PHMG alone group. However, there was no upregulation in the expression of inflammatory cytokines following exposure to a combination of PHMG and CS. Our results demonstrate that repeated exposure to CS may promote the development of PHMG-induced pulmonary fibrosis.
Collapse
Affiliation(s)
- Young-Jun Shin
- Inhalation Toxicity Research Group, Korea Institute of Toxicology, Korea
| | - Sung-Hwan Kim
- Inhalation Toxicology Center for Airborne Risk Factors, Korea Institute of Toxicology, Korea
| | - Chul Min Park
- Inhalation Toxicity Research Group, Korea Institute of Toxicology, Korea
| | - Hyeon-Young Kim
- Inhalation Toxicology Center for Airborne Risk Factors, Korea Institute of Toxicology, Korea
| | - In-Hyeon Kim
- Inhalation Toxicology Center for Airborne Risk Factors, Korea Institute of Toxicology, Korea
| | - Mi-Jin Yang
- Pathology Research Group, Korea Institute of Toxicology, Korea
| | - Kyuhong Lee
- Inhalation Toxicology Center for Airborne Risk Factors, Korea Institute of Toxicology, Korea.,Department of Human and Environmental Toxicology, University of Science & Technology, Korea
| | - Min-Seok Kim
- Inhalation Toxicity Research Group, Korea Institute of Toxicology, Korea
| |
Collapse
|
25
|
Loading of Beclomethasone in Liposomes and Hyalurosomes Improved with Mucin as Effective Approach to Counteract the Oxidative Stress Generated by Cigarette Smoke Extract. NANOMATERIALS 2021; 11:nano11040850. [PMID: 33810420 PMCID: PMC8065961 DOI: 10.3390/nano11040850] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 03/22/2021] [Accepted: 03/23/2021] [Indexed: 01/01/2023]
Abstract
In this work beclomethasone dipropionate was loaded into liposomes and hyalurosomes modified with mucin to improve the ability of the payload to counteract the oxidative stress and involved damages caused by cigarette smoke in the airway. The vesicles were prepared by dispersing all components in the appropriate vehicle and sonicating them, thus avoiding the use of organic solvents. Unilamellar and bilamellar vesicles small in size (~117 nm), homogeneously dispersed (polydispersity index lower than 0.22) and negatively charged (~−11 mV), were obtained. Moreover, these vesicle dispersions were stable for five months at room temperature (~25 °C). In vitro studies performed using the Next Generation Impactor confirmed the suitability of the formulations to be nebulized as they were capable of reaching the last stages of the impactor that mimic the deeper airways, thus improving the deposition of beclomethasone in the target site. Further, biocompatibility studies performed by using 16HBE bronchial epithelial cells confirmed the high biocompatibility and safety of all the vesicles. Among the tested formulations, only mucin-hyalurosomes were capable of effectively counteracting the production of reactive oxygen species (ROS) induced by cigarette smoke extract, suggesting that this formulation may represent a promising tool to reduce the damaging effects of cigarette smoke in the lung tissues, thus reducing the pathogenesis of cigarette smoke-associated diseases such as chronic obstructive pulmonary disease, emphysema, and cancer.
Collapse
|
26
|
Application of text mining to develop AOP-based mucus hypersecretion genesets and confirmation with in vitro and clinical samples. Sci Rep 2021; 11:6091. [PMID: 33731770 PMCID: PMC7969622 DOI: 10.1038/s41598-021-85345-9] [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: 04/08/2020] [Accepted: 02/26/2021] [Indexed: 11/28/2022] Open
Abstract
Mucus hypersecretion contributes to lung function impairment observed in COPD (chronic obstructive pulmonary disease), a tobacco smoking-related disease. A detailed mucus hypersecretion adverse outcome pathway (AOP) has been constructed from literature reviews, experimental and clinical data, mapping key events (KEs) across biological organisational hierarchy leading to an adverse outcome. AOPs can guide the development of biomarkers that are potentially predictive of diseases and support the assessment frameworks of nicotine products including electronic cigarettes. Here, we describe a method employing manual literature curation supported by a focused automated text mining approach to identify genes involved in 5 KEs contributing to decreased lung function observed in tobacco-related COPD. KE genesets were subsequently confirmed by unsupervised clustering against 3 different transcriptomic datasets including (1) in vitro acute cigarette smoke and e-cigarette aerosol exposure, (2) in vitro repeated incubation with IL-13, and (3) lung biopsies from COPD and healthy patients. The 5 KE genesets were demonstrated to be predictive of cigarette smoke exposure and mucus hypersecretion in vitro, and less conclusively predict the COPD status of lung biopsies. In conclusion, using a focused automated text mining and curation approach with experimental and clinical data supports the development of risk assessment strategies utilising AOPs.
Collapse
|
27
|
Thirupathi A, Scarparo S, Silva PL, Marqueze LF, Vasconcelos FTF, Nagashima S, Cunha EBB, de Noronha L, Silveira PCL, Nesi RT, Gu Y, Pinho RA. Physical Exercise-Mediated Changes in Redox Profile Contribute to Muscle Remodeling After Passive Hand-Rolled Cornhusk Cigarette Smoke Exposure. Front Physiol 2020; 11:590962. [PMID: 33281621 PMCID: PMC7705113 DOI: 10.3389/fphys.2020.590962] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 10/29/2020] [Indexed: 12/17/2022] Open
Abstract
Consumption of non-traditional cigarettes has increased considerably worldwide, and they can induce skeletal muscle dysfunction. Physical exercise has been demonstrated to be important for prevention and treatment of smoking-related diseases. Therfore, the aim of this study was to investigate the effects of combined physical exercise (aerobic plus resistance exercise) on muscle histoarchitecture and oxidative stress in the animals exposed chronically to smoke from hand-rolled cornhusk cigarette (HRCC). Male Swiss mice were exposed to ambient air or passively to the smoke of 12 cigarettes over three daily sessions (four cigarettes per session) for 30 consecutive days with or without combined physical training. 48 h after the last training session, total leukocyte count was measured in bronchoalveolar lavage fluid (BALF), and the quadriceps were removed for histological/immunohistochemical analysis and measurement of oxidative stress parameters. The effects of HRCC on the number of leukocytes in BALF, muscle fiber diameter, central nuclei, and nuclear factor kappa B (NF-κB) were reverted after combined physical training. In addition, increased myogenic factor 5, tumor necrosis factor alpha (TNFα), reduced transforming growth factor beta (TGF-β), and nitrate levels were observed after physical training. However, the reduction in superoxide dismutase and glutathione/glutathione oxidized ratio induced by HRCC was not affected by the training program. These results suggest the important changes in the skeletal muscle brought about by HRCC-induced alteration in the muscle redox profile. In addition, combined physical exercise contributes to remodeling without disrupting muscle morphology.
Collapse
Affiliation(s)
| | - Silvia Scarparo
- Laboratory of Experimental Pathophysiology, Graduate Program in Health Sciences, Universidade do Extremo Sul Catarinense, Criciúma, Brazil
| | - Paulo L Silva
- Laboratory of Experimental Pathophysiology, Graduate Program in Health Sciences, Universidade do Extremo Sul Catarinense, Criciúma, Brazil
| | - Luis F Marqueze
- Laboratory of Exercise Biochemistry in Health, Graduate Program in Health Sciences, School of Medicine, Pontifícia Universidade Católica do Paraná, Curitiba, Brazil
| | - Franciane T F Vasconcelos
- Laboratory of Exercise Biochemistry in Health, Graduate Program in Health Sciences, School of Medicine, Pontifícia Universidade Católica do Paraná, Curitiba, Brazil
| | - Seigo Nagashima
- Laboratory of Experimental Pathology, Graduate Program in Health Sciences, School of Medicine, Pontifícia Universidade Católica do Paraná, Curitiba, Brazil
| | - Eduardo B B Cunha
- Laboratory of Exercise Biochemistry in Health, Graduate Program in Health Sciences, School of Medicine, Pontifícia Universidade Católica do Paraná, Curitiba, Brazil
| | - Lúcia de Noronha
- Laboratory of Experimental Pathology, Graduate Program in Health Sciences, School of Medicine, Pontifícia Universidade Católica do Paraná, Curitiba, Brazil
| | - Paulo C L Silveira
- Laboratory of Experimental Pathophysiology, Graduate Program in Health Sciences, Universidade do Extremo Sul Catarinense, Criciúma, Brazil
| | - Renata T Nesi
- Laboratory of Exercise Biochemistry in Health, Graduate Program in Health Sciences, School of Medicine, Pontifícia Universidade Católica do Paraná, Curitiba, Brazil
| | - Yaodong Gu
- Faculty of Sports Science, Ningbo University, Ningbo, China
| | - Ricardo A Pinho
- Laboratory of Exercise Biochemistry in Health, Graduate Program in Health Sciences, School of Medicine, Pontifícia Universidade Católica do Paraná, Curitiba, Brazil
| |
Collapse
|
28
|
Protective Effects of Quercetin on Livers from Mice Exposed to Long-Term Cigarette Smoke. BIOMED RESEARCH INTERNATIONAL 2020; 2020:2196207. [PMID: 33282940 PMCID: PMC7685793 DOI: 10.1155/2020/2196207] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 10/15/2020] [Accepted: 10/19/2020] [Indexed: 01/24/2023]
Abstract
Cigarette smoke is highly toxic, and it can promote increased production of reactive species and inflammatory response and leads to liver diseases. Quercetin is a flavonoid that displays antioxidant and anti-inflammatory activities in liver diseases. This study aimed at evaluating the protective effects of quercetin on livers from mice exposed to long-term cigarette smoke exposure. Male C57BL/6 mice were divided into five groups: control (CG), vehicle (VG), quercetin (QG), cigarette smoke (CSG), quercetin, and cigarette smoke (QCSG). CSG and QCSG were exposed to cigarette smoke for sixty consecutive days; at the end of the exposures, all animals were euthanized. Mice that received quercetin daily and were exposed to cigarette smoke showed a reduced influx of inflammatory cells, oxidative stress, inflammatory reaction, and histopathological changes in the liver, compared to CSG. These results suggest that quercetin may be an effective adjuvant for treating damage to the liver due to cigarette smoke exposure.
Collapse
|
29
|
Peterson LA, Balbo S, Fujioka N, Hatsukami DK, Hecht SS, Murphy SE, Stepanov I, Tretyakova NY, Turesky RJ, Villalta PW. Applying Tobacco, Environmental, and Dietary-Related Biomarkers to Understand Cancer Etiology and Evaluate Prevention Strategies. Cancer Epidemiol Biomarkers Prev 2020; 29:1904-1919. [PMID: 32051197 PMCID: PMC7423750 DOI: 10.1158/1055-9965.epi-19-1356] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 12/18/2019] [Accepted: 01/27/2020] [Indexed: 01/20/2023] Open
Abstract
Many human cancers are caused by environmental and lifestyle factors. Biomarkers of exposure and risk developed by our team have provided critical data on internal exposure to toxic and genotoxic chemicals and their connection to cancer in humans. This review highlights our research using biomarkers to identify key factors influencing cancer risk as well as their application to assess the effectiveness of exposure intervention and chemoprevention protocols. The use of these biomarkers to understand individual susceptibility to the harmful effects of tobacco products is a powerful example of the value of this type of research and has provided key data confirming the link between tobacco smoke exposure and cancer risk. Furthermore, this information has led to policy changes that have reduced tobacco use and consequently, the tobacco-related cancer burden. Recent technological advances in mass spectrometry led to the ability to detect DNA damage in human tissues as well as the development of adductomic approaches. These new methods allowed for the detection of DNA adducts in tissues from patients with cancer, providing key evidence that exposure to carcinogens leads to DNA damage in the target tissue. These advances will provide valuable insights into the etiologic causes of cancer that are not tobacco-related.See all articles in this CEBP Focus section, "Environmental Carcinogenesis: Pathways to Prevention."
Collapse
Affiliation(s)
- Lisa A Peterson
- Division of Environmental Health Sciences, University of Minnesota, Minneapolis, Minnesota.
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
| | - Silvia Balbo
- Division of Environmental Health Sciences, University of Minnesota, Minneapolis, Minnesota
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
| | - Naomi Fujioka
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
- Division of Hematology, Oncology and Transplantation, University of Minnesota, Minneapolis, Minnesota
| | - Dorothy K Hatsukami
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
- Department of Psychiatry, University of Minnesota, Minneapolis, Minnesota
| | - Stephen S Hecht
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, Minnesota
| | - Sharon E Murphy
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, Minnesota
| | - Irina Stepanov
- Division of Environmental Health Sciences, University of Minnesota, Minneapolis, Minnesota
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
| | - Natalia Y Tretyakova
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
- Department of Medicinal Chemistry, University of Minnesota, Minneapolis, Minnesota
| | - Robert J Turesky
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
- Department of Medicinal Chemistry, University of Minnesota, Minneapolis, Minnesota
| | - Peter W Villalta
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
| |
Collapse
|
30
|
Mustra Rakic J, Wang XD. Role of lycopene in smoke-promoted chronic obstructive pulmonary disease and lung carcinogenesis. Arch Biochem Biophys 2020; 689:108439. [PMID: 32504553 DOI: 10.1016/j.abb.2020.108439] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 05/20/2020] [Accepted: 05/27/2020] [Indexed: 12/30/2022]
Abstract
Chronic obstructive pulmonary disease (COPD) and lung cancer are a major cause of morbidity and mortality worldwide, with cigarette smoking being the single most important risk factor for both. Emerging evidence indicates alterations in reverse cholesterol transport-mediated removal of excess cholesterol from lung, and intracellular cholesterol overload to be involved in smoke-promoted COPD and lung cancer development. Since there are currently few effective treatments for COPD and lung cancer, it is important to identify food-derived, biologically active compounds, which can protect against COPD and lung cancer development. High intake of the carotenoid lycopene, as one of phytochemicals, is associated with a decreased risk of chronic lung lesions. This review article summarizes and discusses epidemiologic evidence, in vitro and in vivo studies regarding the prevention of smoke-promoted COPD and lung carcinogenesis through dietary lycopene as an effective intervention strategy. We focus on the recent research implying that lycopene preventive effect is through targeting the main genes involved in reverse cholesterol transport. This review also indicates gaps in knowledge about the function of lycopene against COPD and lung cancer, offering directions for further research.
Collapse
Affiliation(s)
- Jelena Mustra Rakic
- Nutrition and Cancer Biology Lab, Jean Mayer USDA-Human Nutrition Research Center on Aging (HNRCA) at Tufts University, Boston, MA, USA; Biochemical and Molecular Nutrition Program, Friedman School of Nutrition and Policy, Tufts University, Boston, MA, USA
| | - Xiang-Dong Wang
- Nutrition and Cancer Biology Lab, Jean Mayer USDA-Human Nutrition Research Center on Aging (HNRCA) at Tufts University, Boston, MA, USA; Biochemical and Molecular Nutrition Program, Friedman School of Nutrition and Policy, Tufts University, Boston, MA, USA.
| |
Collapse
|
31
|
Halappanavar S, van den Brule S, Nymark P, Gaté L, Seidel C, Valentino S, Zhernovkov V, Høgh Danielsen P, De Vizcaya A, Wolff H, Stöger T, Boyadziev A, Poulsen SS, Sørli JB, Vogel U. Adverse outcome pathways as a tool for the design of testing strategies to support the safety assessment of emerging advanced materials at the nanoscale. Part Fibre Toxicol 2020; 17:16. [PMID: 32450889 PMCID: PMC7249325 DOI: 10.1186/s12989-020-00344-4] [Citation(s) in RCA: 133] [Impact Index Per Article: 26.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Accepted: 04/02/2020] [Indexed: 12/11/2022] Open
Abstract
Toxicity testing and regulation of advanced materials at the nanoscale, i.e. nanosafety, is challenged by the growing number of nanomaterials and their property variants requiring assessment for potential human health impacts. The existing animal-reliant toxicity testing tools are onerous in terms of time and resources and are less and less in line with the international effort to reduce animal experiments. Thus, there is a need for faster, cheaper, sensitive and effective animal alternatives that are supported by mechanistic evidence. More importantly, there is an urgency for developing alternative testing strategies that help justify the strategic prioritization of testing or targeting the most apparent adverse outcomes, selection of specific endpoints and assays and identifying nanomaterials of high concern. The Adverse Outcome Pathway (AOP) framework is a systematic process that uses the available mechanistic information concerning a toxicological response and describes causal or mechanistic linkages between a molecular initiating event, a series of intermediate key events and the adverse outcome. The AOP framework provides pragmatic insights to promote the development of alternative testing strategies. This review will detail a brief overview of the AOP framework and its application to nanotoxicology, tools for developing AOPs and the role of toxicogenomics, and summarize various AOPs of relevance to inhalation toxicity of nanomaterials that are currently under various stages of development. The review also presents a network of AOPs derived from connecting all AOPs, which shows that several adverse outcomes induced by nanomaterials originate from a molecular initiating event that describes the interaction of nanomaterials with lung cells and involve similar intermediate key events. Finally, using the example of an established AOP for lung fibrosis, the review will discuss various in vitro tests available for assessing lung fibrosis and how the information can be used to support a tiered testing strategy for lung fibrosis. The AOPs and AOP network enable deeper understanding of mechanisms involved in inhalation toxicity of nanomaterials and provide a strategy for the development of alternative test methods for hazard and risk assessment of nanomaterials.
Collapse
Affiliation(s)
- Sabina Halappanavar
- Environmental Health Science and Research Bureau, Health Canada, Ottawa, Ontario, Canada.
| | - Sybille van den Brule
- Louvain centre for Toxicology and Applied Pharmacology, Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain, Brussels, Belgium
| | - Penny Nymark
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
- Department of Toxicology, Misvik Biology, Turku, Finland
| | - Laurent Gaté
- Institut National de Recherche et de Sécurité, Vandoeuvre-lès-Nancy, France
| | - Carole Seidel
- Institut National de Recherche et de Sécurité, Vandoeuvre-lès-Nancy, France
| | - Sarah Valentino
- Institut National de Recherche et de Sécurité, Vandoeuvre-lès-Nancy, France
| | - Vadim Zhernovkov
- Systems Biology Ireland, University College Dublin, Dublin 4, Ireland
| | | | - Andrea De Vizcaya
- Departamento de Toxicologia, CINVESTAV-IPN, Ciudad de México, Mexico
- Sabbatical leave at Environmental Health Science and Research Bureau, Health Canada, Ottawa, Canada
| | - Henrik Wolff
- Finnish Institute of Occupational Health, Helsinki, Finland
| | - Tobias Stöger
- Research Center for Environmental Health (GmbH), Neuherberg, Germany
- German Center for Lung Research (DZL), Giessen, Germany
- Institute of Lung Biology and Disease, Comprehensive Pneumology Center, Helmholtz Zentrum München - German, Oberschleißheim, Germany
| | - Andrey Boyadziev
- Environmental Health Science and Research Bureau, Health Canada, Ottawa, Ontario, Canada
| | - Sarah Søs Poulsen
- National Research Centre for the Working Environment, Copenhagen Ø, Denmark
| | | | - Ulla Vogel
- National Research Centre for the Working Environment, Copenhagen Ø, Denmark.
- DTU Health Tech, Technical University of Denmark, Kgs. Lyngby, Denmark.
| |
Collapse
|
32
|
Ashraf-Uz-Zaman M, Bhalerao A, Mikelis CM, Cucullo L, German NA. Assessing the Current State of Lung Cancer Chemoprevention: A Comprehensive Overview. Cancers (Basel) 2020; 12:E1265. [PMID: 32429547 PMCID: PMC7281533 DOI: 10.3390/cancers12051265] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 05/06/2020] [Accepted: 05/14/2020] [Indexed: 12/14/2022] Open
Abstract
Chemoprevention of lung cancer is thought to significantly reduce the risk of acquiring these conditions in the subpopulation of patients with underlying health issues, such as chronic obstructive pulmonary disorder and smoking-associated lung problems. Many strategies have been tested in the previous decades, with very few translating to successful clinical trials in specific subpopulations of patients. In this review, we analyze these strategies, as well as new approaches that have emerged throughout the last few years, including synthetic lethality concept and microbiome-induced regulation of lung carcinogenesis. Overall, the continuous effort in the area of lung chemoprevention is required to develop practical therapeutical approaches. Given the inconsistency of results obtained in clinical trials targeting lung cancer chemoprevention in various subgroups of patients that differ in the underlying health condition, race, and gender, we believe that individualized approaches will have more promise than generalized treatments.
Collapse
Affiliation(s)
- Md Ashraf-Uz-Zaman
- Department of Pharmaceutical Sciences, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA; (M.A.-U.-Z.); (A.B.); (C.M.M.); (L.C.)
| | - Aditya Bhalerao
- Department of Pharmaceutical Sciences, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA; (M.A.-U.-Z.); (A.B.); (C.M.M.); (L.C.)
| | - Constantinos M. Mikelis
- Department of Pharmaceutical Sciences, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA; (M.A.-U.-Z.); (A.B.); (C.M.M.); (L.C.)
- Center for Blood-Brain Barrier Research, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA
| | - Luca Cucullo
- Department of Pharmaceutical Sciences, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA; (M.A.-U.-Z.); (A.B.); (C.M.M.); (L.C.)
- Center for Blood-Brain Barrier Research, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA
| | - Nadezhda A. German
- Department of Pharmaceutical Sciences, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA; (M.A.-U.-Z.); (A.B.); (C.M.M.); (L.C.)
- Center for Blood-Brain Barrier Research, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA
- Center of Excellence for Translational Neuroscience and Therapeutics, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| |
Collapse
|
33
|
Nagler R, Zeineh N, Azrad M, Yassin N, Weizman A, Gavish M. 18-kDa Translocator Protein Ligands Protect H9C2 Cardiomyocytes from Cigarette Smoke-induced Cell Death: In Vitro Study. In Vivo 2020; 34:549-556. [PMID: 32111753 PMCID: PMC7157870 DOI: 10.21873/invivo.11807] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Revised: 12/30/2019] [Accepted: 01/02/2020] [Indexed: 12/14/2022]
Abstract
BACKGROUND Cigarette smoke (CS) can induce cellular damage via alterations in 18 kDa translocator protein (TSPO)-related functions, leading to cardiovascular diseases. The current study focused on the possible protective effect of TSPO ligands against CS-induced damage to cardiac cells. MATERIALS AND METHODS H9C2 Cardiomyocyte cell line of rat origin was pre-treated with TSPO ligands. Cell death, TSPO binding, and TSPO protein expression levels were assessed following 30-min CS exposure with/without TSPO ligands. RESULTS CS exposure of H9C2 cells significantly incensed cell death (by 26%, p<0.001). Pre-treatment with TSPO ligands at two concentrations prevented cell death. Neither CS nor ligands affected TSPO protein expression in H9C2 cells. CS led to increased cell death and reduced TSPO binding. CONCLUSION Reduced TSPO binding may have a role in CS-induced cell death, and TSPO ligand MGV-1 can prevent suppression of TSPO binding and corresponding cell death. These results may be relevant to treatment of cardiovascular diseases associated with CS.
Collapse
Affiliation(s)
- Rafael Nagler
- Department of Neuroscience, Faculty of Medicine, Rappaport Family Institute for Research in the Medical Sciences, Technion - Israel Institute of Technology, Haifa, Israel
| | - Nidal Zeineh
- Department of Neuroscience, Faculty of Medicine, Rappaport Family Institute for Research in the Medical Sciences, Technion - Israel Institute of Technology, Haifa, Israel
| | - Maya Azrad
- Department of Neuroscience, Faculty of Medicine, Rappaport Family Institute for Research in the Medical Sciences, Technion - Israel Institute of Technology, Haifa, Israel
| | - Nasra Yassin
- Department of Neuroscience, Faculty of Medicine, Rappaport Family Institute for Research in the Medical Sciences, Technion - Israel Institute of Technology, Haifa, Israel
| | - Abraham Weizman
- Research Unit at Geha Mental Health Center and Laboratory of Biological Psychiatry at Felsenstein Medical Research Center, Petah Tikva, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Moshe Gavish
- Department of Neuroscience, Faculty of Medicine, Rappaport Family Institute for Research in the Medical Sciences, Technion - Israel Institute of Technology, Haifa, Israel
| |
Collapse
|
34
|
Mukharjee S, Bank S, Maiti S. Chronic Tobacco Exposure by Smoking Develops Insulin Resistance. Endocr Metab Immune Disord Drug Targets 2020; 20:869-877. [PMID: 32065107 DOI: 10.2174/1871530320666200217123901] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Revised: 11/29/2019] [Accepted: 12/05/2019] [Indexed: 02/05/2023]
Abstract
BACKGROUND AND OBJECTIVES The present review critically discusses the high occurrence rate, insulin resistance and type-2 diabetes in tobacco exposed individuals. Tobacco extracts and smoke contain a large number of toxic materials and a significant number of those are metabolic disintegrators. DISCUSSION Glucose and lipid homeostasis is severely impaired by this compound. Tobacco exposure contributes to adverse effects by impairing the physical, biochemical and molecular mechanisms in the tissues. The immunological components are damaged by tobacco with high production of proinflammatory cytokines (IL-6, TNF-∞) and augmentation of inflammatory responses. These events result in damages to cytoskeletal structures of different tissues. Degradation of matrix structure (by activation of different types of MMPs) results in the permanent damages to the tissues and their metabolic functions. Cellular antioxidant defense system mostly cannot or hardly nullify CS-induced ROS production that activates polymorphonuclear neutrophils (PMNs), which are a major source of cytokines and chemokines (TNFα, IL6, IL8, INFγ). Additive effects of these immediately promote the low energy-metabolism as well as inflammation. Oxidative stress, mitochondrial dysfunction, and inflammation contribute to the direct nicotine toxicity via nAChRs in diabetes. The investigator identified that skeletal muscle insulin-resistance occurs in smokers due to phosphorylation of insulin receptor substrate1 (IRS1) at Ser-636 position. CONCLUSION Tobacco exposure initiates free radical related immunological impairment, DNA damage, and inflammation. So, the present analysis is of importance to figure out the mechanistic layout of tobacco-induced tissue damage and its possible therapeutic interventions.
Collapse
Affiliation(s)
- Suchismita Mukharjee
- Post Graduate Department of Biochemistry, Cell and Molecular Therapeutics Laboratory, Oriental Institute of Science and Technology, Rangamati, Midnapore, WB, 721102, India
| | - Sarbashri Bank
- Post Graduate Department of Biochemistry, Cell and Molecular Therapeutics Laboratory, Oriental Institute of Science and Technology, Rangamati, Midnapore, WB, 721102, India
| | - Smarajit Maiti
- Post Graduate Department of Biochemistry, Cell and Molecular Therapeutics Laboratory, Oriental Institute of Science and Technology, Rangamati, Midnapore, WB, 721102, India
| |
Collapse
|
35
|
Albacar N, Faner R, Sellarés J. Smoking and Interstitial lung damage/effects: A Plausible Association? Arch Bronconeumol 2019; 56:422-423. [PMID: 31753678 DOI: 10.1016/j.arbres.2019.09.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Revised: 09/13/2019] [Accepted: 09/25/2019] [Indexed: 10/25/2022]
Affiliation(s)
- Nuria Albacar
- Servei de Pneumologia, Respiratory Institute, Hospital Clínic, Universitat de Barcelona, Barcelona, España
| | - Rosa Faner
- Centro de Investigación Biomedica en Red-Enfermedades Respiratorias (CIBERES); Institut d'investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, España
| | - Jacobo Sellarés
- Servei de Pneumologia, Respiratory Institute, Hospital Clínic, Universitat de Barcelona, Barcelona, España; Centro de Investigación Biomedica en Red-Enfermedades Respiratorias (CIBERES); Institut d'investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, España.
| |
Collapse
|
36
|
Frobert O, Reitan C, Hatsukami DK, Pernow J, Omerovic E, Andell P. Smokeless tobacco, snus, at admission for percutaneous coronary intervention and future risk for cardiac events. Open Heart 2019; 6:e001109. [PMID: 31673392 PMCID: PMC6803000 DOI: 10.1136/openhrt-2019-001109] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Revised: 09/18/2019] [Accepted: 09/23/2019] [Indexed: 02/01/2023] Open
Abstract
Objective To assess the risk of future death and cardiac events following percutaneous coronary intervention (PCI) in patients using smokeless tobacco, snus, compared with patients not using snus at admission for a first PCI. Methods The Swedish Coronary Angiography and Angioplasty Registry is a prospective registry on coronary diagnostic procedures and interventions. A total of 74 958 patients admitted for a first PCI were enrolled between 2009 and 2018, 6790 snus users and 68 168 not using snus. We used Cox proportional hazards regression for statistical modelling on imputed datasets as well as complete-case datasets. Results Patients using snus were younger (mean (SD) age 61.0 (±10.2) years) than patients not using snus (67.6 (±11.1), p<0.001) and more often male (95.4% vs 67.4%, p<0.001). After multivariable adjustment, snus use was not associated with the primary composite outcome of all-cause mortality, new coronary revascularisation or new hospitalisation for heart failure at 1 year (HR 0.98, 95% CI 0.91 to 1.05). In patients using snus at baseline who underwent a second PCI (n=1443), the duration from the index intervention was shorter for subjects who continued using snus (n=921, 63.8%) compared with subjects who had stopped (mean number of days 285 vs 406, p value=0.001). Conclusions Snus use at admission for a first PCI was not associated with a higher occurrence of all-cause mortality, new revascularisation or heart failure hospitalisation. Discontinuing snus after a first PCI was associated with a significantly longer duration to a subsequent PCI.
Collapse
Affiliation(s)
- Ole Frobert
- Faculty of Health, Department of Cardiology, Örebro University, Örebro, Sweden
| | - Christian Reitan
- Faculty of Medicine, Department of Cardiology, Lund University, Lund, Sweden
| | - Dorothy K Hatsukami
- Department of Psychiatry, Tobacco Research Programs, University of Minnesota, Minneapolis, Minnesota, USA
| | - John Pernow
- Unit of Cardiology, Department of Medicine, Heart and Vascular Division, Karolinska Institutet, Stockholm, Sweden
| | - Elmir Omerovic
- Department of Cardiology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Pontus Andell
- Unit of Cardiology, Department of Medicine, Heart and Vascular Division, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
37
|
Modeling drug exposure in rodents using e-cigarettes and other electronic nicotine delivery systems. J Neurosci Methods 2019; 330:108458. [PMID: 31614162 DOI: 10.1016/j.jneumeth.2019.108458] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2019] [Revised: 10/05/2019] [Accepted: 10/06/2019] [Indexed: 12/25/2022]
Abstract
Smoking tobacco products is the leading cause of preventable death worldwide. Coordinated efforts have successfully reduced tobacco cigarette smoking in the United States; however, electronic cigarettes (e-cigarette) and other electronic nicotine delivery systems (ENDS) recently have replaced traditional cigarettes for many users. While the clinical risks associated with long-term ENDS use remain unclear, advancements in preclinical rodent models will enhance our understanding of their overall health effects. This review examines the peripheral and central effects of ENDS-mediated exposure to nicotine and other drugs of abuse in rodents and evaluates current techniques for implementing ENDS in preclinical research.
Collapse
|
38
|
Topic A, Milovanovic V, Lazic Z, Ivosevic A, Radojkovic D. Oxidized Alpha-1-Antitrypsin as a Potential Biomarker Associated with Onset and Severity of Chronic Obstructive Pulmonary Disease in Adult Population. COPD 2019; 15:472-478. [PMID: 30822244 DOI: 10.1080/15412555.2018.1541448] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Oxidative stress could reduce inhibitor activity of the alpha-1-antitrypsin (A1AT). Oxidative-modified A1AT (oxidized alpha-1-antitrypsin, OxyA1AT) significantly loses ability to protect the lungs from neutrophil elastase. We aimed to investigate OxyA1AT as a potential biomarker associated with onset and severity of chronic obstructive pulmonary disease (COPD) in adult population. The study included 65 patients with COPD (33 smokers and 32 no-smokers) and 46 healthy participants (17 smokers and 29 no-smokers). Determination of OxyA1AT in serum was based on the difference between the inhibitory activities of normal and oxidized A1AT against trypsin and elastase. The level of OxyA1AT was significantly increased in the group of COPD smokers compared to healthy no-smokers (p = 0.030) and COPD no-smokers (p = 0.009). The highest level of OxyA1AT was found in group of smokers with severe and very severe COPD in comparison to the following: no-smokers with the same stage of disease (p = 0.038), smokers with moderate COPD (p = 0.022), and the healthy control group, regardless of the smoking status (control no-smokers p = 0.001 and control smokers p = 0.034). In conclusion, serum level of OxyA1AT would be potentially good biomarker for the assessment of harmful effect of smoking to the onset and severity of COPD. Also, clinical significance of OxyA1AT as prognostic biomarker could be useful in assessing the effectiveness of antioxidant therapy for COPD and emphysema. Suitable and inexpensive laboratory method for determination of OxyA1AT is additional benefit for the introduction of OxyA1AT into routine clinical practice for diagnosis and monitoring of COPD.
Collapse
Affiliation(s)
- A Topic
- a Department of Medical Biochemistry , University of Belgrade-Faculty of Pharmacy , Belgrade , Serbia
| | - V Milovanovic
- a Department of Medical Biochemistry , University of Belgrade-Faculty of Pharmacy , Belgrade , Serbia
| | - Z Lazic
- b Faculty of Medical Sciences , University of Kragujevac , Kragujevac , Serbia
| | - A Ivosevic
- b Faculty of Medical Sciences , University of Kragujevac , Kragujevac , Serbia
| | - D Radojkovic
- c Institute of Molecular Genetics and Genetic Engineering, University of Belgrade , Belgrade , Serbia
| |
Collapse
|
39
|
Lebron IDSL, da Silva LF, Paletta JT, da Silva RA, Sant'Ana M, Costa SDS, Iyomasa-Pilon MM, Souza HR, Possebon L, Girol AP. Modulation of the endogenous Annexin A1 in a cigarette smoke cessation model: Potential therapeutic target in reversing the damage caused by smoking? Pathol Res Pract 2019; 215:152614. [PMID: 31500927 DOI: 10.1016/j.prp.2019.152614] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Revised: 08/18/2019] [Accepted: 08/23/2019] [Indexed: 10/26/2022]
Abstract
BACKGROUND Smoking cessation may help in the reversal of inflammation and damage caused by smoking. The endogenous annexin A1 (AnxA1) protein has anti-inflammatory effects which instigates the understanding of its role in the attenuation of inflammatory processes caused by smoking. MATERIAL AND METHODS Wistar rats were exposed to cigarette smoke for 8 weeks. After the exposure period, one of the groups remained other 8 weeks in the absence of smoke. Animals not exposed to smoke were used as control. Blood, trachea and lungs were obtained for histopathological, immunohistochemical and biochemical analyses. RESULTS Loss of cilia of the tracheal lining epithelium was found by smoke exposure, but smoking cessation led to recovery of the tracheal epithelium. Similarly, chronically exposed-to-smoke animals showed increased lymphocytes and macrophages in bronchoalveolar lavage and higher levels of glucose and gamma-GT in their blood. Reduction of lymphocytes, glucose and gamma-GT occurred after smoking cessation. In addition, IL-1β, IL-6, IL-10, TNF-α and MCP-1 levels were elevated by smoke exposure. Smoking cessation significantly reduced the levels of IL-1β, IL-6 and MCP-1 but increased the IL-10 concentration. Numerous mast cells and macrophages were observed in the lung of chronically exposed-to-smoke animals with reduction by smoking cigarette abstinence. AnxA1 increased expression and concomitant NF-κB reduction were found in the smoking cessation group. CONCLUSION Our results showed that cigarette abstinence promoted partial recovery of the inflammatory process. The attenuation of the inflammatory profile may be associated with the overexpression of AnxA1 protein.
Collapse
Affiliation(s)
| | | | | | | | | | - Sara de Souza Costa
- University Center Padre Albino (UNIFIPA), Catanduva, SP, Brazil; Department of Biology, Laboratory of Immunomorphology, São Paulo State University (UNESP), Institute of Biosciences, Humanities and Exact Sciences (IBILCE), São José do Rio Preto Campus, SP, Brazil.
| | | | - Helena Ribeiro Souza
- University Center Padre Albino (UNIFIPA), Catanduva, SP, Brazil; Department of Biology, Laboratory of Immunomorphology, São Paulo State University (UNESP), Institute of Biosciences, Humanities and Exact Sciences (IBILCE), São José do Rio Preto Campus, SP, Brazil.
| | - Lucas Possebon
- University Center Padre Albino (UNIFIPA), Catanduva, SP, Brazil; Department of Biology, Laboratory of Immunomorphology, São Paulo State University (UNESP), Institute of Biosciences, Humanities and Exact Sciences (IBILCE), São José do Rio Preto Campus, SP, Brazil.
| | - Ana Paula Girol
- University Center Padre Albino (UNIFIPA), Catanduva, SP, Brazil; Department of Biology, Laboratory of Immunomorphology, São Paulo State University (UNESP), Institute of Biosciences, Humanities and Exact Sciences (IBILCE), São José do Rio Preto Campus, SP, Brazil.
| |
Collapse
|
40
|
Ergosterol attenuates cigarette smoke extract-induced COPD by modulating inflammation, oxidative stress and apoptosis in vitro and in vivo. Clin Sci (Lond) 2019; 133:1523-1536. [PMID: 31270147 DOI: 10.1042/cs20190331] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Revised: 06/24/2019] [Accepted: 07/03/2019] [Indexed: 02/06/2023]
Abstract
Cigarette smoke (CS) is the major cause of chronic obstructive pulmonary disease (COPD). CS heightens inflammation, oxidative stress and apoptosis. Ergosterol is the main bioactive ingredient in Cordyceps sinensis (C. sinensis), a traditional medicinal herb for various diseases. The objective of this work was to investigate the effects of ergosterol on anti-inflammatory and antioxidative stress as well as anti-apoptosis in a cigarette smoke extract (CSE)-induced COPD model both in vitro and in vivo Our results demonstrate that CSE induced inflammatory and oxidative stress and apoptosis with the involvement of the Bcl-2 family proteins via the nuclear factor kappa B (NF-κB)/p65 pathway in both 16HBE cells and Balb/c mice. CSE induced epithelial cell death and increased the expression of nitric oxide (NO), interleukin-6 (IL-6), tumor necrosis factor α (TNF-α), malondialdehyde (MAD) and the apoptosis-related proteins cleaved caspase 3/7/9 and cleaved-poly-(ADP)-ribose polymerase (PARP) both in vitro and in vivo, whereas decreased the levels of superoxide dismutase (SOD) and catalase (CAT). Treatment of 16HBE cells and Balb/c mice with ergosterol inhibited CSE-induced inflammatory and oxidative stress and apoptosis by inhibiting the activation of NF-κB/p65. Ergosterol suppressed apoptosis by inhibiting the expression of the apoptosis-related proteins both in vitro and in vivo Moreover, the usage of QNZ (an inhibitor of NF-κB) also partly demonstrated that NF-κB/p65 pathway was involved in the ergosterol protective progress. These results show that ergosterol suppressed COPD inflammatory and oxidative stress and apoptosis through the NF-κB/p65 pathway, suggesting that ergosterol may be partially responsible for the therapeutic effects of cultured C. sinensis on COPD patients.
Collapse
|
41
|
Mustra Rakic J, Liu C, Veeramachaneni S, Wu D, Paul L, Chen CYO, Ausman LM, Wang XD. Lycopene Inhibits Smoke-Induced Chronic Obstructive Pulmonary Disease and Lung Carcinogenesis by Modulating Reverse Cholesterol Transport in Ferrets. Cancer Prev Res (Phila) 2019; 12:421-432. [PMID: 31177203 DOI: 10.1158/1940-6207.capr-19-0063] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Revised: 04/01/2019] [Accepted: 05/15/2019] [Indexed: 12/17/2022]
Abstract
Chronic obstructive pulmonary disease (COPD) and lung cancer share the same etiologic factor, cigarette smoking. Higher consumption of dietary lycopene has been associated with lower risks of COPD and lung cancer in smokers. We investigated whether lycopene feeding protects against COPD and lung cancer in ferrets, a nonrodent model that closely mimics cigarette smoke (CS)-induced chronic bronchitis, emphysema, and lung tumorigenesis in human. We also explored whether the protective effect of lycopene is associated with restoring reverse cholesterol transport (RCT), a key driver in persistent inflammation with CS exposure. Ferrets (4 groups, n = 12-16/group) were exposed to a combination of tobacco carcinogen (NNK) and CS with or without consuming lycopene at low and high doses (equivalent to ∼30 and ∼90 mg lycopene/day in human, respectively) for 22 weeks. Results showed that dietary lycopene at a high dose significantly inhibited NNK/CS-induced chronic bronchitis, emphysema, and preneoplastic lesions, including squamous metaplasia and atypical adenomatous hyperplasia, as compared with the NNK/CS alone (P < 0.05). Lycopene feeding also tended to decrease the lung neoplastic lesions. Furthermore, lycopene feeding significantly inhibited NNK/CS-induced accumulation of total cholesterol, and increased mRNA expression of critical genes related to the RCT (PPARα, LXRα, and ATP-binding cassette transporters ABCA1 and ABCG1) in the lungs, which were downregulated by the NNK/CS exposure. The present study has provided the first evidence linking a protective role of dietary lycopene against COPD and preneoplastic lesions to RCT-mediated cholesterol accumulation in lungs.
Collapse
Affiliation(s)
- Jelena Mustra Rakic
- Nutrition and Cancer Biology Lab, Jean Mayer USDA-Human Nutrition Research Center on Aging (HNRCA) at Tufts University, Boston, Massachusetts.,Biochemical and Molecular Nutrition Program, Friedman School of Nutrition Science and Policy, Tufts University, Boston, Massachusetts
| | - Chun Liu
- Nutrition and Cancer Biology Lab, Jean Mayer USDA-Human Nutrition Research Center on Aging (HNRCA) at Tufts University, Boston, Massachusetts
| | - Sudipta Veeramachaneni
- Nutrition and Cancer Biology Lab, Jean Mayer USDA-Human Nutrition Research Center on Aging (HNRCA) at Tufts University, Boston, Massachusetts
| | - Dayong Wu
- Biochemical and Molecular Nutrition Program, Friedman School of Nutrition Science and Policy, Tufts University, Boston, Massachusetts.,Nutritional Immunology Lab, JM USDA-HNRCA at Tufts University, Boston, Massachusetts
| | - Ligi Paul
- Biochemical and Molecular Nutrition Program, Friedman School of Nutrition Science and Policy, Tufts University, Boston, Massachusetts.,Department of Developmental, Molecular and Chemical Biology, Tufts University School of Medicine, Boston, Massachusetts
| | - C-Y Oliver Chen
- Biochemical and Molecular Nutrition Program, Friedman School of Nutrition Science and Policy, Tufts University, Boston, Massachusetts
| | - Lynne M Ausman
- Nutrition and Cancer Biology Lab, Jean Mayer USDA-Human Nutrition Research Center on Aging (HNRCA) at Tufts University, Boston, Massachusetts.,Biochemical and Molecular Nutrition Program, Friedman School of Nutrition Science and Policy, Tufts University, Boston, Massachusetts
| | - Xiang-Dong Wang
- Nutrition and Cancer Biology Lab, Jean Mayer USDA-Human Nutrition Research Center on Aging (HNRCA) at Tufts University, Boston, Massachusetts. .,Biochemical and Molecular Nutrition Program, Friedman School of Nutrition Science and Policy, Tufts University, Boston, Massachusetts
| |
Collapse
|
42
|
Hussain M, Xu C, Yao M, Zhang Q, Wu J, Wu X, Lu M, Tang L, Wu F, Wu X. CRTH2 antagonist, CT‑133, effectively alleviates cigarette smoke-induced acute lung injury. Life Sci 2019; 216:156-167. [PMID: 30468833 DOI: 10.1016/j.lfs.2018.11.039] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Revised: 11/16/2018] [Accepted: 11/18/2018] [Indexed: 01/01/2023]
Abstract
AIMS Acute lung injury (ALI) and acute respiratory distress syndrome (ARDS), characterized by overwhelming lung inflammation, are associated with high mortality. Cigarette smoke (CS) is one of the major causes of ALI/ARDS. Since high expression of prostaglandin (PG) D2 has been observed in CS-induced lung injury. Currently, no effective pharmacological therapies are available to treat ALI, and supportive therapies remain the mainstay of treatment. Therefore, we investigated the protective effect of CT‑133, a newly discovered selective CRTH2 antagonist, on CS-induced ALI in vivo and in vitro. MAIN METHODS CT‑133 (10 and 30 mg/kg), dexamethasone (1 mg/kg) and normal saline were intratracheally administrated 1 hr prior to whole-body CS-exposure for seven consecutive days to study the key characteristics of ALI. Subsequently, CSE (4%)- and PGD2-stimulated RAW 264.7 macrophages were used to evaluate the protective effect of CT‑133. KEY FINDINGS CT‑133 remarkably attenuated infiltration of inflammatory cells, neutrophils, and macrophages in the BALF, albumin contents, expression of IL‑1β, IL‑6, TNF‑α and KC, lung myeloperoxidase (MPO) activity and lung histopathological alterations caused by CS exposure in mice. Moreover, CT‑133 not only reversed the uncontrolled secretion of IL‑1β, IL-6, TNF‑α and KC from CSE- and PGD2-stimulated RAW 264.7 macrophages but also augmented IL-10 production in both in vivo and in vitro studies. Additionally, CT‑133 alleviated in vitro neutrophil migration chemoattracted by PGD2. SIGNIFICANCE Our results provide the first evidence that targeting CRTH2 could be a new potential therapeutic option to treat CS-induced ALI.
Collapse
Affiliation(s)
- Musaddique Hussain
- Department of Pharmacology, Zhejiang University, School of Medicine, Hangzhou City 310058, China
| | - Chengyun Xu
- Department of Pharmacology, Zhejiang University, School of Medicine, Hangzhou City 310058, China
| | - Minli Yao
- Department of Pharmacology, Zhejiang University, School of Medicine, Hangzhou City 310058, China
| | - Qin Zhang
- Department of Pharmacology, Zhejiang University, School of Medicine, Hangzhou City 310058, China
| | - Junsong Wu
- Department of Critical Care Medicine and Orthopedics, The First Affiliated Hospital, Zhejiang University, School of Medicine, Hangzhou City 310009, China
| | - Xiling Wu
- Department of Respiratory Medicine, the Affiliated Children Hospital, Zhejiang University School of Medicine, Hangzhou City 310052, China
| | - Meiping Lu
- Department of Respiratory Medicine, the Affiliated Children Hospital, Zhejiang University School of Medicine, Hangzhou City 310052, China
| | - Lanfang Tang
- Department of Respiratory Medicine, the Affiliated Children Hospital, Zhejiang University School of Medicine, Hangzhou City 310052, China
| | - Fugen Wu
- Department of Pediatrics, The First People's Hospital of Wenling City, Wenling City 317500, China.
| | - Ximei Wu
- Department of Pharmacology, Zhejiang University, School of Medicine, Hangzhou City 310058, China.
| |
Collapse
|
43
|
Possebon L, Costa SS, Souza HR, Azevedo LR, Sant'Ana M, Iyomasa-Pilon MM, Oliani SM, Girol AP. Mimetic peptide AC2-26 of annexin A1 as a potential therapeutic agent to treat COPD. Int Immunopharmacol 2018; 63:270-281. [DOI: 10.1016/j.intimp.2018.08.011] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Revised: 08/08/2018] [Accepted: 08/10/2018] [Indexed: 12/27/2022]
|
44
|
Pezzuto A, Stellato M, Catania G, Mazzara C, Tonini S, Caricato M, Crucitti P, Tonini G. Short-term benefit of smoking cessation along with glycopirronium on lung function and respiratory symptoms in mild COPD patients: a retrospective study. J Breath Res 2018; 12:046007. [PMID: 29967309 DOI: 10.1088/1752-7163/aad0a8] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
INTRODUCTION Tobacco smoke is the leading cause of chronic obstructive pulmonary disease (COPD). Smoking cessation can change the natural history of COPD, as we know from the GOLD guidelines. Little is known about the short-term clinical and functional effects of smoking cessation treatment combined with anti-muscarinic bronchodilators. OBJECTIVE To determine whether quitting smoking, obtained by smoking cessation treatment combined with the use of a new long-acting muscarinic antagonist bronchodilator (LAMA), can improve lung function tests and respiratory symptoms more than the use of LAMA alone. METHODS We evaluated, in a retrospective analysis, the functional and clinical data, collected in one year, of 120 patients who were current smokers affected by mild COPD and who quit smoking using smoking cessation treatment combined with glycopirronium. We compared them with a group of 80 patients with mild COPD undergoing the same treatment but who did not quit smoking. All patients underwent functional and clinical tests at baseline and at a third-month check. MEASUREMENTS AND MAIN RESULTS The two groups were homogeneous in terms of demographic data without significant differences. All patients used varenicline for smoking cessation. They all performed the following tests: a spirometry with detection of resistances, the 6 min walking test, haemogasanalysis, the exhaled CO test, the COPD assessment test (CAT) and finally the modified Medical Research Council test (mMRC). A significant improvement in the functional tests at the third-month check was found in both groups-quitters and non-quitters. However, a notable increase in the examined parameters was registered in the group of patients who quit smoking, in particular, we observed a significant increase at the third-month check of the parameter forced expiratory volume in 1 s (FEV1) of more than 200 ml with p < 0.001. A comparison between quitters and non-quitters revealed a major benefit derived from smoking cessation in terms of functional changes and symptom relief. In particular, not only FEV1 but also forced expiratory flow at 25%-75% of vital capacity (FEF 25-75) (p < 0.01) and CAT (p < 0.001) were found to be significantly improved in patients who quit than in patients who did not at the check time point. CONCLUSIONS Smoking cessation treatment obtained by varenicline was confirmed as a crucial therapeutic option, especially when combined with bronchodilator in mild COPD. Patients who quit smoking could already benefit from both treatments in the short term, improving lung function and respiratory symptoms and therefore improving their quality of life.
Collapse
Affiliation(s)
- Aldo Pezzuto
- Cardiovascular-pulmonary Department, Sant' Andrea Hospital-Sapienza University, Via di Grottarossa, 1035/39 , I-00189; Rome, Italy
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Strzelak A, Ratajczak A, Adamiec A, Feleszko W. Tobacco Smoke Induces and Alters Immune Responses in the Lung Triggering Inflammation, Allergy, Asthma and Other Lung Diseases: A Mechanistic Review. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2018; 15:E1033. [PMID: 29883409 PMCID: PMC5982072 DOI: 10.3390/ijerph15051033] [Citation(s) in RCA: 370] [Impact Index Per Article: 52.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Revised: 05/08/2018] [Accepted: 05/09/2018] [Indexed: 02/06/2023]
Abstract
Many studies have been undertaken to reveal how tobacco smoke skews immune responses contributing to the development of chronic obstructive pulmonary disease (COPD) and other lung diseases. Recently, environmental tobacco smoke (ETS) has been linked with asthma and allergic diseases in children. This review presents the most actual knowledge on exact molecular mechanisms responsible for the skewed inflammatory profile that aggravates inflammation, promotes infections, induces tissue damage, and may promote the development of allergy in individuals exposed to ETS. We demonstrate how the imbalance between oxidants and antioxidants resulting from exposure to tobacco smoke leads to oxidative stress, increased mucosal inflammation, and increased expression of inflammatory cytokines (such as interleukin (IL)-8, IL-6 and tumor necrosis factor α ([TNF]-α). Direct cellular effects of ETS on epithelial cells results in increased permeability, mucus overproduction, impaired mucociliary clearance, increased release of proinflammatory cytokines and chemokines, enhanced recruitment of macrophages and neutrophils and disturbed lymphocyte balance towards Th2. The plethora of presented phenomena fully justifies a restrictive policy aiming at limiting the domestic and public exposure to ETS.
Collapse
Affiliation(s)
- Agnieszka Strzelak
- Department of Pediatric Pulmonology and Allergy, Medical University of Warsaw, Zwirki i Wigury 61, 02-091 Warszawa, Poland.
| | - Aleksandra Ratajczak
- Department of Pediatric Pulmonology and Allergy, Medical University of Warsaw, Zwirki i Wigury 61, 02-091 Warszawa, Poland.
| | - Aleksander Adamiec
- Department of Pediatric Pulmonology and Allergy, Medical University of Warsaw, Zwirki i Wigury 61, 02-091 Warszawa, Poland.
| | - Wojciech Feleszko
- Department of Pediatric Pulmonology and Allergy, Medical University of Warsaw, Zwirki i Wigury 61, 02-091 Warszawa, Poland.
| |
Collapse
|
46
|
Herman M, Warkentin MT, Shepshelovich D, Latifovic L, Hung R, Liu G. Heritable Germline Variation and Lung Cancer Susceptibility: One Size Does not Fit All. J Thorac Oncol 2018; 13:601-602. [PMID: 29703536 DOI: 10.1016/j.jtho.2018.02.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Accepted: 02/27/2018] [Indexed: 11/16/2022]
Affiliation(s)
- Michael Herman
- Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| | | | | | | | - Rayjean Hung
- Mount Sinai Hospital-Lunenfeld Research Institute, Toronto, Ontario, Canada
| | - Geoffrey Liu
- Princess Margaret Cancer Centre, Toronto, Ontario, Canada.
| |
Collapse
|
47
|
López-Hernández Y, Rivas-Santiago CE, López JA, Mendoza-Almanza G, Hernandez-Pando R. Tuberculosis and cigarette smoke exposure: An update of in vitro and in vivo studies. Exp Lung Res 2018; 44:113-126. [PMID: 29565741 DOI: 10.1080/01902148.2018.1444824] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Tuberculosis (TB) has been declared the first cause of death by an infectious agent. Annually, 10.4 million people suffer active TB. Most infected individuals live in low-income countries, where social and economic conditions enhance the dissemination and progression of the disease. These countries have a high percentage of smokers. Thousands of studies have linked cigarette smoke (CS) with increased risk of many diseases, such as cancer and lung diseases. Numerous in vitro studies have been conducted to evaluate the general and specific toxic effects of CS in lung immune function. Smoke exposure increases the risk of TB development three-fold. However, until now, only few animal studies have been performed to analyze the association between smoke and TB. In the present work, we review in vitro and in vivo studies whose aim was to analyze the molecular basis of TB susceptibility caused by exposure to CS.
Collapse
Affiliation(s)
- Y López-Hernández
- a CONACyT, Unidad Academica de Ciencias Biologicas , Universidad Autónoma de Zacatecas , Zacatecas , Mexico
| | - C E Rivas-Santiago
- a CONACyT, Unidad Academica de Ciencias Biologicas , Universidad Autónoma de Zacatecas , Zacatecas , Mexico
| | - J A López
- b Laboratorio de MicroRNAs, Unidad Academica de Ciencias Biologicas , Universidad Autónoma de Zacatecas , Zacatecas , Mexico
| | - G Mendoza-Almanza
- a CONACyT, Unidad Academica de Ciencias Biologicas , Universidad Autónoma de Zacatecas , Zacatecas , Mexico
| | - R Hernandez-Pando
- c Departamento de Patologia, Unidad de Patologia Experimental , Instituto Nacional de Ciencias Medicas y Nutricion Salvador Zubiran , Mexico
| |
Collapse
|
48
|
Ryu IS, Kim J, Seo SY, Yang JH, Oh JH, Lee DK, Cho HW, Lee K, Yoon SS, Seo JW, Shim I, Choe ES. Repeated Administration of Cigarette Smoke Condensate Increases Glutamate Levels and Behavioral Sensitization. Front Behav Neurosci 2018; 12:47. [PMID: 29615877 PMCID: PMC5864865 DOI: 10.3389/fnbeh.2018.00047] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Accepted: 02/28/2018] [Indexed: 01/09/2023] Open
Abstract
Nicotine, a nicotinic acetylcholine receptor agonist, produces the reinforcing effects of tobacco dependence by potentiating dopaminergic and glutamatergic neurotransmission. Non-nicotine alkaloids in tobacco also contribute to dependence by activating the cholinergic system. However, glutamatergic neurotransmission in the dorsal striatum associated with behavioral changes in response to cigarette smoking has not been investigated. In this study, the authors investigated alterations in glutamate levels in the rat dorsal striatum related to behavioral alterations after repeated administration of cigarette smoke condensate (CSC) using the real-time glutamate biosensing and an open-field behavioral assessment. Repeated administration of CSC including 0.4 mg nicotine (1.0 mL/kg/day, subcutaneous) for 14 days significantly increased extracellular glutamate concentrations more than repeated nicotine administration. In parallel with the hyperactivation of glutamate levels, repeated administration of CSC-evoked prolonged hypersensitization of psychomotor activity, including locomotor and rearing activities. These findings suggest that the CSC-induced psychomotor activities are closely associated with the elevation of glutamate concentrations in the rat dorsal striatum.
Collapse
Affiliation(s)
- In Soo Ryu
- Department of Biological Sciences, Pusan National University, Busan, South Korea.,Research Center for Safety Pharmacology, Korea Institute of Toxicology, Daejeon, South Korea
| | - Jieun Kim
- Department of Biological Sciences, Pusan National University, Busan, South Korea
| | - Su Yeon Seo
- Department of Biological Sciences, Pusan National University, Busan, South Korea.,Fundamental Research Division, Korea Institute of Oriental Medicine, Daejeon, South Korea
| | - Ju Hwan Yang
- Department of Biological Sciences, Pusan National University, Busan, South Korea
| | - Jeong Hwan Oh
- College of Fisheries Sciences, National Institute of Fisheries (NIFS), Busan, South Korea
| | - Dong Kun Lee
- Department of Physiology, Institute of Health Sciences, Gyeongsang National University School of Medicine, Jinju, South Korea
| | - Hyun-Wook Cho
- Department of Biology, Sunchon National University, Sunchon, South Korea
| | - Kyuhong Lee
- Inhalation Toxicology Research Center, Korea Institute of Toxicology, Jeongeup, South Korea
| | - Seong Shoon Yoon
- Research Center for Safety Pharmacology, Korea Institute of Toxicology, Daejeon, South Korea
| | - Joung-Wook Seo
- Research Center for Safety Pharmacology, Korea Institute of Toxicology, Daejeon, South Korea
| | - Insop Shim
- Department of Science in Korean Medicine, Kyung Hee University, Seoul, South Korea
| | - Eun Sang Choe
- Department of Biological Sciences, Pusan National University, Busan, South Korea
| |
Collapse
|
49
|
Kim W, Lim D, Kim J. p-Coumaric Acid, a Major Active Compound of Bambusae Caulis in Taeniam, Suppresses Cigarette Smoke-Induced Pulmonary Inflammation. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2018; 46:407-421. [PMID: 29433391 DOI: 10.1142/s0192415x18500209] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
[Formula: see text]-coumaric acid ([Formula: see text]-CA) is a common compound found in medicinal herbs, including Bambusae Caulis in Taeniam (BC). It has been used to treat various diseases in China and Korea. Our previous study demonstrated that BC inhibits pulmonary and intestinal inflammation. In the present study, we used cigarette smoke (CS) to induce lung inflammation in vivo, and investigated the anti-inflammatory effects of [Formula: see text]-CA on CS-induced inflammatory mice model. Mice were treated with BC and [Formula: see text]-CA via oral injection 2[Formula: see text]h before CS exposure. The body weight and the inflammatory cells in the bronchoalveolar lavage fluid were measured. The levels of relative inflammatory factors were confirmed by enzyme-linked immunosorbent assay. The lung histological changes were examined by hematoxylin and eosin staining. Also, the protein level of nuclear factor-[Formula: see text]B (NF-[Formula: see text]B) was evaluated by Western blotting. Our results indicated that BC and [Formula: see text]-CA inhibited CS-induced lung inflammation by regulating pro-inflammatory productions such as cytokines, chemokine, protease and NF-[Formula: see text]B. Consequently, these data demonstrated that [Formula: see text]-CA inhibited pulmonary inflammation by suppressing NF-[Formula: see text]B activity, through which pro-inflammatory mediators were regulated. Therefore, [Formula: see text]-CA, which was shown to be a major component of BC, can be considered as a strong therapeutic candidate for treating pulmonary inflammatory diseases.
Collapse
Affiliation(s)
- Woogyeong Kim
- 1 Department of Korean Physiology, College of Pharmacy, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Dahae Lim
- 1 Department of Korean Physiology, College of Pharmacy, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Jinju Kim
- 1 Department of Korean Physiology, College of Pharmacy, Kyung Hee University, Seoul 02447, Republic of Korea
| |
Collapse
|
50
|
van 't Erve TJ, Lih FB, Kadiiska MB, Deterding LJ, Mason RP. Elevated plasma 8-iso-prostaglandin F 2α levels in human smokers originate primarily from enzymatic instead of non-enzymatic lipid peroxidation. Free Radic Biol Med 2018; 115:105-112. [PMID: 29162517 PMCID: PMC5767525 DOI: 10.1016/j.freeradbiomed.2017.11.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Revised: 10/13/2017] [Accepted: 11/09/2017] [Indexed: 11/18/2022]
Abstract
It is widely accepted that free radicals in tobacco smoke lead to oxidative stress and generate the popular lipid peroxidation biomarker 8-iso-prostaglandin F2α (8-iso-PGF2α). However, 8-iso-PGF2α can simultaneously be produced in vivo by the prostaglandin-endoperoxide synthases (PGHS) induced by inflammation. This inflammation-dependent mechanism has never been considered as a source of elevated 8-iso-PGF2α in tobacco smokers. The goal of this study is to quantify the distribution of chemical- and PGHS-dependent 8-iso-PGF2α formation in the plasma of tobacco smokers and non-smokers. The influences of gender and hormonal contraceptive use were accounted for. The distribution was determined by measuring the 8-iso-PGF2α/prostaglandin F2α (PGF2α) ratio. When comparing smokers (n = 28) against non-smokers (n = 30), there was a statistically significant increase in the 8-iso-PGF2α concentration. The source of this increased 8-iso-PGF2α was primarily from PGHS. When stratifying for gender, the increase in 8-iso-PGF2α in male smokers (n = 9) was primarily from PGHS. Interestingly, female smokers on hormonal contraceptives had increased 8-iso-PGF2α in both pathways, whereas those not on hormonal contraceptives did not have increased 8-iso-PGF2α. In conclusion, increased plasma 8-iso-PGF2α in tobacco smokers has complex origins, with PGHS-dependent formation as the primary source. Accounting for both pathways provides a definitive measurement of both oxidative stress and inflammation.
Collapse
Affiliation(s)
- Thomas J van 't Erve
- Immunity, Inflammation and Disease Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, 27709 NC, USA.
| | - Fred B Lih
- Epigenetic and Stem Cell Biology Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, 27709 NC, USA
| | - Maria B Kadiiska
- Immunity, Inflammation and Disease Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, 27709 NC, USA
| | - Leesa J Deterding
- Epigenetic and Stem Cell Biology Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, 27709 NC, USA
| | - Ronald P Mason
- Immunity, Inflammation and Disease Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, 27709 NC, USA
| |
Collapse
|