51
|
Alexander LEC, Bellinghausen AL, Eakin MN. What are the mechanisms underlying vaping-induced lung injury? J Clin Invest 2020; 130:2754-2756. [PMID: 32391805 PMCID: PMC7260008 DOI: 10.1172/jci138644] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Affiliation(s)
- Laura E. Crotty Alexander
- Pulmonary and Critical Care Section, VA San Diego Healthcare System, La Jolla, California, USA
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, UCSD, La Jolla, California, USA
| | - Amy L. Bellinghausen
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, UCSD, La Jolla, California, USA
| | - Michelle N. Eakin
- Division of Pulmonary and Critical Care, Department of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| |
Collapse
|
52
|
Sato K, Inoue S, Igarashi A, Tokairin Y, Yamauchi K, Kimura T, Nishiwaki M, Nemoto T, Nakano H, Sato M, Machida H, Yang S, Minegishi Y, Furuyama K, Watanabe M, Shibata Y. Effect of Iron Deficiency on a Murine Model of Smoke-induced Emphysema. Am J Respir Cell Mol Biol 2020; 62:588-597. [DOI: 10.1165/rcmb.2018-0239oc] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Affiliation(s)
- Kento Sato
- Department of Cardiology, Pulmonology, and Nephrology, Yamagata University Faculty of Medicine, Yamagata, Japan; and
| | - Sumito Inoue
- Department of Cardiology, Pulmonology, and Nephrology, Yamagata University Faculty of Medicine, Yamagata, Japan; and
| | - Akira Igarashi
- Department of Cardiology, Pulmonology, and Nephrology, Yamagata University Faculty of Medicine, Yamagata, Japan; and
| | - Yoshikane Tokairin
- Department of Cardiology, Pulmonology, and Nephrology, Yamagata University Faculty of Medicine, Yamagata, Japan; and
| | - Keiko Yamauchi
- Department of Cardiology, Pulmonology, and Nephrology, Yamagata University Faculty of Medicine, Yamagata, Japan; and
| | - Tomomi Kimura
- Department of Cardiology, Pulmonology, and Nephrology, Yamagata University Faculty of Medicine, Yamagata, Japan; and
| | - Michiko Nishiwaki
- Department of Cardiology, Pulmonology, and Nephrology, Yamagata University Faculty of Medicine, Yamagata, Japan; and
| | - Takako Nemoto
- Department of Cardiology, Pulmonology, and Nephrology, Yamagata University Faculty of Medicine, Yamagata, Japan; and
| | - Hiroshi Nakano
- Department of Cardiology, Pulmonology, and Nephrology, Yamagata University Faculty of Medicine, Yamagata, Japan; and
| | - Masamichi Sato
- Department of Cardiology, Pulmonology, and Nephrology, Yamagata University Faculty of Medicine, Yamagata, Japan; and
| | - Hiroyoshi Machida
- Department of Cardiology, Pulmonology, and Nephrology, Yamagata University Faculty of Medicine, Yamagata, Japan; and
| | - Sujeong Yang
- Department of Cardiology, Pulmonology, and Nephrology, Yamagata University Faculty of Medicine, Yamagata, Japan; and
| | - Yukihiro Minegishi
- Department of Cardiology, Pulmonology, and Nephrology, Yamagata University Faculty of Medicine, Yamagata, Japan; and
| | - Kodai Furuyama
- Department of Cardiology, Pulmonology, and Nephrology, Yamagata University Faculty of Medicine, Yamagata, Japan; and
| | - Masafumi Watanabe
- Department of Cardiology, Pulmonology, and Nephrology, Yamagata University Faculty of Medicine, Yamagata, Japan; and
| | - Yoko Shibata
- Department of Pulmonary Medicine, Fukushima Medical University, Fukushima, Japan
| |
Collapse
|
53
|
Passaro A, Peters S, Mok TSK, Attili I, Mitsudomi T, de Marinis F. Testing for COVID-19 in lung cancer patients. Ann Oncol 2020; 31:832-834. [PMID: 32278879 PMCID: PMC7144604 DOI: 10.1016/j.annonc.2020.04.002] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Accepted: 04/03/2020] [Indexed: 01/08/2023] Open
Affiliation(s)
- A Passaro
- Division of Thoracic Oncology, IEO, European Institute of Oncology IRCCS, Milan, Italy.
| | - S Peters
- Department of Oncology, Centre Hospitalier Universitaire Vaudois, Lausanne University, Lausanne, Switzerland
| | - T S K Mok
- Department of Clinical Oncology, State Key Laboratory of Translational Oncology, Chinese University of Hong Kong, Hong Kong, China
| | - I Attili
- Division of Thoracic Oncology, IEO, European Institute of Oncology IRCCS, Milan, Italy
| | - T Mitsudomi
- Thoracic Surgery, Kinki University Faculty of Medicine, Osaka-Sayama, Japan
| | - F de Marinis
- Division of Thoracic Oncology, IEO, European Institute of Oncology IRCCS, Milan, Italy
| |
Collapse
|
54
|
da Silva Araújo NP, de Matos NA, Leticia Antunes Mota S, Farias de Souza AB, Dantas Cangussú S, Cunha Alvim de Menezes R, Silva Bezerra F. Quercetin Attenuates Acute Lung Injury Caused by Cigarette Smoke Both In Vitro and In Vivo. COPD 2020; 17:205-214. [PMID: 32237913 DOI: 10.1080/15412555.2020.1749253] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Cigarette smoke is highly toxic and is a major risk factor for airway inflammation, oxidative stress, and decline in lung function-the starting points for chronic obstructive pulmonary disease. Quercetin is a potent dietary antioxidant that displays anti-inflammatory activities. The goal of this study was to evaluate the effects of quercetin on reducing the redox imbalance and inflammation induced by short-term cigarette smoke exposure. In vitro, 25 and 50 μM quercetin attenuated the effects of cigarette smoke extract (increased generation of reactive oxygen species and nitric oxide) on J774A.1 cells (macrophages). We further examined the effects of quercetin in vivo. Male C57Bl/6 mice that received 10 mg/kg/day of quercetin via orogastric gavage before exposure to five days of cigarette smoke demonstrated reduced levels of leukocyte, oxidative stress, histological pattern changes of pulmonary parenchyma, and lung function alterations compared to the group that did not receive quercetin. These results suggest that quercetin may be an effective adjuvant for treating the effects of cigarette smoke exposure.
Collapse
Affiliation(s)
- Natália Pereira da Silva Araújo
- Laboratory of Experimental Pathophysiology, Department of Biological Sciences and Center of Research in Biological Sciences, Federal University of Ouro Preto, Ouro Preto, Brazil
| | - Natália Alves de Matos
- Laboratory of Experimental Pathophysiology, Department of Biological Sciences and Center of Research in Biological Sciences, Federal University of Ouro Preto, Ouro Preto, Brazil
| | - Suianne Leticia Antunes Mota
- Laboratory of Parasitic Diseases, School of Medicine, Department of Biological Sciences and NUPEB, Federal University of Ouro Preto, Ouro Preto, Brazil
| | - Ana Beatriz Farias de Souza
- Laboratory of Experimental Pathophysiology, Department of Biological Sciences and Center of Research in Biological Sciences, Federal University of Ouro Preto, Ouro Preto, Brazil
| | - Sílvia Dantas Cangussú
- Laboratory of Experimental Pathophysiology, Department of Biological Sciences and Center of Research in Biological Sciences, Federal University of Ouro Preto, Ouro Preto, Brazil
| | - Rodrigo Cunha Alvim de Menezes
- Laboratory of Cardiovascular Physiology, Department of Biological Sciences and Center of Research in Biological Sciences, Federal University of Ouro Preto, Ouro Preto, Brazil
| | - Frank Silva Bezerra
- Laboratory of Experimental Pathophysiology, Department of Biological Sciences and Center of Research in Biological Sciences, Federal University of Ouro Preto, Ouro Preto, Brazil
| |
Collapse
|
55
|
Herr C, Tsitouras K, Niederstraßer J, Backes C, Beisswenger C, Dong L, Guillot L, Keller A, Bals R. Cigarette smoke and electronic cigarettes differentially activate bronchial epithelial cells. Respir Res 2020; 21:67. [PMID: 32164736 PMCID: PMC7068890 DOI: 10.1186/s12931-020-1317-2] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Accepted: 02/10/2020] [Indexed: 01/25/2023] Open
Abstract
BACKGROUND The use of electronic cigarettes (ECIGs) is increasing, but the impact of ECIG-vapor on cellular processes like inflammation or host defense are less understood. The aim of the present study was to compare the acute effects of traditional cigarettes (TCIGs) and ECIG-exposure on host defense, inflammation, and cellular activation of cell lines and primary differentiated human airway epithelial cells (pHBE). METHODS We exposed pHBEs and several cell lines to TCIG-smoke or ECIG-vapor. Epithelial host defense and barrier integrity were determined. The transcriptome of airway epithelial cells was compared by gene expression array analysis. Gene interaction networks were constructed and differential gene expression over all groups analyzed. The expression of several candidate genes was validated by qRT-PCR. RESULTS Bacterial killing, barrier integrity and the expression of antimicrobial peptides were not affected by ECIG-vapor compared to control samples. In contrast, TCIGs negatively affected host defense and reduced barrier integrity in a significant way. Furthermore ECIG-exposure significantly induced IL-8 secretion from Calu-3 cells but had no effect on NCI-H292 or primary cells. The gene expression based on array analysis distinguished TCIG-exposed cells from ECIG and room air-exposed samples. CONCLUSION The transcriptome patterns of host defense and inflammatory genes are significantly distinct between ECIG-exposed and TCIG-treated cells. The overall effects of ECIGs on epithelial cells are less in comparison to TCIG, and ECIG-vapor does not affect host defense. Nevertheless, although acute exposure to ECIG-vapor induces inflammation, and the expression of S100 proteins, long term in vivo data is needed to evaluate the chronic effects of ECIG use.
Collapse
Affiliation(s)
- Christian Herr
- Department of Internal Medicine V - Pulmonology, Allergology and Critical Care Medicine, Saarland University, D-66421, Homburg, Germany.
| | - Konstantinos Tsitouras
- Department of Internal Medicine V - Pulmonology, Allergology and Critical Care Medicine, Saarland University, D-66421, Homburg, Germany
| | - Julia Niederstraßer
- Department of Internal Medicine V - Pulmonology, Allergology and Critical Care Medicine, Saarland University, D-66421, Homburg, Germany
| | - Christina Backes
- Clinical Bioinformatics, Saarland University, University Hospital, 66123, Saarbrücken, Germany
| | - Christoph Beisswenger
- Department of Internal Medicine V - Pulmonology, Allergology and Critical Care Medicine, Saarland University, D-66421, Homburg, Germany
| | - Li Dong
- Department of Clinical Laboratory, Shanghai Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China
| | - Loïc Guillot
- Sorbonne Universités, UPMC Univ Paris 06, INSERM, Centre de Recherche Saint-Antoine (CRSA), 75012, Paris, France
| | - Andreas Keller
- Clinical Bioinformatics, Saarland University, University Hospital, 66123, Saarbrücken, Germany
| | - Robert Bals
- Department of Internal Medicine V - Pulmonology, Allergology and Critical Care Medicine, Saarland University, D-66421, Homburg, Germany
| |
Collapse
|
56
|
He X, Li T, Luo L, Zeng H, Chen Y, Cai S. PRMT6 mediates inflammation via activation of the NF-κB/p65 pathway on a cigarette smoke extract-induced murine emphysema model. Tob Induc Dis 2020; 18:8. [PMID: 32047419 PMCID: PMC7008391 DOI: 10.18332/tid/116413] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Revised: 12/30/2019] [Accepted: 01/09/2020] [Indexed: 01/09/2023] Open
Abstract
INTRODUCTION Smoke-driven lung inflammation is considered to be the major pathophysiology mechanism of Chronic Obstructive Pulmonary Disease (COPD)/emphysema. Protein arginine methyltransferase 6 (PRMT6) is a key epigenetic enzyme, which is related to protecting the tri-methylation of H3K4 (H3K4me3). We hypothesized that PTMT6 protects lung inflammation through the nuclear factor kappa B (NF-κB) pathway. METHODS Mice were injected with cigarette smoke extract (CSE) or PBS to establish a mice model, intratracheally instilled with overexpressed PRMT6 or negative control vector. Morphometry of lung slides and lung function were measured. We determined the protein expression of PRMT6 and its related histone targets, the activation of NF-κB pathway, the level of tumor necrosis factor α (TNFα) and interleukin-1β (IL-1β). RESULTS After PRMT6 overexpression, the morphometry indexes and lung function were improved. Also, the expression of H3K4me3 was decreased. Overexpressed PRMT6 could suppress CSE-induced NF-κB activation and pro-inflammation genes expression. CONCLUSIONS The overexpressed PRMT6 could serve as an inflammation inhibitor, potentially through blocking the NF-κB/p65 pathway in the murine emphysema model.
Collapse
Affiliation(s)
- Xue He
- Department of Pulmonary and Critical Care Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
- Research Unit of Respiratory Disease, Central South University, Changsha, China
- Diagnosis and Treatment Center of Respiratory Disease, Central South University, Changsha, China
| | - Tiao Li
- Department of Pulmonary and Critical Care Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
- Research Unit of Respiratory Disease, Central South University, Changsha, China
- Diagnosis and Treatment Center of Respiratory Disease, Central South University, Changsha, China
| | - Lijuan Luo
- Department of Pulmonary and Critical Care Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
- Research Unit of Respiratory Disease, Central South University, Changsha, China
- Diagnosis and Treatment Center of Respiratory Disease, Central South University, Changsha, China
| | - Huihui Zeng
- Department of Pulmonary and Critical Care Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
- Research Unit of Respiratory Disease, Central South University, Changsha, China
- Diagnosis and Treatment Center of Respiratory Disease, Central South University, Changsha, China
| | - Yan Chen
- Department of Pulmonary and Critical Care Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
- Research Unit of Respiratory Disease, Central South University, Changsha, China
- Diagnosis and Treatment Center of Respiratory Disease, Central South University, Changsha, China
| | - Shan Cai
- Department of Pulmonary and Critical Care Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
- Research Unit of Respiratory Disease, Central South University, Changsha, China
- Diagnosis and Treatment Center of Respiratory Disease, Central South University, Changsha, China
| |
Collapse
|
57
|
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
|
58
|
Corriden R, Moshensky A, Bojanowski CM, Meier A, Chien J, Nelson RK, Crotty Alexander LE. E-cigarette use increases susceptibility to bacterial infection by impairment of human neutrophil chemotaxis, phagocytosis, and NET formation. Am J Physiol Cell Physiol 2019; 318:C205-C214. [PMID: 31664858 DOI: 10.1152/ajpcell.00045.2019] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
E-cigarettes are portrayed as safer relative to conventional tobacco. However, burgeoning evidence suggests that E-cigarettes may adversely affect host defenses. However, the precise mechanisms by which E-cigarette vapor alters innate immune cell function have not been fully elucidated. We determined the effects of E-cigarette exposure on the function and responses to infectious challenge of the most abundant innate immune cell, the neutrophil, using isolated human neutrophils and a mouse model of gram-negative infection. Our results revealed that human neutrophils exposed to E-cigarette vapor had 4.2-fold reductions in chemotaxis toward the bacterial cell-well component f-Met-Leu-Phe (P < 0.001). F-actin polarization and membrane fluidity were also adversely affected by E-cigarette vapor exposure. E-cigarette-exposed human neutrophils exhibited a 48% reduction in production of reactive oxygen species (ROS; P < 0.001). Given the central role of ROS in neutrophil extracellular trap (NET) production, NET production was quantified, and E-cigarette vapor exposure was found to reduce NETosis by 3.5-fold (P < 0.01); formulations with and without nicotine containing propylene glycol exhibiting significant suppressive effects. However, noncanonical NETosis was unaffected. In addition, exposure to E-cigarette vapor lowered the rate of phagocytosis of bacterial bioparticles by 47% (P < 0.05). In our physiological mouse model of chronic E-cigarette exposure and sepsis, E-cigarette vapor inhalation led to reduced neutrophil migration in infected spaces and a higher burden of Pseudomonas. These findings provide evidence that E-cigarette use adversely impacts the innate immune system and may place E-cigarette users at higher risk for dysregulated inflammatory responses and invasive bacterial infections.
Collapse
Affiliation(s)
- Ross Corriden
- Department of Pharmacology, University of California San Diego, La Jolla, California
| | - Alexander Moshensky
- Pulmonary and Critical Care Section, Veterans Affairs San Diego Healthcare System, La Jolla, California.,Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of California San Diego, La Jolla, California
| | - Christine M Bojanowski
- Pulmonary and Critical Care Section, Veterans Affairs San Diego Healthcare System, La Jolla, California.,Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of California San Diego, La Jolla, California
| | - Angela Meier
- Department of Anesthesiology, Division of Critical Care, University of California, San Diego, La Jolla, California
| | - Jason Chien
- Department of Pharmacology, University of California San Diego, La Jolla, California.,Pulmonary and Critical Care Section, Veterans Affairs San Diego Healthcare System, La Jolla, California.,Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of California San Diego, La Jolla, California
| | - Ryan K Nelson
- Pulmonary and Critical Care Section, Veterans Affairs San Diego Healthcare System, La Jolla, California.,Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of California San Diego, La Jolla, California
| | - Laura E Crotty Alexander
- Pulmonary and Critical Care Section, Veterans Affairs San Diego Healthcare System, La Jolla, California.,Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of California San Diego, La Jolla, California
| |
Collapse
|
59
|
Sayyed K, Aljebeai AK, Al-Nachar M, Chamieh H, Taha S, Abdel-Razzak Z. Interaction of cigarette smoke condensate and some of its components with chlorpromazine toxicity on Saccharomyces cerevisiae. Drug Chem Toxicol 2019; 45:77-87. [PMID: 31514548 DOI: 10.1080/01480545.2019.1659809] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Chlorpromazine (CPZ) is an antipsychotic phenothiazine which is still commonly prescribed though it causes idiosyncratic toxicity such as cholestasis. CPZ toxicity mechanisms involve oxidative stress among others. Cigarette smoke (CS) causes deleterious effects through diverse mechanisms such as oxidative stress. CS alters drug metabolizing enzymes expression and drug transporters expression and activity in animal cell models as well as in Saccharomyces cerevisiae. CS therefore alters pharmacokinetic and pharmacodynamics of many drugs including CPZ and caffeine whose toxicity is promoted by CS condensate (CSC). CSC interaction with CPZ toxicity deserves investigation. In this study, CSC exerted mild toxicity on Saccharomyces cerevisiae which resisted to this chemical stress after several hours. CPZ toxicity on yeast was dose-dependent and the cells resisted to CPZ up to 40 µM after 24 h of treatment. Yeast cells treated simultaneously with CPZ and a nontoxic CSC dose were less sensitive to CPZ. CSC probably triggers cross-resistance to CPZ. Using Sod1 mutant strain, we showed that this gene is potentially involved in the potential cross-resistance. Other genes encoding stress-related transcription factors could be involved in this process. Nicotine and cadmium chloride, which caused a dose-dependent toxicity individually, acted with CPZ in an additive or synergistic manner in terms of toxicity. Although our results cannot be extrapolated to humans, they clearly show that CSC and its components interact with CPZ toxicity.
Collapse
Affiliation(s)
- Katia Sayyed
- EDST-AZM-center-LBA3B - Tripoli and Faculty of Sciences, Lebanese University , Beirut , Lebanon
| | - Abdel-Karim Aljebeai
- EDST-AZM-center-LBA3B - Tripoli and Faculty of Sciences, Lebanese University , Beirut , Lebanon
| | - Mariam Al-Nachar
- EDST-AZM-center-LBA3B - Tripoli and Faculty of Sciences, Lebanese University , Beirut , Lebanon
| | - Hala Chamieh
- EDST-AZM-center-LBA3B - Tripoli and Faculty of Sciences, Lebanese University , Beirut , Lebanon
| | - Samir Taha
- EDST-AZM-center-LBA3B - Tripoli and Faculty of Sciences, Lebanese University , Beirut , Lebanon
| | - Ziad Abdel-Razzak
- EDST-AZM-center-LBA3B - Tripoli and Faculty of Sciences, Lebanese University , Beirut , Lebanon
| |
Collapse
|
60
|
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
|
61
|
Fallacara A, Busato L, Pozzoli M, Ghadiri M, Ong HX, Young PM, Manfredini S, Traini D. Co-Spray-Dried Urea Cross-Linked Hyaluronic Acid and Sodium Ascorbyl Phosphate as Novel Inhalable Dry Powder Formulation. J Pharm Sci 2019; 108:2964-2971. [DOI: 10.1016/j.xphs.2019.04.015] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Revised: 03/14/2019] [Accepted: 04/10/2019] [Indexed: 12/18/2022]
|
62
|
Hamer M, O'Donovan G, Stamatakis E. Lifestyle risk factors, obesity and infectious disease mortality in the general population: Linkage study of 97,844 adults from England and Scotland. Prev Med 2019; 123:65-70. [PMID: 30844499 DOI: 10.1016/j.ypmed.2019.03.002] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 02/26/2019] [Accepted: 03/02/2019] [Indexed: 11/25/2022]
Abstract
We examined associations between lifestyle variables and infectious disease mortality in a large general population cohort. A sample of 97,844 men and women (aged 47.1 ± 17.7 yrs.; 46.6% male) recruited from general population, household-based surveys were followed up over mean [SD] 9.4 ± 4.5 years. Exposure measurements included self-reported physical activity, cigarette smoking, alcohol intake, and objective body mass index and waist to hip ratio. There were 9027 deaths, of which 14.1% were attributed to infectious diseases. Compared to physically inactive participants both insufficiently active (Hazard ratio = 0.61; 95% CI, 0.50, 0.75) and sufficiently active (at least 150 min/wk. moderate - vigorous activity) (0.60; 0.45, 0.78) was associated with reduced risk of infectious disease mortality in models mutually adjusted for other lifestyle factors. Ex-smokers and current smokers were at increased risk of infectious disease mortality compared with never smoker, with the strongest associations being observed for heavy smoking (>20 cigarettes/day) and pneumonia (3.30; 2.35, 4.63). Underweight was associated with increased risk of infectious disease mortality (3.65; 2.64, 5.06) compared with normal weight; the risk of viral infection was lower in overweight (0.56; 0.44, 0.72) and obesity (0.39; 0.26, 0.58). Central obesity was, however, related to higher risk of bacterial infections, but only in normal weight centrally obese participants (1.71; 1.10, 2.64). A physically active lifestyle and lifelong absence from cigarette smoking had protective associations against infectious disease mortality. Obesity has divergent associations dependent on peripheral and visceral fat depots, and the specific outcome.
Collapse
Affiliation(s)
- Mark Hamer
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough LE11 3TU, United Kingdom; Department of Epidemiology and Public Health, University College London, London WC1E 6BT, United Kingdom.
| | - Gary O'Donovan
- School of Medicine, Universidad de los Andes, Bogotá, Colombia
| | - Emmanuel Stamatakis
- Department of Epidemiology and Public Health, University College London, London WC1E 6BT, United Kingdom; Charles Perkins Centre Epidemiology Unit, University of Sydney, Sydney, Australia; Prevention Research Collaboration, School of Public Health, University of Sydney, Sydney, Australia
| |
Collapse
|
63
|
Moore PJ, Sesma J, Alexis NE, Tarran R. Tobacco exposure inhibits SPLUNC1-dependent antimicrobial activity. Respir Res 2019; 20:94. [PMID: 31113421 PMCID: PMC6530064 DOI: 10.1186/s12931-019-1066-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2019] [Accepted: 05/06/2019] [Indexed: 11/24/2022] Open
Abstract
Background Tobacco smoke exposure impairs the lung’s innate immune response, leading to an increased risk of chronic infections. SPLUNC1 is a secreted, multifunctional innate defense protein that has antimicrobial activity against Gram negative organisms. We hypothesize that tobacco smoke-induced SPLUNC1 dysfunction contributes to the observed defect in innate immunity in tobacco smokers and that this dysfunction can be used as a potential biomarker of harm. Methods We collected sputum from never-smokers and otherwise healthy smokers. We performed Western blotting to determine SPLUNC1 levels and determined antimicrobial activity against nontypeable Haemophilus influenzae. An in vitro exposure model was utilized to measure the effect of tobacco exposure on human bronchial epithelial culture (HBEC) antimicrobial activity against H. influenzae. The direct effects of cigarette and little cigar smoke exposure on SPLUNC1 function was determined using 24 h growth measurements and LPS binding assays. Results H. influenzae growth in cigarette smoker’s sputum was significantly greater compared to never-smokers sputum over 24 h. HBEC supernatants and lysates contained significantly higher numbers of H. influenzae following chronic cigarette and little cigar smoke exposure compared to air-exposed controls. Furthermore, SPLUNC1’s antimicrobial activity and LPS-binding capability against both H. influenzae and P. aeruginosa was attenuated following cigarette and little cigar exposure. Conclusions These data suggest that cigarette and little cigar exposure impairs SPLUNC1’s antimicrobial ability and that this inhibition may serve as a novel biomarker of harm that can be used to assess the toxicity of commercial tobacco products.
Collapse
Affiliation(s)
- Patrick J Moore
- Marsico Lung Institute, University of North Carolina at Chapel Hill, 7118A Marsico Hall, 125 Mason Farm Road, Chapel Hill, NC, 27599, USA.
| | - Juliana Sesma
- CONICET, Facultad de Ciencias Médicas, Universidad Nacional de Rosario, Rosario, Argentina
| | - Neil E Alexis
- Center for Environmental Medicine, Asthma and Lung Biology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Robert Tarran
- Marsico Lung Institute, University of North Carolina at Chapel Hill, 7118A Marsico Hall, 125 Mason Farm Road, Chapel Hill, NC, 27599, USA.,Department of Cell Biology & Physiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| |
Collapse
|
64
|
Jubinville É, Routhier J, Maranda-Robitaille M, Pineault M, Milad N, Talbot M, Beaulieu MJ, Aubin S, Paré MÈ, Laplante M, Morissette MC. Pharmacological activation of liver X receptor during cigarette smoke exposure adversely affects alveolar macrophages and pulmonary surfactant homeostasis. Am J Physiol Lung Cell Mol Physiol 2019; 316:L669-L678. [PMID: 30702343 DOI: 10.1152/ajplung.00482.2018] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Smoking alters pulmonary reverse lipid transport and leads to intracellular lipid accumulation in alveolar macrophages. We investigated whether stimulating reverse lipid transport with an agonist of the liver X receptor (LXR) would help alveolar macrophages limit lipid accumulation and dampen lung inflammation in response to cigarette smoke. Mice were exposed to cigarette smoke and treated intraperitoneally with the LXR agonist T0901317. Expression of lipid capture and lipid export genes was assessed in lung tissue and alveolar macrophages. Pulmonary inflammation was assessed in the bronchoalveolar lavage (BAL). Finally, cholesterol efflux capacity and pulmonary surfactant levels were determined. In room air-exposed mice, T0901317 increased the expression of lipid export genes in macrophages and the whole lung and increased cholesterol efflux capacity without inducing inflammation or affecting the pulmonary surfactant. However, cigarette smoke-exposed mice treated with T0901317 showed a marked increase in BAL neutrophils, IL-1α, C-C motif chemokine ligand 2, and granulocyte-colony-stimulating factor levels. T0901317 treatment in cigarette smoke-exposed mice failed to increase the ability of alveolar macrophages to export cholesterol and markedly exacerbated IL-1α release. Finally, T0901317 led to pulmonary surfactant depletion only in cigarette smoke-exposed mice. This study shows that hyperactivation of LXR and the associated lipid capture/export mechanisms only have minor pulmonary effects on the normal lung. However, in the context of cigarette smoke exposure, where the pulmonary surfactant is constantly oxidized, hyperactivation of LXR has dramatic adverse effects, once again showing the central role of lipid homeostasis in the pulmonary response to cigarette smoke exposure.
Collapse
Affiliation(s)
- Éric Jubinville
- Faculty of Medicine, Université Laval , Quebec City, Quebec , Canada
- Quebec Heart and Lung Institute, Université Laval , Quebec City, Quebec , Canada
| | - Joanie Routhier
- Faculty of Medicine, Université Laval , Quebec City, Quebec , Canada
- Quebec Heart and Lung Institute, Université Laval , Quebec City, Quebec , Canada
| | | | - Marie Pineault
- Faculty of Medicine, Université Laval , Quebec City, Quebec , Canada
- Quebec Heart and Lung Institute, Université Laval , Quebec City, Quebec , Canada
| | - Nadia Milad
- Faculty of Medicine, Université Laval , Quebec City, Quebec , Canada
- Quebec Heart and Lung Institute, Université Laval , Quebec City, Quebec , Canada
| | - Maude Talbot
- Faculty of Medicine, Université Laval , Quebec City, Quebec , Canada
- Quebec Heart and Lung Institute, Université Laval , Quebec City, Quebec , Canada
| | - Marie-Josée Beaulieu
- Quebec Heart and Lung Institute, Université Laval , Quebec City, Quebec , Canada
| | - Sophie Aubin
- Quebec Heart and Lung Institute, Université Laval , Quebec City, Quebec , Canada
| | - Marie-Ève Paré
- Quebec Heart and Lung Institute, Université Laval , Quebec City, Quebec , Canada
| | - Mathieu Laplante
- Quebec Heart and Lung Institute, Université Laval , Quebec City, Quebec , Canada
- Department of Medicine, Université Laval , Quebec City, Quebec , Canada
- Centre de Recherche sur le Cancer de l'Université Laval, Quebec City, Quebec, Canada
| | - Mathieu C Morissette
- Quebec Heart and Lung Institute, Université Laval , Quebec City, Quebec , Canada
- Department of Medicine, Université Laval , Quebec City, Quebec , Canada
| |
Collapse
|
65
|
Fireman Klein E, Adir Y, Krencel A, Peri R, Vasserman B, Fireman E, Kessel A. Ultrafine particles in airways: a novel marker of COPD exacerbation risk and inflammatory status. Int J Chron Obstruct Pulmon Dis 2019; 14:557-564. [PMID: 30880945 PMCID: PMC6402613 DOI: 10.2147/copd.s187560] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Purpose Ultrafine particles (UFP) are toxic due to their small size and penetration into deeper lung compartments. We aimed to evaluate the exhaled breath condensate (EBC)-UFP content as a reflection of inflammation and oxidative stress status in COPD patients and as an exacerbation risk marker. Methods EBC was collected by conventional methods. Particles were analyzed with NanoSight LM20. EBC carbonyl and 8-hydroxydeoxyguanosine (8-OHdG) levels were measured using ELISA kits. Study population (58 COPD patients and 40 healthy smoker and non-smoker controls) underwent spirometry, diffusion capacity, EBC testing, and blood sampling. Results Absolute eosinophil count, C-reactive protein (CRP), and lactate dehydrogenase in serum were elevated in the COPD group compared with the controls (224 U/L, 5 mg/L, and 391 U/L vs 154 U/L, 3 mg/L, and 330 U/L, P=0.009, P=0.05, and P=0.004, respectively). COPD patients had lower UFP concentrations in EBC compared with controls (0.24 E8/mL vs 0.51 E8/mL, P≤0.001). A mirror image was detected in serum: COPD patients had higher UFP concentrations compared with controls (9.8 E8/mL vs 6.7 E8/mL, respectively, P=0.03). EBC carbonyl and 8-OHdG levels were higher among COPD patients compared with controls (5.1 per 1 µg/mL protein and 0.036 ng/mL vs 0.41 per 1 µg/mL protein and 0.003 ng/mL, P=0.001 and P≤0.001, respectively). EBC UFP concentrations were negatively correlated with pack years (R=−0.44, P ≤0.001) and positively correlated with FEV1 and diffusing lung capacity for carbon monoxide (R=0.46, 0.23, P ≤0.001 and P=0.04, respectively). Low EBC UFP concentrations (≤0.18 E8/mL) and CRP levels ≥5 mg/L were independent predictors of the frequent exacerbator phenotype (OR 3.6; 95% CI: 1.06–7.97; P=0.04 and OR 4.4; 95% CI: 1.24–10.2; P=0.02, respectively). Conclusion UFP content in EBC reflects the inflammatory state of airways. Low UFP concentrations in EBC and high in serum of COPD patients support our hypothesis that increased epithelial permeability could be the mechanism behind those findings.
Collapse
Affiliation(s)
- Einat Fireman Klein
- Pulmonary Division, Carmel Medical Center, Rappaport Faculty of Medicine, Technion - Institute of Technology, Haifa, Israel,
| | - Yochai Adir
- Pulmonary Division, Carmel Medical Center, Rappaport Faculty of Medicine, Technion - Institute of Technology, Haifa, Israel,
| | - Amir Krencel
- Division of Allergy and Clinical Immunology, Bnai Zion Medical Center, Rappaport Faculty of Medicine, Technion - Institute of Technology, Haifa, Israel
| | - Regina Peri
- Division of Allergy and Clinical Immunology, Bnai Zion Medical Center, Rappaport Faculty of Medicine, Technion - Institute of Technology, Haifa, Israel
| | - Bella Vasserman
- Division of Allergy and Clinical Immunology, Bnai Zion Medical Center, Rappaport Faculty of Medicine, Technion - Institute of Technology, Haifa, Israel
| | - Elizabeth Fireman
- Institute of Pulmonary Diseases, National Laboratory Service for Interstitial Lung Diseases, Tel-Aviv Sourasky Medical Center, Tel Aviv, Israel.,Department of Occupational and Environmental Medicine, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Aharon Kessel
- Division of Allergy and Clinical Immunology, Bnai Zion Medical Center, Rappaport Faculty of Medicine, Technion - Institute of Technology, Haifa, Israel
| |
Collapse
|
66
|
Qin Q, Wu Q, Wang Y, Xiong R, Guo L, Fu X, Rosenfeldt H, Bryant M, Cao X. Effects of cellular differentiation in human primary bronchial epithelial cells: Metabolism of 4-(methylnitrosamine)-1-(3-pyridyl)-1-butanone. Toxicol In Vitro 2019; 55:185-194. [PMID: 30552994 PMCID: PMC7953429 DOI: 10.1016/j.tiv.2018.12.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Revised: 12/06/2018] [Accepted: 12/11/2018] [Indexed: 12/13/2022]
Abstract
Many of the toxicants in tobacco smoke undergo biotransformation in the lungs of smokers, both to reactive and to detoxified derivatives. Human air-liquid-interface (ALI) airway tissue models have emerged as an advanced in vitro model for evaluating the toxicity of inhaled substances; however, the metabolic potential of these cultures has not been evaluated extensively. In this study, we compared the metabolic activities of an ALI tissue model to the undifferentiated normal human primary bronchial epithelial (NHBE) cells from which it was derived. Measurement of the basal levels of gene expression for 84 phase I drug metabolism enzymes indicated that most genes were upregulated in ALI cultures compared to NHBE cells. Furthermore, the enzymatic activities of three cytochrome P450s involved in the bioactivation of tobacco-specific nitrosamines were higher in the ALI cultures, and the bioactivation of 4-(methylnitrosamine)-1-(3-pyridyl)-1-butanone (NNK), as measured by the formation of two of its major metabolites, i.e., keto acid and 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanol (NNAL), was significantly greater in the ALI cultures. Finally, NNK was a direct-acting genotoxicant in the ALI cultures, while the genotoxicity of NNK was detected in NHBE cells only in the presence of an exogenous liver S9 activation system. Taken together, our findings demonstrate the greater metabolic potential of well-differentiated ALI cultures than primary NHBE cells, supporting the potential use of ALI airway cultures as an alternative in vitro model for evaluating inhaled toxicants that require metabolic transformation.
Collapse
Affiliation(s)
- Qin Qin
- Division of Genetic and Molecular Toxicology, Division of Biochemical Toxicology, National Center for Toxicological Research, U.S Food and Drug Administration, Jefferson, AR 72079, United States
| | - Qiangen Wu
- Division of Biochemical Toxicology, National Center for Toxicological Research, U.S Food and Drug Administration, Jefferson, AR 72079, United States
| | - Yiying Wang
- Division of Genetic and Molecular Toxicology, Division of Biochemical Toxicology, National Center for Toxicological Research, U.S Food and Drug Administration, Jefferson, AR 72079, United States
| | - Rui Xiong
- Division of Genetic and Molecular Toxicology, Division of Biochemical Toxicology, National Center for Toxicological Research, U.S Food and Drug Administration, Jefferson, AR 72079, United States
| | - Lei Guo
- Division of Biochemical Toxicology, National Center for Toxicological Research, U.S Food and Drug Administration, Jefferson, AR 72079, United States
| | - Xin Fu
- Division of Nonclinical Science, Center for Tobacco Products, U.S Food and Drug Administration, Silver Spring, MD 20993, United States; Division of Clinical Review, Office of Bioequivalence, Office of Generic Drugs, Center for Drug Evaluation and Research, U.S Food and Drug Administration, Silver Spring, MD 20993, United States
| | - Hans Rosenfeldt
- Division of Nonclinical Science, Center for Tobacco Products, U.S Food and Drug Administration, Silver Spring, MD 20993, United States
| | - Matthew Bryant
- Division of Biochemical Toxicology, National Center for Toxicological Research, U.S Food and Drug Administration, Jefferson, AR 72079, United States
| | - Xuefei Cao
- Division of Genetic and Molecular Toxicology, Division of Biochemical Toxicology, National Center for Toxicological Research, U.S Food and Drug Administration, Jefferson, AR 72079, United States.
| |
Collapse
|
67
|
Manley GCA, Parker LC, Zhang Y. Emerging Regulatory Roles of Dual-Specificity Phosphatases in Inflammatory Airway Disease. Int J Mol Sci 2019; 20:E678. [PMID: 30764493 PMCID: PMC6387402 DOI: 10.3390/ijms20030678] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Revised: 01/31/2019] [Accepted: 02/01/2019] [Indexed: 12/16/2022] Open
Abstract
Inflammatory airway disease, such as asthma and chronic obstructive pulmonary disease (COPD), is a major health burden worldwide. These diseases cause large numbers of deaths each year due to airway obstruction, which is exacerbated by respiratory viral infection. The inflammatory response in the airway is mediated in part through the MAPK pathways: p38, JNK and ERK. These pathways also have roles in interferon production, viral replication, mucus production, and T cell responses, all of which are important processes in inflammatory airway disease. Dual-specificity phosphatases (DUSPs) are known to regulate the MAPKs, and roles for this family of proteins in the pathogenesis of airway disease are emerging. This review summarizes the function of DUSPs in regulation of cytokine expression, mucin production, and viral replication in the airway. The central role of DUSPs in T cell responses, including T cell activation, differentiation, and proliferation, will also be highlighted. In addition, the importance of this protein family in the lung, and the necessity of further investigation into their roles in airway disease, will be discussed.
Collapse
Affiliation(s)
- Grace C A Manley
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117545, Singapore.
- Immunology Programme, Life Science Institute, National University of Singapore, Singapore 117597, Singapore.
| | - Lisa C Parker
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield S10 2RX, UK.
| | - Yongliang Zhang
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117545, Singapore.
- Immunology Programme, Life Science Institute, National University of Singapore, Singapore 117597, Singapore.
| |
Collapse
|
68
|
Iron Homeostasis in the Lungs-A Balance between Health and Disease. Pharmaceuticals (Basel) 2019; 12:ph12010005. [PMID: 30609678 PMCID: PMC6469191 DOI: 10.3390/ph12010005] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Revised: 12/23/2018] [Accepted: 12/25/2018] [Indexed: 12/15/2022] Open
Abstract
A strong mechanistic link between the regulation of iron homeostasis and oxygen sensing is evident in the lung, where both systems must be properly controlled to maintain lung function. Imbalances in pulmonary iron homeostasis are frequently associated with respiratory diseases, such as chronic obstructive pulmonary disease and with lung cancer. However, the underlying mechanisms causing alterations in iron levels and the involvement of iron in the development of lung disorders are incompletely understood. Here, we review current knowledge about the regulation of pulmonary iron homeostasis, its functional importance, and the link between dysregulated iron levels and lung diseases. Gaining greater knowledge on how iron contributes to the pathogenesis of these diseases holds promise for future iron-related therapeutic strategies.
Collapse
|
69
|
Dong J, Liao W, Tan LH, Yong A, Peh WY, Wong WSF. Gene silencing of receptor-interacting protein 2 protects against cigarette smoke-induced acute lung injury. Pharmacol Res 2019; 139:560-568. [PMID: 30394320 DOI: 10.1016/j.phrs.2018.10.016] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2018] [Revised: 10/15/2018] [Accepted: 10/15/2018] [Indexed: 12/12/2022]
Abstract
BACKGROUND AND PURPOSE Chronic obstructive pulmonary disease (COPD) is characterized by progressive alveolar damage and generally irreversible airflow limitation. Nuclear factor-κB (NF-κB) plays a critical role in COPD pathogenesis. Receptor-interacting protein 2 (Rip2), a 60 kDa adaptor protein, is a positive regulator of NF-κB pathway and also an inducible transcriptional product of NF-κB activation. We sought to investigate if Rip2 gene silencing could protect against cigarette smoke (CS)-induced acute lung injury. EXPERIMENTAL APPROACH Gene silencing efficacy of Rip2 siRNA was characterized in mouse macrophage and mouse lung epithelial cell lines, and in a CS-induced acute lung injury mouse model. Bronchoalveolar lavage (BAL) fluid cell counts, levels of pro-inflammatory and oxidative damage markers, lung section inflammatory and epithelium thickness scorings, and nuclear NF-κB translocation were measured. KEY RESULTS CS was found to upregulate Rip2 level in mouse lungs. Rip2 siRNA was able to suppress Rip2 levels in both macrophage and lung epithelial cell lines and in mouse lungs, block CS extract (CSE)-induced mediator release by the cultured cells, and abate neutrophil counts in BAL fluid from CS-challenged mice. Rip2 siRNA suppressed CS-induced inflammatory and oxidative damage markers, and nuclear p65 accumulation and transcriptional activation in lung tissues. Besides, Rip2 siRNA was able to disrupt CSE-induced NF-κB activation in a NF-κB reporter gene assay. CONCLUSIONS AND IMPLICATIONS Taken together, we report for the first time that Rip2 gene silencing ameliorated CS-induced acute lung injury probably via disruption of the NF-κB activity, postulating that Rip2 may be a novel therapeutic target for COPD.
Collapse
Affiliation(s)
- Jinrui Dong
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University Health System, Singapore
| | - Wupeng Liao
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University Health System, Singapore
| | - Lay Hong Tan
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University Health System, Singapore
| | - Amy Yong
- Department of Pharmacology and Therapeutics, Faculty of Life Sciences and Medicine, King's College London, UK
| | - Wen Yan Peh
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University Health System, Singapore
| | - W S Fred Wong
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University Health System, Singapore; Immunology Program, Life Science Institute, National University of Singapore, Singapore; Singapore-HUJ Alliance for Research and Enterprise, Molecular Mechanisms of Inflammatory Diseases Interdisciplinary Research Group, Singapore.
| |
Collapse
|
70
|
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]
|
71
|
Hasby Saad MA, Watany M, Tomoum M, El-Mehy D, Elsheikh M, Sharshar R. Acidic mammalian chitinase tuning after enteric helminths eradication in inflammatory respiratory disease patients. Parasite Immunol 2018; 40:e12583. [DOI: 10.1111/pim.12583] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Revised: 08/07/2018] [Accepted: 08/09/2018] [Indexed: 01/18/2023]
Affiliation(s)
- Marwa A. Hasby Saad
- Medical Parasitology Department; Faculty of Medicine; Tanta University; Tanta Egypt
| | - Mona Watany
- Clinical Pathology Department; Faculty of Medicine; Tanta University; Tanta Egypt
| | - Mohamed Tomoum
- Otorhinolaryngeology Department; Faculty of Medicine; Tanta University; Tanta Egypt
| | - Dalia El-Mehy
- Medical Parasitology Department; Faculty of Medicine; Tanta University; Tanta Egypt
| | - May Elsheikh
- Paediatric Department; Faculty of Medicine; Tanta University; Tanta Egypt
| | - Ragia Sharshar
- Pulmonology Department; Faculty of Medicine; Tanta University; Tanta Egypt
| |
Collapse
|
72
|
Dong J, Liao W, Peh HY, Tan WSD, Zhou S, Wong WSF. Ribosomal Protein S3 Gene Silencing Protects Against Cigarette Smoke-Induced Acute Lung Injury. MOLECULAR THERAPY. NUCLEIC ACIDS 2018; 12:370-380. [PMID: 30195775 PMCID: PMC6031153 DOI: 10.1016/j.omtn.2018.05.027] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/08/2017] [Revised: 05/11/2018] [Accepted: 05/12/2018] [Indexed: 01/18/2023]
Abstract
Chronic obstructive pulmonary disease (COPD) is estimated to be the third leading cause of death by 2030. Transcription factor NF-κB may play a critical role in COPD pathogenesis. Ribosomal protein S3 (RPS3), a 40S ribosomal protein essential for executing protein translation, has recently been found to interact with the NF-κB p65 subunit and promote p65 DNA-binding activity. We sought to study whether RPS3 gene silencing could protect against cigarette-smoke (CS)-induced acute lung injury in a mouse model. Effects of an intratracheal RPS3 siRNA in CS-induced lung injury were determined by measuring bronchoalveolar lavage (BAL) fluid cell counts, levels of inflammatory and oxidative damage markers, and NF-κB translocation. Lung RPS3 level was found to be upregulated for the first time with CS exposure, and RPS3 siRNA blocked CS-induced neutrophil counts in BAL fluid. RPS3 siRNA suppressed CS-induced lung inflammatory mediator and oxidative damage marker levels, as well as nuclear p65 accumulation and transcriptional activation. RPS3 siRNA was able to disrupt CS extract (CSE)-induced NF-κB activation in an NF-κB reporter gene assay. We report for the first time that RPS3 gene silencing ameliorated CS-induced acute lung injury, probably via interruption of the NF-κB activity, postulating that RPS3 is a novel therapeutic target for COPD.
Collapse
Affiliation(s)
- Jinrui Dong
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University Health System, Singapore, Singapore
| | - Wupeng Liao
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University Health System, Singapore, Singapore
| | - Hong Yong Peh
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University Health System, Singapore, Singapore
| | - W S Daniel Tan
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University Health System, Singapore, Singapore
| | - Shuo Zhou
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University Health System, Singapore, Singapore
| | - W S Fred Wong
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University Health System, Singapore, Singapore; Immunology Program, Life Science Institute, National University of Singapore, Singapore, Singapore; Singapore-HUJ Alliance for Research and Enterprise, Molecular Mechanisms of Inflammatory Diseases Interdisciplinary Research Group, Singapore, Singapore.
| |
Collapse
|
73
|
Kim YH, An YJ, Jo S, Lee SH, Lee SJ, Choi SJ, Lee K. Comparison of volatile organic compounds between cigarette smoke condensate (CSC) and extract (CSE) samples. ENVIRONMENTAL HEALTH AND TOXICOLOGY 2018; 33:e2018012-0. [PMID: 30286588 PMCID: PMC6182245 DOI: 10.5620/eht.e2018012] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Accepted: 08/27/2018] [Indexed: 06/08/2023]
Abstract
Cigarette smoke is a major risk factor for several diseases, including chronic obstructive pulmonary and cardiovascular diseases. The toxicity of the cigarette smoke can be determined in vitro. The cytotoxicity test of the cigarette smoke is commonly conducted using the cigarette smoke condensate (CSC) and cigarette smoke extract (CSE). The CSC and CSE methods are well known for sampling of the particles and water-soluble compounds in the cigarette smoke, respectively. In this study, the CSC and CSE were analyzed by using a gas chromatography-mass spectrometry (GC-MS) system equipped with a wax column for separation of the volatile organic compounds. The cytotoxic effect of the CSC and CSE were evaluated thoroughly by comparing the analytical results of the CSC and CSE samples. The total concentration of the volatile organic compounds detected in the CSC sample was similar to that in the CSE sample based on the peak area. Except for the dimethyl sulfoxide solvent, nicotine had the highest concentration in the CSC sample, while acetonitrile had the highest concentration in the CSE sample. The compositions were as follows: (1) CSC sample: 55.8% nicotine, 18.0% nicotyrine, 3.20% 1,2,3-propanetriol, triacetate, 1.28% ethyl chloride, 1.22% phenol, etc. and (2) CSE sample: 18.7% acetonitrile, 18.0% acetone, 12.5% 2-hydroxy-2-methyl-propanenitrile, 8.98% nicotine, 5.86% nicotyrine, etc. In this manner, to accurately examine the cytotoxicity of the cigarette smoke using CSC or CSE, the components and their concentrations in the CSC and CSE samples should be considered.
Collapse
Affiliation(s)
- Yong-Hyun Kim
- Jeonbuk Department of Inhalation Research, Korea Institute of Toxicology, Jeongeup 56212, Republic of Korea
- Human and Environmental Toxicology, University of Science and Technology, Daejeon 34113, Republic of Korea
| | - Young-Ji An
- Department of Toxicology Evaluation, Konyang University, Daejeon 35365 Republic of Korea
- National Center for Efficacy Evaluation of Respiratory Disease Product, Korea Institute of Toxicology, Jeongeup 56212, Republic of Korea
| | - Seongwon Jo
- Jeonbuk Department of Inhalation Research, Korea Institute of Toxicology, Jeongeup 56212, Republic of Korea
| | - Sang-Hyub Lee
- Jeonbuk Department of Inhalation Research, Korea Institute of Toxicology, Jeongeup 56212, Republic of Korea
| | - Sang Jin Lee
- National Center for Efficacy Evaluation of Respiratory Disease Product, Korea Institute of Toxicology, Jeongeup 56212, Republic of Korea
| | - Seong-Jin Choi
- Jeonbuk Department of Inhalation Research, Korea Institute of Toxicology, Jeongeup 56212, Republic of Korea
| | - Kyuhong Lee
- Human and Environmental Toxicology, University of Science and Technology, Daejeon 34113, Republic of Korea
- National Center for Efficacy Evaluation of Respiratory Disease Product, Korea Institute of Toxicology, Jeongeup 56212, Republic of Korea
| |
Collapse
|
74
|
Tejero JD, Armand NC, Finn CM, Dhume K, Strutt TM, Chai KX, Chen LM, McKinstry KK. Cigarette smoke extract acts directly on CD4 T cells to enhance Th1 polarization and reduce memory potential. Cell Immunol 2018; 331:121-129. [PMID: 29935764 PMCID: PMC6092241 DOI: 10.1016/j.cellimm.2018.06.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Revised: 06/11/2018] [Accepted: 06/14/2018] [Indexed: 12/12/2022]
Abstract
Although cigarette smoke is known to alter immune responses, whether and how CD4 T cells are affected is not well-described. We aimed to characterize how exposure to cigarette smoke extract impacts CD4 T cell effector generation in vitro under Th1-polarizing conditions. Our results demonstrate that cigarette smoke directly acts on CD4 T cells to impair effector expansion by decreasing division and increasing apoptosis. Furthermore, cigarette smoke enhances Th1-associated cytokine production and increases expression of the transcription factor T-bet, the master regulator of Th1 differentiation. Finally, we show that exposure to cigarette smoke extract during priming impairs the ability of effectors to form memory cells. Our findings thus demonstrate that cigarette smoke simultaneously enhances effector functions but promotes terminal differentiation of CD4 T cell effectors. This study may be relevant to understanding how smoking can both aggravate autoimmune symptoms and reduce vaccine efficacy.
Collapse
Affiliation(s)
- Joanne D Tejero
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL, USA
| | - Nicole C Armand
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL, USA
| | - Caroline M Finn
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL, USA
| | - Kunal Dhume
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL, USA
| | - Tara M Strutt
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL, USA
| | - Karl X Chai
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL, USA
| | - Li-Mei Chen
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL, USA
| | - K Kai McKinstry
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL, USA.
| |
Collapse
|
75
|
Glynos C, Bibli SI, Katsaounou P, Pavlidou A, Magkou C, Karavana V, Topouzis S, Kalomenidis I, Zakynthinos S, Papapetropoulos A. Comparison of the effects of e-cigarette vapor with cigarette smoke on lung function and inflammation in mice. Am J Physiol Lung Cell Mol Physiol 2018; 315:L662-L672. [PMID: 30091379 DOI: 10.1152/ajplung.00389.2017] [Citation(s) in RCA: 126] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Electronic cigarettes (e-cigs) are advertised as a less harmful nicotine delivery system or as a new smoking cessation tool. We aimed to assess the in vivo effects of e-cig vapor in the lung and to compare them to those of cigarette smoke (CS). We exposed C57BL/6 mice for either 3 days or 4 wk to ambient air, CS, or e-cig vapor containing 1) propylene glycol/vegetable glycerol (PG:VG-Sol; 1:1), 2) PG:VG with nicotine (G:VG-N), or 3) PG:VG with nicotine and flavor (PG:VG-N+F) and determined oxidative stress, inflammation, and pulmonary mechanics. E-cig vapors, especially PG:VG-N+F, increased bronchoalveolar lavage fluid (BALF) cellularity, Muc5ac production, as well as BALF and lung oxidative stress markers at least comparably and in many cases more than CS. BALF protein content at both time points studied was only elevated in the PG:VG-N+F group. After 3 days, PG:VG-Sol altered tissue elasticity, static compliance, and airway resistance, whereas after 4 wk CS was the only treatment adversely affecting these parameters. Airway hyperresponsiveness in response to methacholine was increased similarly in the CS and PG:VG-N+F groups. Our findings suggest that exposure to e-cig vapor can trigger inflammatory responses and adversely affect respiratory system mechanics. In many cases, the added flavor in e-cigs exacerbated the detrimental effects of e-cig vapor. We conclude that both e-cig vaping and conventional cigarette smoking negatively impact lung biology.
Collapse
Affiliation(s)
- Constantinos Glynos
- George P. Livanos and Marianthi Simou Laboratories, Evangelismos Hospital, 1st Department of Pulmonary and Critical Care, National and Kapodistrian University of Athens Medical School , Greece
| | - Sofia-Iris Bibli
- Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens , Athens , Greece.,Institute for Vascular Signaling, Centre for Molecular Medicine, Goethe University , Frankfurt am Main , Germany
| | - Paraskevi Katsaounou
- George P. Livanos and Marianthi Simou Laboratories, Evangelismos Hospital, 1st Department of Pulmonary and Critical Care, National and Kapodistrian University of Athens Medical School , Greece
| | - Athanasia Pavlidou
- George P. Livanos and Marianthi Simou Laboratories, Evangelismos Hospital, 1st Department of Pulmonary and Critical Care, National and Kapodistrian University of Athens Medical School , Greece.,Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens , Athens , Greece
| | - Christina Magkou
- Department of Histopathology, Evangelismos Hospital , Athens , Greece
| | - Vassiliki Karavana
- George P. Livanos and Marianthi Simou Laboratories, Evangelismos Hospital, 1st Department of Pulmonary and Critical Care, National and Kapodistrian University of Athens Medical School , Greece
| | - Stavros Topouzis
- Laboratory of Molecular Pharmacology, Department of Pharmacy, University of Patras , Patras , Greece
| | - Ioannis Kalomenidis
- George P. Livanos and Marianthi Simou Laboratories, Evangelismos Hospital, 1st Department of Pulmonary and Critical Care, National and Kapodistrian University of Athens Medical School , Greece
| | - Spyros Zakynthinos
- George P. Livanos and Marianthi Simou Laboratories, Evangelismos Hospital, 1st Department of Pulmonary and Critical Care, National and Kapodistrian University of Athens Medical School , Greece
| | - Andreas Papapetropoulos
- George P. Livanos and Marianthi Simou Laboratories, Evangelismos Hospital, 1st Department of Pulmonary and Critical Care, National and Kapodistrian University of Athens Medical School , Greece.,Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens , Athens , Greece
| |
Collapse
|
76
|
Zuo J, Hu Z, Liu T, Chen C, Tao Z, Chen S, Li F. Calpeptin attenuates cigarette smoke-induced pulmonary inflammation via suppressing calpain/IκBα signaling in mice and BEAS-2B cells. Pathol Res Pract 2018; 214:1199-1209. [PMID: 30078403 DOI: 10.1016/j.prp.2018.06.019] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Revised: 06/19/2018] [Accepted: 06/25/2018] [Indexed: 11/29/2022]
Abstract
Exposure to cigarette smoke including secondhand smoking is the most important risk factor in the development of chronic obstructive pulmonary disease where incidence has substantially increased in recent decades. The mechanisms responsible for cigarette smoke-induced pulmonary inflammation remain unclear, and thus lack of effective treatment. The present study investigated the effect of calpeptin on attenuating cigarette smoke induced pulmonary inflammation and its potential mechanism and function. When BALB/c mice were exposed to cigarette smoke and received calpeptin intraperitoneally injection after 90 days, calpeptin histologically attenuated the accumulation of neutrophils (P < 0.001), eosinophils (P < 0.001), macrophages (P < 0.01), fibrinous exudation and proliferation within the interstitial and alveolar spaces. BEAS-2B cells were added with cigarette smoke extract in vitro and treated with calpeptin for 24 h in the treatment group. The markedly upregulation of μ-calpain (P < 0.01), m-calpain (P < 0.001) and IκBα (P < 0.01) in cigarette smoke-induced lungs were simultaneously decreased by calpeptin treatment (P < 0.05). The increased expression of μ-calpain, m-calpain and IκBα (P < 0.05) in cigarette smoke extract-stimulated BEAS-2B cells were also decreased by calpeptin treatment (P < 0.05). These data indicated that calpeptin attenuated cigarette smoke-induced pulmonary inflammation by suppressing the pathway of μ-calpain, m-calpain and IκBα in vivo and in vitro. Calpeptin might have a potential for prevention of the development of inflammatory pulmonary diseases and warrant further pharmaceutical investigation.
Collapse
Affiliation(s)
- Jingjing Zuo
- Department of Otolaryngology Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, PR China; Research Institute of Otolaryngology Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, PR China; Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, PR China
| | - Zhangwei Hu
- Department of Otolaryngology Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, PR China; Research Institute of Otolaryngology Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, PR China; Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, PR China
| | - Tao Liu
- Department of Otolaryngology Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, PR China; Research Institute of Otolaryngology Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, PR China; Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, PR China
| | - Chen Chen
- Research Institute of Otolaryngology Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, PR China
| | - Zezhang Tao
- Department of Otolaryngology Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, PR China; Research Institute of Otolaryngology Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, PR China.
| | - Shiming Chen
- Department of Otolaryngology Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, PR China; Research Institute of Otolaryngology Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, PR China
| | - Fen Li
- Research Institute of Otolaryngology Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, PR China
| |
Collapse
|
77
|
Demb J, Wei EK, Izano M, Kritchevsky S, Swede H, Newman AB, Shlipak M, Akinyemiju T, Gregorich S, Braithwaite D. Chronic inflammation and risk of lung cancer in older adults in the health, aging and body composition cohort study. J Geriatr Oncol 2018; 10:265-271. [PMID: 30078713 DOI: 10.1016/j.jgo.2018.07.008] [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] [Received: 02/06/2018] [Revised: 06/21/2018] [Accepted: 07/11/2018] [Indexed: 11/29/2022]
Abstract
OBJECTIVES We examined the association between three inflammatory markers (Interleukin (IL)-6, C-reactive protein (CRP), tumor necrosis factor (TNF)-α) and incident lung cancer using baseline, updated, and averaged inflammatory measures in older adults. METHODS We fitted multivariable Cox models to assess whether circulating levels of inflammation markers were associated with incident lung cancers in the Health Aging, Body and Composition (HealthABC) prospective cohort of 3075 older adults aged 70-79 years at baseline. IL-6 and CRP were measured biennially, whereas TNF-α was measured at baseline. RESULTS Baseline levels of IL-6 were significantly associated with incident lung cancer risk in a model that adjusted for age, gender, race, and site (Model 1) (Hazard RatioT3 vs. T1: 3.34, 95% Confidence Interval: 1.91, 5.85) and in a model adjusted for health factors linked to chronic inflammation (Model 2) (HR T3 vs. T1: 2.57, 95% CI: 1.41, 4.65). The associations observed in time-updated IL-6 (HR T3 vs. T1: 2.47, 95% CI: 1.43, 4.28), cumulatively averaged IL-6 (HR T3 vs. T1: 2.47, 95% CI: 1.43, 4.35), and baseline CRP levels (HR T3 vs. T1: 1.85, 95% CI: 1.11, 3.08) with incident lung cancer in Model 1 were not statistically significant in Model 2. CONCLUSIONS Baseline CRP and IL-6 levels were associated with increased risk of lung cancer in Model 1 and both models, respectively. Chronic IL-6 inflammation, as quantified by repeated measures was associated with incident lung cancer in Model 1, but not Model 2. Further research is needed to understand the role of CRP and IL-6 in lung carcinogenesis.
Collapse
Affiliation(s)
- Joshua Demb
- Department of Epidemiology and Biostatistics, University of California, San Francisco, CA, United States; Department of Oncology, Georgetown University, Washington, DC, United States.
| | - Esther K Wei
- California Pacific Medical Center Research Institute, San Francisco, CA, United States.
| | - Monika Izano
- Department of Epidemiology, University of California, Berkeley, CA, United States
| | | | - Helen Swede
- Department of Community Medicine and Healthcare, University of Connecticut School of Medicine, Farmington, CT, United States.
| | - Anne B Newman
- School of Public Health, University of Pittsburgh, Pittsburgh, PA, United States.
| | - Michael Shlipak
- Department of Epidemiology and Biostatistics, University of California, San Francisco, CA, United States.
| | - Tomi Akinyemiju
- Department of Epidemiology, University of Kentucky, Lexington, KY, United States.
| | - Steven Gregorich
- Department of Medicine, University of California, San Francisco, CA, United States.
| | - Dejana Braithwaite
- Department of Oncology, Georgetown University, Washington, DC, United States.
| |
Collapse
|
78
|
A Novel In Vivo Model to Study Impaired Tissue Regeneration Mediated by Cigarette Smoke. Sci Rep 2018; 8:10926. [PMID: 30026555 PMCID: PMC6053433 DOI: 10.1038/s41598-018-28687-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Accepted: 06/13/2018] [Indexed: 12/27/2022] Open
Abstract
Cigarette smoke is associated with several pathologies including chronic respiratory diseases and cancer. In addition, exposure to cigarette smoke is correlated with impaired wound healing, where a significant decrease in the regenerative capacity of smokers is well documented and broadly considered a negative risk factor after trauma or surgery. So far, some in vitro and in vivo models have been described to study how exposure to cigarette smoke diminishes the regenerative potential in different organisms. However, although useful, many of these models are difficult and expensive to implement and do not allow high-throughput screening approaches. In order to establish a reliable and accessible model, we have evaluated the effects of cigarette smoke extract (CSE) on zebrafish development and regeneration. In this work, zebrafish embryos and larvae were exposed to low doses of aqueous CSE showing severe developmental abnormalities in a dose-dependent manner. Furthermore, when adult zebrafish were subjected to caudal fin amputation, we observed a significant decrease in the regenerative capacity of animals exposed to CSE. The effect was exacerbated in male and aged fish compared to female or young organisms. The establishment of a zebrafish model to assess the consequences of cigarette smoke and its effects on animal physiology could provide a new tool to study the underlying mechanisms involved in impaired tissue regeneration, and aid the development of novel approaches to treat complications associated with cigarette smoke toxicity.
Collapse
|
79
|
Huang SJ, Xu YM, Lau ATY. Electronic cigarette: A recent update of its toxic effects on humans. J Cell Physiol 2018; 233:4466-4478. [PMID: 29215738 DOI: 10.1002/jcp.26352] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Accepted: 11/29/2017] [Indexed: 02/05/2023]
Abstract
Electronic cigarettes (e-cigarettes), battery-powered and liquid-vaporizing devices, were invented to replace the conventional cigarette (c-cigarette) smoking for the sake of reducing the adverse effects on multiple organ systems that c-cigarettes have induced. Although some of the identified harmful components in e-cigarettes were alleged to be measured in lower quantity than those in c-cigarettes, researchers unveiled that the toxic effects of e-cigarettes should not be understated. This review is sought for an attempt to throw light on several typical types of e-cigarette components (tobacco-specific nitrosamines, carbonyl compounds, and volatile organic compounds) by revealing their possible impacts on human bodies through different action mechanisms characterized by alteration of specific biomarkers on cellular and molecular levels. In addition, this review is intended to draw the limelight that like c-cigarettes, e-cigarettes could also be accompanied with toxic effects on whole human body, which are especially apparent on respiratory system. From head to foot, from physical aspect to chemical aspect, from genotype to phenotype, potential alterations will take place upon the intake of the liquid aerosol.
Collapse
Affiliation(s)
- Shu-Jie Huang
- Laboratory of Cancer Biology and Epigenetics, Department of Cell Biology and Genetics, Shantou University Medical College, Shantou, Guangdong, People's Republic of China
| | - Yan-Ming Xu
- Laboratory of Cancer Biology and Epigenetics, Department of Cell Biology and Genetics, Shantou University Medical College, Shantou, Guangdong, People's Republic of China
| | - Andy T Y Lau
- Laboratory of Cancer Biology and Epigenetics, Department of Cell Biology and Genetics, Shantou University Medical College, Shantou, Guangdong, People's Republic of China
| |
Collapse
|
80
|
Wielgat P, Trofimiuk E, Czarnomysy R, Holownia A, Braszko JJ. Sialylation pattern in lung epithelial cell line and Siglecs expression in monocytic THP-1 cells as cellular indicators of cigarette smoke - induced pathology in vitro. Exp Lung Res 2018; 44:167-177. [PMID: 29781747 DOI: 10.1080/01902148.2018.1461959] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
PURPOSE Cellular response to cigarette smoke (CS) involves activation of recognition receptors resulting in changes in immune status, oxidative stress and cell turnover. We investigated the effects of CS on sialic acid-binding immunoglobulin type lectins (Siglecs) expression and their sialylated ligands in human immune and non-immune cells. METHODS Human monocytes (THP-1) and epithelial cells (A549) were cultured in CS-conditioned medium (CSM). Expression of Siglec-8 and Siglec-5/Siglec-14 was analysed in THP-1 cells using flow cytometry. The effects of CS on immune activity was evaluated flow cytometrically in these cells by assessment of phagocytosis and intracellular expression IL-1β and IL-10. Detection and differentiation of sialic acids was analyzed by dot blot, western blot and flow cytometry using plant lectins and antibodies. RESULTS Exposure to CS significantly increased expression of Siglec-8 and Siglec-5/Siglec-14 in THP-1 cells. These changes were accompanied by enhanced intracellular level of IL-1β and IL-10 but reduced phagocytic activity. In THP-1 and A549 cells, the level of α2,3-sialic acids, but not α2,6-sialic acid, was significantly increased when compared to naïve cells. The level of α2,8-sialic acids increased significantly in A549 cells, but not in THP-1 cells, after exposure to CS. CONCLUSION These results show that cellular response to CS involves changes in expression of Siglec receptors and sialylated ligands functionally associated with immunity.
Collapse
Affiliation(s)
- Przemyslaw Wielgat
- a Department of Clinical Pharmacology , Medical University of Bialystok , Bialystok , Poland
| | - Emil Trofimiuk
- a Department of Clinical Pharmacology , Medical University of Bialystok , Bialystok , Poland
| | - Robert Czarnomysy
- b Department of Synthesis and Technology of Drugs , Medical University of Bialystok , Bialystok , Poland
| | - Adam Holownia
- c Department of Pharmacology , Medical University of Bialystok , Bialystok , Poland
| | - Jan J Braszko
- a Department of Clinical Pharmacology , Medical University of Bialystok , Bialystok , Poland
| |
Collapse
|
81
|
Benedikter BJ, Wouters EFM, Savelkoul PHM, Rohde GGU, Stassen FRM. Extracellular vesicles released in response to respiratory exposures: implications for chronic disease. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART B, CRITICAL REVIEWS 2018; 21:142-160. [PMID: 29714636 DOI: 10.1080/10937404.2018.1466380] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Extracellular vesicles (EV) are secreted signaling entities that enhance various pathological processes when released in response to cellular stresses. Respiratory exposures such as cigarette smoke and air pollution exert cellular stresses and are associated with an increased risk of several chronic diseases. The aim of this review was to examine the evidence that modifications in EV contribute to respiratory exposure-associated diseases. Publications were searched using PubMed and Google Scholar with the search terms (cigarette smoke OR tobacco smoke OR air pollution OR particulate matter) AND (extracellular vesicles OR exosomes OR microvesicles OR microparticles OR ectosomes). All original research articles were included and reviewed. Fifty articles were identified, most of which investigated the effect of respiratory exposures on EV release in vitro (25) and/or on circulating EV in human plasma (24). The majority of studies based their main observations on the relatively insensitive scatter-based flow cytometry of EV (29). EV induced by respiratory exposures were found to modulate inflammation (19), thrombosis (13), endothelial dysfunction (11), tissue remodeling (6), and angiogenesis (3). By influencing these processes, EV may play a key role in the development of cardiovascular diseases and chronic obstructive pulmonary disease and possibly lung cancer and allergic asthma. The current findings warrant additional research with improved methodologies to evaluate the contribution of respiratory exposure-induced EV to disease etiology, as well as their potential as biomarkers of exposure or risk and as novel targets for preventive or therapeutic strategies.
Collapse
Affiliation(s)
- Birke J Benedikter
- a Department of Medical Microbiology , NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center , Maastricht , The Netherlands
- b Department of Respiratory Medicine , NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center , Maastricht , The Netherlands
| | - Emiel F M Wouters
- b Department of Respiratory Medicine , NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center , Maastricht , The Netherlands
| | - Paul H M Savelkoul
- a Department of Medical Microbiology , NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center , Maastricht , The Netherlands
- c Department of Medical Microbiology & Infection Control , VU University Medical Center , Amsterdam , The Netherlands
| | - Gernot G U Rohde
- d Medical clinic I, Department of Respiratory Medicine , Goethe University Hospital , Frankfurt/Main , Germany
| | - Frank R M Stassen
- a Department of Medical Microbiology , NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center , Maastricht , The Netherlands
| |
Collapse
|
82
|
Fallacara A, Busato L, Pozzoli M, Ghadiri M, Ong HX, Young PM, Manfredini S, Traini D. Combination of urea-crosslinked hyaluronic acid and sodium ascorbyl phosphate for the treatment of inflammatory lung diseases: An in vitro study. Eur J Pharm Sci 2018; 120:96-106. [PMID: 29723596 DOI: 10.1016/j.ejps.2018.04.042] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Revised: 03/16/2018] [Accepted: 04/27/2018] [Indexed: 12/29/2022]
Abstract
This in vitro study evaluated, for the first time, the safety and the biological activity of a novel urea-crosslinked hyaluronic acid component and sodium ascorbyl phosphate (HA-CL - SAP), singularly and/or in combination, intended for the treatment of inflammatory lung diseases. The aim was to understand if the combination HA-CL - SAP had an enhanced activity with respect to the combination native hyaluronic acid (HA) - SAP and the single SAP, HA and HA-CL components. Sample solutions displayed pH, osmolality and viscosity values suitable for lung delivery and showed to be not toxic on epithelial Calu-3 cells at the concentrations used in this study. The HA-CL - SAP displayed the most significant reduction in interleukin-6 (IL-6) and reactive oxygen species (ROS) levels, due to the combined action of HA-CL and SAP. Moreover, this combination showed improved cellular healing (wound closure) with respect to HA - SAP, SAP and HA, although at a lower rate than HA-CL alone. These preliminary results showed that the combination HA-CL - SAP could be suitable to reduce inflammation and oxidative stress in lung disorders like acute respiratory distress syndrome, asthma, emphysema and chronic obstructive pulmonary disease, where inflammation is prominent.
Collapse
Affiliation(s)
- Arianna Fallacara
- Respiratory Technology, Woolcock Institute of Medical Research and Discipline of Pharmacology, Sydney Medical School, The University of Sydney, 431 Glebe Point Road, Glebe, NSW 2037, Australia; Department of Life Sciences and Biotechnology, University of Ferrara, Via L. Borsari 46, 44121 Ferrara, Italy..
| | - Laura Busato
- Respiratory Technology, Woolcock Institute of Medical Research and Discipline of Pharmacology, Sydney Medical School, The University of Sydney, 431 Glebe Point Road, Glebe, NSW 2037, Australia; Department of Life Sciences and Biotechnology, University of Ferrara, Via L. Borsari 46, 44121 Ferrara, Italy..
| | - Michele Pozzoli
- Respiratory Technology, Woolcock Institute of Medical Research and Discipline of Pharmacology, Sydney Medical School, The University of Sydney, 431 Glebe Point Road, Glebe, NSW 2037, Australia.
| | - Maliheh Ghadiri
- Respiratory Technology, Woolcock Institute of Medical Research and Discipline of Pharmacology, Sydney Medical School, The University of Sydney, 431 Glebe Point Road, Glebe, NSW 2037, Australia.
| | - Hui Xin Ong
- Respiratory Technology, Woolcock Institute of Medical Research and Discipline of Pharmacology, Sydney Medical School, The University of Sydney, 431 Glebe Point Road, Glebe, NSW 2037, Australia.
| | - Paul M Young
- Respiratory Technology, Woolcock Institute of Medical Research and Discipline of Pharmacology, Sydney Medical School, The University of Sydney, 431 Glebe Point Road, Glebe, NSW 2037, Australia.
| | - Stefano Manfredini
- Department of Life Sciences and Biotechnology, University of Ferrara, Via L. Borsari 46, 44121 Ferrara, Italy..
| | - Daniela Traini
- Respiratory Technology, Woolcock Institute of Medical Research and Discipline of Pharmacology, Sydney Medical School, The University of Sydney, 431 Glebe Point Road, Glebe, NSW 2037, Australia.
| |
Collapse
|
83
|
Madani A, Alack K, Richter MJ, Krüger K. Immune-regulating effects of exercise on cigarette smoke-induced inflammation. J Inflamm Res 2018; 11:155-167. [PMID: 29731655 PMCID: PMC5923223 DOI: 10.2147/jir.s141149] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Long-term cigarette smoking (LTCS) represents an important risk factor for cardiac infarction and stroke and the central risk factor for the development of a bronchial carcinoma, smoking-associated interstitial lung fibrosis, and chronic obstructive pulmonary disease. The pathophysiologic development of these diseases is suggested to be promoted by chronic and progressive inflammation. Cigarette smoking induces repetitive inflammatory insults followed by a chronic and progressive activation of the immune system. In the pulmonary system of cigarette smokers, oxidative stress, cellular damage, and a chronic activation of pattern recognition receptors are described which are followed by the translocation of the NF-kB, the release of pro-inflammatory cytokines, chemokines, matrix metalloproteases, and damage-associated molecular patterns. In parallel, smoke pollutants cross directly through the alveolus-capillary interface and spread through the systemic bloodstream targeting different organs. Consequently, LTCS induces a systemic low-grade inflammation and increased oxidative stress in the vascular system. In blood, these processes promote an increased coagulation and endothelial dysfunction. In muscle tissue, inflammatory processes activate catabolic signaling pathways followed by muscle wasting and sarcopenia. In brain, several characteristics of neuroinflammation were described. Regular exercise training has been shown to be an effective nonpharmacological treatment strategy in smoke-induced pulmonary diseases. It is well established that exercise training exerts immune-regulating effects by activating anti-inflammatory signaling pathways. In this regard, the release of myokines from contracting skeletal muscle, the elevations of cortisol and adrenalin, the reduced expression of Toll-like receptors, and the increased mobilization of immune-regulating leukocyte subtypes might be of vital importance. Exercise training also increases the local and systemic antioxidative capacity and several compensatory mechanisms in tissues such as an increased anabolic signaling in muscle or an increased compliance of the vascular system. Accordingly, regular exercise training seems to protect long-term smokers against some important negative local and systemic consequences of smoking. Data suggest that it seems to be important to start exercise training as early as possible.
Collapse
Affiliation(s)
- Ashkan Madani
- Department of Exercise and Health, Institute of Sports Science, Leibniz University Hannover, Germany
| | - Katharina Alack
- Department of Sports Medicine, University of Giessen, Germany
| | - Manuel Jonas Richter
- Department of Internal Medicine, Justus Liebig University Giessen, Universities of Giessen and Marburg Lung Center (UGMLC), Germany
- German Center for Lung Research (DZL), Giessen, Germany
| | - Karsten Krüger
- Department of Exercise and Health, Institute of Sports Science, Leibniz University Hannover, Germany
| |
Collapse
|
84
|
Titz B, Kogel U, Martin F, Schlage WK, Xiang Y, Nury C, Dijon S, Baumer K, Peric D, Bornand D, Dulize R, Phillips B, Leroy P, Vuillaume G, Lebrun S, Elamin A, Guedj E, Trivedi K, Ivanov NV, Vanscheeuwijck P, Peitsch MC, Hoeng J. A 90-day OECD TG 413 rat inhalation study with systems toxicology endpoints demonstrates reduced exposure effects of the aerosol from the carbon heated tobacco product version 1.2 (CHTP1.2) compared with cigarette smoke. II. Systems toxicology assessment. Food Chem Toxicol 2018; 115:284-301. [PMID: 29545142 DOI: 10.1016/j.fct.2018.02.058] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2017] [Accepted: 02/27/2018] [Indexed: 12/11/2022]
Abstract
Modified risk tobacco products (MRTPs) have the potential to reduce smoking-related health risks. The Carbon Heated Tobacco Product 1.2 (CHTP1.2) is a potential MRTP that uses a pressed carbon heat source to generate an aerosol by heating tobacco. Here, we report the results from the systems toxicology arm of a 90-day rat inhalation study (OECD test guideline 413) to assess the effects of CHTP1.2 aerosol compared with cigarette smoke (CS). Transcriptomics, proteomics, and lipidomics analyses complemented the standard endpoints. In the respiratory nasal epithelium, CS induced an adaptive tissue and inflammatory response, which was much weaker after CHTP1.2 aerosol exposure, mostly limited to the highest CHTP1.2 concentration (at twice the 3R4F CS concentration: 50 vs. 23 μg nicotine/L), in female rats. In the lungs, the effects of CS exposure included inflammatory and cellular stress responses, which were absent or much lower after CHTP1.2 aerosol exposure. Outside of the respiratory tract, CS and CHTP1.2 aerosol induced effects that were previously associated with exposure to any nicotine-containing aerosol, e.g., lower lipid concentrations in serum. Overall, this systems toxicology analysis complements and confirms the results from classical toxicological endpoints and further suggests potentially reduced respiratory health risks of CHTP1.2.
Collapse
Affiliation(s)
- Bjoern Titz
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, 2000, Neuchatel, Switzerland(2)
| | - Ulrike Kogel
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, 2000, Neuchatel, Switzerland(2)
| | - Florian Martin
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, 2000, Neuchatel, Switzerland(2)
| | - Walter K Schlage
- Biology Consultant, Max-Baermann-Str. 21, 51429, Bergisch Gladbach, Germany
| | - Yang Xiang
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, 2000, Neuchatel, Switzerland(2)
| | - Catherine Nury
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, 2000, Neuchatel, Switzerland(2)
| | - Sophie Dijon
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, 2000, Neuchatel, Switzerland(2)
| | - Karine Baumer
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, 2000, Neuchatel, Switzerland(2)
| | - Dariusz Peric
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, 2000, Neuchatel, Switzerland(2)
| | - David Bornand
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, 2000, Neuchatel, Switzerland(2)
| | - Remi Dulize
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, 2000, Neuchatel, Switzerland(2)
| | - Blaine Phillips
- PMI R&D, Philip Morris International Research Laboratories Pte. Ltd., Science Park II, Singapore(2)
| | - Patrice Leroy
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, 2000, Neuchatel, Switzerland(2)
| | - Gregory Vuillaume
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, 2000, Neuchatel, Switzerland(2)
| | - Stefan Lebrun
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, 2000, Neuchatel, Switzerland(2)
| | - Ashraf Elamin
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, 2000, Neuchatel, Switzerland(2)
| | - Emmanuel Guedj
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, 2000, Neuchatel, Switzerland(2)
| | - Keyur Trivedi
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, 2000, Neuchatel, Switzerland(2)
| | - Nikolai V Ivanov
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, 2000, Neuchatel, Switzerland(2)
| | - Patrick Vanscheeuwijck
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, 2000, Neuchatel, Switzerland(2)
| | - Manuel C Peitsch
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, 2000, Neuchatel, Switzerland(2)
| | - Julia Hoeng
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, 2000, Neuchatel, Switzerland(2).
| |
Collapse
|
85
|
Long-term smoking alters abundance of over half of the proteome in bronchoalveolar lavage cell in smokers with normal spirometry, with effects on molecular pathways associated with COPD. Respir Res 2018. [PMID: 29514648 PMCID: PMC5842534 DOI: 10.1186/s12931-017-0695-6] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Background Smoking represents a significant risk factor for many chronic inflammatory diseases, including chronic obstructive pulmonary disease (COPD). Methods To identify dysregulation of specific proteins and pathways in bronchoalveolar lavage (BAL) cells associated with smoking, isobaric tags for relative and absolute quantitation (iTRAQ)-based shotgun proteomics analyses were performed on BAL cells from healthy never-smokers and smokers with normal lung function from the Karolinska COSMIC cohort. Multivariate statistical modeling, multivariate correlations with clinical data, and pathway enrichment analysis were performed. Results Smoking exerted a significant impact on the BAL cell proteome, with more than 500 proteins representing 15 molecular pathways altered due to smoking. The majority of these alterations occurred in a gender-independent manner. The phagosomal- and leukocyte trans endothelial migration (LTM) pathways significantly correlated with FEV1/FVC as well as the percentage of CD8+ T-cells and CD8+CD69+ T-cells in smokers. The correlations to clinical parameters in healthy never-smokers were minor. Conclusion The significant correlations of proteins in the phagosome- and LTM pathways with activated cytotoxic T-cells (CD69+) and the level of airway obstruction (FEV1/FVC) in smokers, both hallmarks of COPD, suggests that these two pathways may play a role in the molecular events preceding the development of COPD in susceptible smokers. Both pathways were found to be further dysregulated in COPD patients from the same cohort, thereby providing further support to this hypothesis. Given that not all smokers develop COPD in spite of decades of smoking, it is also plausible that some of the molecular pathways associated with response to smoking exert protective mechanisms to smoking-related pathologies in resilient individuals. Trial registration ClinicalTrials.gov identifier NCT02627872; Retrospectively registered on December 9, 2015. Electronic supplementary material The online version of this article (10.1186/s12931-017-0695-6) contains supplementary material, which is available to authorized users.
Collapse
|
86
|
Possebon L, de Souza Lima Lebron I, Furlan da Silva L, Tagliaferri Paletta J, Glad BG, Sant’Ana M, Iyomasa-Pilon MM, Ribeiro Souza H, de Souza Costa S, Pereira da Silva Rodriguesa G, Pereira MDL, de Haro Moreno A, Girol AP. Anti-inflammatory actions of herbal medicines in a model of chronic obstructive pulmonary disease induced by cigarette smoke. Biomed Pharmacother 2018; 99:591-597. [DOI: 10.1016/j.biopha.2018.01.106] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Revised: 01/18/2018] [Accepted: 01/24/2018] [Indexed: 12/20/2022] Open
|
87
|
Wu J, Li X, Qin Y, Cheng J, Hao G, Jin R, Zhu C. Jinwei Tang modulates HDAC2 expression in a rat model of COPD. Exp Ther Med 2018; 15:2604-2610. [PMID: 29456664 DOI: 10.3892/etm.2018.5707] [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] [Received: 05/15/2016] [Accepted: 04/07/2017] [Indexed: 11/06/2022] Open
Abstract
The aim of the present study was to investigate the effect of a Traditional Chinese Herbal Medicine (TCHM), named Jinwei Tang on histone deacetylase 2 (HDAC2) and its role in the regulation of corticosteroid resistance in a rat model of chronic obstructive pulmonary disease (COPD). Male Wistar rats were divided into five groups (each n=10): COPD group, established by the intratracheal instillation of lipopolysaccharide and passive smoke exposure, and control, budesonide, theophylline + budesonide and Jinwei Tang + budesonide groups. Lung function was measured, lung tissue histopathology was examined and HDAC2 expression in the lung was assessed by immunohistochemistry. In addition, protein levels of interleukin-8 (IL-8), tumor necrosis factor (TNF)-α and HDAC2 in lung homogenate were quantified by ELISA. The rat COPD model exhibited alterations of the ratio of forced expiratory volume in 0.2 sec (FEV0.2) to the forced vital capacity, FEV0.2, dynamic compliance and airway resistance. HDAC2 expression was markedly reduced in the lung tissue of the COPD group compared with the control group, and treatment with Jinwei Tang + budesonide or theophylline + budesonide resulted in significant attenuation of the reduction of HDAC2 expression in the lungs (P<0.05). However, treatment with budesonide alone did not significantly alter HDAC2 expression. In the Jinwei Tang + budesonide and theophylline + budesonide groups, IL-8 and TNF-α expression was significantly decreased (P<0.05) and the HDAC2 level increased (P<0.05) compared with that in the COPD group. In conclusion, Jinwei Tang modulates airway inflammation and may enhance the anti-inflammatory effect of glucocorticoid through the upregulation of HADC2 expression in a rat model of COPD.
Collapse
Affiliation(s)
- Jianjun Wu
- Department of Respiratory Disease, The Third Affiliated Hospital of Beijing University of Traditional Chinese Medicine, Beijing 100029, P.R. China
| | - Xin Li
- Department of Ocular Diseases, Eye Hospital, China Academy of Chinese Medical Sciences, Beijing 100040, P.R. China
| | - Yang Qin
- Department of Respiratory Disease, The Third Affiliated Hospital of Beijing University of Traditional Chinese Medicine, Beijing 100029, P.R. China
| | - Juan Cheng
- Department of Pathology, Dongzhimen Hospital Affiliated to Beijing University of Traditional Chinese Medicine, Beijing 100700, P.R. China
| | - Gaimei Hao
- Department of Pathology, Beijing University of Traditional Chinese Medicine, Beijing 100029, P.R. China
| | - Ruifeng Jin
- Department of Respiratory Disease, The Third Affiliated Hospital of Beijing University of Traditional Chinese Medicine, Beijing 100029, P.R. China
| | - Chenjun Zhu
- Department of Encephalopathy, The Third Affiliated Hospital of Beijing University of Traditional Chinese Medicine, Beijing 100029, P.R. China
| |
Collapse
|
88
|
Crotty Alexander LE, Drummond CA, Hepokoski M, Mathew D, Moshensky A, Willeford A, Das S, Singh P, Yong Z, Lee JH, Vega K, Du A, Shin J, Javier C, Tian J, Brown JH, Breen EC. Chronic inhalation of e-cigarette vapor containing nicotine disrupts airway barrier function and induces systemic inflammation and multiorgan fibrosis in mice. Am J Physiol Regul Integr Comp Physiol 2018; 314:R834-R847. [PMID: 29384700 DOI: 10.1152/ajpregu.00270.2017] [Citation(s) in RCA: 158] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Electronic (e)-cigarettes theoretically may be safer than conventional tobacco. However, our prior studies demonstrated direct adverse effects of e-cigarette vapor (EV) on airway cells, including decreased viability and function. We hypothesize that repetitive, chronic inhalation of EV will diminish airway barrier function, leading to inflammatory protein release into circulation, creating a systemic inflammatory state, ultimately leading to distant organ injury and dysfunction. C57BL/6 and CD-1 mice underwent nose only EV exposure daily for 3-6 mo, followed by cardiorenal physiological testing. Primary human bronchial epithelial cells were grown at an air-liquid interface and exposed to EV for 15 min daily for 3-5 days before functional testing. Daily inhalation of EV increased circulating proinflammatory and profibrotic proteins in both C57BL/6 and CD-1 mice: the greatest increases observed were in angiopoietin-1 (31-fold) and EGF (25-fold). Proinflammatory responses were recapitulated by daily EV exposures in vitro of human airway epithelium, with EV epithelium secreting higher IL-8 in response to infection (227 vs. 37 pg/ml, respectively; P < 0.05). Chronic EV inhalation in vivo reduced renal filtration by 20% ( P = 0.017). Fibrosis, assessed by Masson's trichrome and Picrosirius red staining, was increased in EV kidneys (1.86-fold, C57BL/6; 3.2-fold, CD-1; P < 0.05), heart (2.75-fold, C57BL/6 mice; P < 0.05), and liver (1.77-fold in CD-1; P < 0.0001). Gene expression changes demonstrated profibrotic pathway activation. EV inhalation altered cardiovascular function, with decreased heart rate ( P < 0.01), and elevated blood pressure ( P = 0.016). These data demonstrate that chronic inhalation of EV may lead to increased inflammation, organ damage, and cardiorenal and hepatic disease.
Collapse
Affiliation(s)
- Laura E Crotty Alexander
- Pulmonary Critical Care Section, Department of Medicine, Veterans Affairs San Diego Healthcare System, San Diego, California.,Division of Pulmonary Critical Care and Sleep Medicine, Department of Medicine, University of California , San Diego, California
| | | | - Mark Hepokoski
- Pulmonary Critical Care Section, Department of Medicine, Veterans Affairs San Diego Healthcare System, San Diego, California.,Division of Pulmonary Critical Care and Sleep Medicine, Department of Medicine, University of California , San Diego, California
| | - Denzil Mathew
- Pulmonary Critical Care Section, Department of Medicine, Veterans Affairs San Diego Healthcare System, San Diego, California
| | - Alex Moshensky
- Pulmonary Critical Care Section, Department of Medicine, Veterans Affairs San Diego Healthcare System, San Diego, California.,Division of Pulmonary Critical Care and Sleep Medicine, Department of Medicine, University of California , San Diego, California
| | - Andrew Willeford
- Department of Pharmacology, University of California , San Diego, California
| | - Soumita Das
- Department of Pathology, University of California , San Diego, California
| | - Prabhleen Singh
- Division of Nephrology and Hypertension, Department of Medicine, University of California , San Diego, California.,Nephrology Section, Department of Medicine, Veterans Affairs San Diego Healthcare System, San Diego, California
| | - Zach Yong
- Pulmonary Critical Care Section, Department of Medicine, Veterans Affairs San Diego Healthcare System, San Diego, California.,Division of Pulmonary Critical Care and Sleep Medicine, Department of Medicine, University of California , San Diego, California
| | - Jasmine H Lee
- Division of Physiology, Department of Medicine, University of California , San Diego, California
| | - Kevin Vega
- Department of Pathology, University of California , San Diego, California
| | - Ashley Du
- Pulmonary Critical Care Section, Department of Medicine, Veterans Affairs San Diego Healthcare System, San Diego, California.,Division of Pulmonary Critical Care and Sleep Medicine, Department of Medicine, University of California , San Diego, California
| | - John Shin
- Pulmonary Critical Care Section, Department of Medicine, Veterans Affairs San Diego Healthcare System, San Diego, California.,Division of Pulmonary Critical Care and Sleep Medicine, Department of Medicine, University of California , San Diego, California
| | - Christian Javier
- Pulmonary Critical Care Section, Department of Medicine, Veterans Affairs San Diego Healthcare System, San Diego, California.,Division of Pulmonary Critical Care and Sleep Medicine, Department of Medicine, University of California , San Diego, California
| | - Jiang Tian
- Division of Cardiovascular Medicine and Center for Hypertension and Personalized Medicine, University of Toledo , Toledo, Ohio.,Department of Medicine, College of Medicine and Life Sciences, University of Toledo , Toledo, Ohio
| | - Joan Heller Brown
- Department of Pharmacology, University of California , San Diego, California
| | - Ellen C Breen
- Division of Physiology, Department of Medicine, University of California , San Diego, California
| |
Collapse
|
89
|
Liang X, Wang J, Guan R, Zhao L, Li D, Long Z, Yang Q, Xu J, Wang Z, Xie J, Lu W. Limax extract ameliorates cigarette smoke-induced chronic obstructive pulmonary disease in mice. Int Immunopharmacol 2018; 54:210-220. [DOI: 10.1016/j.intimp.2017.11.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Revised: 10/06/2017] [Accepted: 11/03/2017] [Indexed: 01/01/2023]
|
90
|
Smoking-related lung abnormalities on computed tomography images: comparison with pathological findings. Jpn J Radiol 2017; 36:165-180. [DOI: 10.1007/s11604-017-0713-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Accepted: 12/07/2017] [Indexed: 12/17/2022]
|
91
|
Pollard KM, Christy JM, Cauvi DM, Kono DH. Environmental Xenobiotic Exposure and Autoimmunity. CURRENT OPINION IN TOXICOLOGY 2017; 10:15-22. [PMID: 29503968 DOI: 10.1016/j.cotox.2017.11.009] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Susceptibility to autoimmune diseases is dependent on multigenic inheritance, environmental factors, and stochastic events. Although there has been substantial progress in identifying predisposing genetic variants, a significant challenge facing autoimmune disease research is the identification of the specific events that trigger loss of tolerance, autoreactivity and ultimately autoimmune disease. Accordingly, studies have indicated that a wide range of extrinsic factors including drugs, chemicals, microbes, and other environmental factors can induce autoimmunity, particularly systemic autoimmune diseases such as lupus. This review describes a class of environmental factors, namely xenobiotics, epidemiologically linked to human autoimmunity. Mechanisms of xenobiotic autoimmune disease induction are discussed in terms of human and animal model studies with a focus on the role of inflammation and the innate immune response. We argue that localized tissue damage and chronic inflammation elicited by xenobiotic exposure leads to the release of self-antigens and damage-associated molecular patterns as well as the appearance of ectopic lymphoid structures and secondary lymphoid hypertrophy, which provide a milieu for the production of autoreactive B and T cells that contribute to the development and persistence of autoimmunity in predisposed individuals.
Collapse
Affiliation(s)
- K Michael Pollard
- Department of Molecular Medicine, MEM125, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA, USA 92037
| | - Joseph M Christy
- Department of Molecular Medicine, MEM125, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA, USA 92037
| | - David M Cauvi
- Department of Surgery, School of Medicine, University of California, San Diego, 9500 Gilman Drive #0739, La Jolla, CA, USA 92093
| | - Dwight H Kono
- Department of Immunology and Microbiology, IMM310, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA, USA 92037
| |
Collapse
|
92
|
Human lung epithelial cell cultures for analysis of inhaled toxicants: Lessons learned and future directions. Toxicol In Vitro 2017; 47:137-146. [PMID: 29155131 DOI: 10.1016/j.tiv.2017.11.005] [Citation(s) in RCA: 130] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Revised: 09/28/2017] [Accepted: 11/14/2017] [Indexed: 01/01/2023]
Abstract
The epithelium that covers the conducting airways and alveoli is a primary target for inhaled toxic substances, and therefore a focus in inhalation toxicology. The increasing concern about the use of animal models has stimulated the development of in vitro cell culture models for analysis of the biological effects of inhaled toxicants. However, the validity of the current in vitro models and their acceptance by regulatory authorities as an alternative to animal models is a reason for concern, and requires a critical review. In this review, focused on human lung epithelial cell cultures as a model for inhalation toxicology, we discuss the choice of cells for these models, the cell culture system used, the method of exposure as well as the various read-outs to assess the cellular response. We argue that rapid developments in the 3D culture of primary epithelial cells, the use of induced pluripotent stem cells for generation of lung epithelial cells and the development of organ-on-a-chip technology are among the important developments that will allow significant advances in this field. Furthermore, we discuss the various routes of application of inhaled toxicants by air-liquid interface models as well as the vast array of read-outs that may provide essential information. We conclude that close collaboration between researchers from various disciplines is essential for development of valid methods that are suitable for replacement of animal studies for inhalation toxicology.
Collapse
|
93
|
Talio MC, Alesso M, Acosta M, Wills VS, Fernández LP. Sequential determination of nickel and cadmium in tobacco, molasses and refill solutions for e-cigarettes samples by molecular fluorescence. Talanta 2017; 174:221-227. [PMID: 28738572 DOI: 10.1016/j.talanta.2017.06.015] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Revised: 06/01/2017] [Accepted: 06/03/2017] [Indexed: 12/31/2022]
Abstract
In this work, a new procedure was developed for separation and preconcentration of nickel(II) and cadmium(II) in several and varied tobacco samples. Tobacco samples were selected considering the main products consumed by segments of the population, in particular the age (youth) and lifestyle of the consumer. To guarantee representative samples, a randomized strategy of sampling was used. In the first step, a chemofiltration on nylon membrane is carried out employing eosin (Eo) and carbon nanotubes dispersed in sodium dodecylsulfate (SDS) solution (phosphate buffer pH 7). In this condition, Ni(II) was selectively retained on the solid support. After that, the filtrate liquid with Cd(II) was re-conditioned with acetic acid /acetate buffer solution (pH 5) and followed by detection. A spectrofluorimetric determination of both metals was carried out, on the solid support and the filtered aqueous solution, for Ni(II) and Cd(II), respectively. The solid surface fluorescence (SSF) determination was performed at λem = 545nm (λex = 515nm) for Ni(II)-Eo complex and the fluorescence of Cd(II)-Eo was quantified in aqueous solution using λem = 565nm (λex = 540nm). The calibration graphs resulted linear in a range of 0.058-29.35μgL-1 for Ni(II) and 0.124-56.20μgL-1 for Cd(II), with detection limits of 0.019 and 0.041μgL-1 (S/N = 3). The developed methodology shows good sensitivity and adequate selectivity, and it was successfully applied to the determination of trace amounts of nickel and cadmium present in tobacco samples (refill solutions for e-cigarettes, snuff used in narguille (molasses) and traditional tobacco) with satisfactory results. The new methodology was validated by ICP-MS with adequate agreement. The proposed methodology represents a novel fluorescence application to Ni(II) and Cd(II) quantification with sensitivity and accuracy similar to atomic spectroscopies, introducing for the first time the quenching effect on SSF.
Collapse
Affiliation(s)
- María Carolina Talio
- Instituto de Química de San Luis (INQUISAL-CONICET), Chacabuco y Pedernera, 5700 San Luis, Argentina
| | - Magdalena Alesso
- Área de Química Analítica, Facultad de Química, Bioquímica y Farmacia, Universidad Nacional de San Luis, San Luis, Argentina
| | - Mariano Acosta
- Instituto de Química de San Luis (INQUISAL-CONICET), Chacabuco y Pedernera, 5700 San Luis, Argentina
| | | | - Liliana P Fernández
- Área de Química Analítica, Facultad de Química, Bioquímica y Farmacia, Universidad Nacional de San Luis, San Luis, Argentina; Instituto de Química de San Luis (INQUISAL-CONICET), Chacabuco y Pedernera, 5700 San Luis, Argentina.
| |
Collapse
|
94
|
Willinger CM, Rong J, Tanriverdi K, Courchesne PL, Huan T, Wasserman GA, Lin H, Dupuis J, Joehanes R, Jones MR, Chen G, Benjamin EJ, O’Connor GT, Mizgerd JP, Freedman JE, Larson MG, Levy D. MicroRNA Signature of Cigarette Smoking and Evidence for a Putative Causal Role of MicroRNAs in Smoking-Related Inflammation and Target Organ Damage. CIRCULATION. CARDIOVASCULAR GENETICS 2017; 10:e001678. [PMID: 29030400 PMCID: PMC5683429 DOI: 10.1161/circgenetics.116.001678] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Accepted: 07/13/2017] [Indexed: 12/21/2022]
Abstract
BACKGROUND Cigarette smoking increases risk for multiple diseases. MicroRNAs regulate gene expression and may play a role in smoking-induced target organ damage. We sought to describe a microRNA signature of cigarette smoking and relate it to smoking-associated clinical phenotypes, gene expression, and lung inflammatory signaling. METHODS AND RESULTS Expression profiling of 283 microRNAs was conducted on whole blood-derived RNA from 5023 Framingham Heart Study participants (54.0% women; mean age, 55±13 years) using TaqMan assays and high-throughput reverse transcription quantitative polymerase chain reaction. Associations of microRNA expression with smoking status and associations of smoking-related microRNAs with inflammatory biomarkers and pulmonary function were tested with linear mixed effects models. We identified a 6-microRNA signature of smoking. Five of the 6 smoking-related microRNAs were associated with serum levels of C-reactive protein or interleukin-6; miR-1180 was associated with pulmonary function measures at a marginally significant level. Bioinformatic evaluation of smoking-associated genes coexpressed with the microRNA signature of cigarette smoking revealed enrichment for immune-related pathways. Smoking-associated microRNAs altered expression of selected inflammatory mediators in cell culture gain-of-function assays. CONCLUSIONS We characterized a novel microRNA signature of cigarette smoking. The top microRNAs were associated with systemic inflammatory markers and reduced pulmonary function, correlated with expression of genes involved in immune function, and were sufficient to modulate inflammatory signaling. Our results highlight smoking-associated microRNAs and are consistent with the hypothesis that smoking-associated microRNAs serve as mediators of smoking-induced inflammation and target organ damage. These findings call for further mechanistic studies to explore the diagnostic and therapeutic use of smoking-related microRNAs.
Collapse
Affiliation(s)
- Christine M. Willinger
- Framingham Heart Study, Framingham, MA
- Division of Intramural Research and Population Sciences Branch, National Heart, Lung, and Blood Institute, Bethesda, MD
| | - Jian Rong
- Framingham Heart Study, Framingham, MA
- Boston University School of Public Health, Boston
| | - Kahraman Tanriverdi
- Department of Medicine and UMass Memorial Heart & Vascular Center, University of Massachusetts Medical School, Worcester
| | - Paul L. Courchesne
- Framingham Heart Study, Framingham, MA
- Division of Intramural Research and Population Sciences Branch, National Heart, Lung, and Blood Institute, Bethesda, MD
| | - Tianxiao Huan
- Framingham Heart Study, Framingham, MA
- Division of Intramural Research and Population Sciences Branch, National Heart, Lung, and Blood Institute, Bethesda, MD
| | | | - Honghuang Lin
- Framingham Heart Study, Framingham, MA
- Boston University School of Medicine
| | - Josée Dupuis
- Framingham Heart Study, Framingham, MA
- Boston University School of Public Health, Boston
| | - Roby Joehanes
- Framingham Heart Study, Framingham, MA
- Division of Intramural Research and Population Sciences Branch, National Heart, Lung, and Blood Institute, Bethesda, MD
- Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA
| | | | - George Chen
- Framingham Heart Study, Framingham, MA
- Division of Intramural Research and Population Sciences Branch, National Heart, Lung, and Blood Institute, Bethesda, MD
| | - Emelia J. Benjamin
- Framingham Heart Study, Framingham, MA
- Boston University School of Public Health, Boston
- Boston University School of Medicine
| | | | | | - Jane E. Freedman
- Department of Medicine and UMass Memorial Heart & Vascular Center, University of Massachusetts Medical School, Worcester
| | - Martin G. Larson
- Framingham Heart Study, Framingham, MA
- Boston University School of Public Health, Boston
| | - Daniel Levy
- Framingham Heart Study, Framingham, MA
- Division of Intramural Research and Population Sciences Branch, National Heart, Lung, and Blood Institute, Bethesda, MD
- Boston University School of Medicine
| |
Collapse
|
95
|
Toledo-Arruda AC, Vieira RP, Guarnier FA, Suehiro CL, Caleman-Neto A, Olivo CR, Arantes PMM, Almeida FM, Lopes FDTQS, Ramos EMC, Cecchini R, Lin CJ, Martins MA. Time-course effects of aerobic physical training in the prevention of cigarette smoke-induced COPD. J Appl Physiol (1985) 2017; 123:674-683. [PMID: 28729393 DOI: 10.1152/japplphysiol.00819.2016] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Revised: 07/13/2017] [Accepted: 07/13/2017] [Indexed: 11/22/2022] Open
Abstract
A previous study by our group showed that regular exercise training (ET) attenuated pulmonary injury in an experimental model of chronic exposure to cigarette smoke (CS) in mice, but the time-course effects of the mechanisms involved in this protection remain poorly understood. We evaluated the temporal effects of regular ET in an experimental model of chronic CS exposure. Male C57BL/6 mice were divided into four groups: Control (sedentary + air), Exercise (aerobic training + air), Smoke (sedentary + smoke), and Smoke + Exercise (aerobic training + smoke). Mice were exposed to CS and ET for 4, 8, or 12 wk. Exercise protected mice exposed to CS from emphysema and reductions in tissue damping and tissue elastance after 12 wk (P < 0.01). The total number of inflammatory cells in the bronchoalveolar lavage increased in the Smoke group, mainly due to the recruitment of macrophages after 4 wk, neutrophils and lymphocytes after 8 wk, and lymphocytes and macrophages after 12 wk (P < 0.01). Exercise attenuated this increase in mice exposed to CS. The protection conferred by exercise was mainly observed after exercise adaptation. Exercise increased IL-6 and IL-10 in the quadriceps and lungs (P < 0.05) after 12 wk. Total antioxidant capacity and SOD was increased and TNF-α and oxidants decreased in lungs of mice exposed to CS after 12 wk (P < 0.05). The protective effects of exercise against lung injury induced by cigarette smoke exposure suggests that anti-inflammatory mediators and antioxidant enzymes play important roles in chronic obstructive pulmonary disease development mainly after the exercise adaptation.NEW & NOTEWORTHY These experiments investigated for the first time the temporal effects of regular moderate exercise training in cigarette smoke-induced chronic obstructive pulmonary disease. We demonstrate that aerobic conditioning had a protective effect in emphysema development induced by cigarette smoke exposure. This effect was most likely secondary to an effect of exercise on oxidant-antioxidant balance and anti-inflammatory mediators.
Collapse
Affiliation(s)
| | - Rodolfo P Vieira
- Brazilian Institute of Teaching and Research in Pulmonary and Exercise Immunology, School of Medical Sciences Humanitas, Universidade Brasil and Laboratory of Pulmonary and Exercise Immunology, Nove de Julho University, Sao Paulo, Brazil
| | - Flávia A Guarnier
- Department of Pathology, Londrina State University, Londrina, Brazil; and
| | - Camila L Suehiro
- Department of Clinical Medicine, School of Medicine, University of Sao Paulo, Sao Paulo, Brazil
| | - Agostinho Caleman-Neto
- Department of Clinical Medicine, School of Medicine, University of Sao Paulo, Sao Paulo, Brazil
| | - Clarice R Olivo
- Department of Clinical Medicine, School of Medicine, University of Sao Paulo, Sao Paulo, Brazil
| | - Petra M M Arantes
- Department of Clinical Medicine, School of Medicine, University of Sao Paulo, Sao Paulo, Brazil
| | - Francine M Almeida
- Department of Clinical Medicine, School of Medicine, University of Sao Paulo, Sao Paulo, Brazil
| | - Fernanda D T Q S Lopes
- Department of Clinical Medicine, School of Medicine, University of Sao Paulo, Sao Paulo, Brazil
| | - Ercy M C Ramos
- Department of Physiotherapy, State University of Sao Paulo, Presidente Prudente, Brazil
| | - Rubens Cecchini
- Department of Pathology, Londrina State University, Londrina, Brazil; and
| | - Chin Jia Lin
- Department of Pathology, School of Medicine, University of Sao Paulo, Sao Paulo, Brazil
| | - Milton Arruda Martins
- Department of Clinical Medicine, School of Medicine, University of Sao Paulo, Sao Paulo, Brazil
| |
Collapse
|
96
|
Torrelles JB, Schlesinger LS. Integrating Lung Physiology, Immunology, and Tuberculosis. Trends Microbiol 2017; 25:688-697. [PMID: 28366292 PMCID: PMC5522344 DOI: 10.1016/j.tim.2017.03.007] [Citation(s) in RCA: 81] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2016] [Revised: 03/03/2017] [Accepted: 03/10/2017] [Indexed: 11/17/2022]
Abstract
Lungs are directly exposed to the air, have enormous surface area, and enable gas exchange in air-breathing animals. They are constantly 'attacked' by microbes from both outside and inside and thus possess a unique, highly regulated local immune defense system which efficiently allows for microbial clearance while minimizing damaging inflammatory responses. As a prototypic host-adapted airborne pathogen, Mycobacterium tuberculosis traverses the lung and has several 'interaction points' (IPs) which it must overcome to cause infection. These interactions are critical, not only from a pathogenesis perspective but also in considering the effectiveness of therapies and vaccines in the lungs. Here we discuss emerging views on immunologic interactions occurring in the lungs for M. tuberculosis and their impact on infection and persistence.
Collapse
Affiliation(s)
- Jordi B Torrelles
- Department of Microbial Infection and Immunity, College of Medicine, and the Center for Microbial Interface Biology, The Ohio State University, Columbus, OH 43210, USA.
| | - Larry S Schlesinger
- Department of Microbial Infection and Immunity, College of Medicine, and the Center for Microbial Interface Biology, The Ohio State University, Columbus, OH 43210, USA.
| |
Collapse
|
97
|
Shields PG, Berman M, Brasky TM, Freudenheim JL, Mathe E, McElroy JP, Song MA, Wewers MD. A Review of Pulmonary Toxicity of Electronic Cigarettes in the Context of Smoking: A Focus on Inflammation. Cancer Epidemiol Biomarkers Prev 2017; 26:1175-1191. [PMID: 28642230 PMCID: PMC5614602 DOI: 10.1158/1055-9965.epi-17-0358] [Citation(s) in RCA: 81] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2017] [Revised: 05/22/2017] [Accepted: 05/24/2017] [Indexed: 12/30/2022] Open
Abstract
The use of electronic cigarettes (e-cigs) is increasing rapidly, but their effects on lung toxicity are largely unknown. Smoking is a well-established cause of lung cancer and respiratory disease, in part through inflammation. It is plausible that e-cig use might affect similar inflammatory pathways. E-cigs are used by some smokers as an aid for quitting or smoking reduction, and by never smokers (e.g., adolescents and young adults). The relative effects for impacting disease risk may differ for these groups. Cell culture and experimental animal data indicate that e-cigs have the potential for inducing inflammation, albeit much less than smoking. Human studies show that e-cig use in smokers is associated with substantial reductions in blood or urinary biomarkers of tobacco toxicants when completely switching and somewhat for dual use. However, the extent to which these biomarkers are surrogates for potential lung toxicity remains unclear. The FDA now has regulatory authority over e-cigs and can regulate product and e-liquid design features, such as nicotine content and delivery, voltage, e-liquid formulations, and flavors. All of these factors may impact pulmonary toxicity. This review summarizes current data on pulmonary inflammation related to both smoking and e-cig use, with a focus on human lung biomarkers. Cancer Epidemiol Biomarkers Prev; 26(8); 1175-91. ©2017 AACR.
Collapse
Affiliation(s)
- Peter G Shields
- Comprehensive Cancer Center, The Ohio State University and James Cancer Hospital, and College of Medicine, Columbus, Ohio.
| | - Micah Berman
- Comprehensive Cancer Center, The Ohio State University and James Cancer Hospital, and College of Public Health, Ohio
| | - Theodore M Brasky
- Comprehensive Cancer Center, The Ohio State University and James Cancer Hospital, and College of Medicine, Columbus, Ohio
| | - Jo L Freudenheim
- Department of Epidemiology and Environmental Health, School of Public Health and Health Professions, University at Buffalo, Buffalo, New York
| | - Ewy Mathe
- Department of Biomedical Informatics, The Ohio State University, Columbus, Ohio
| | - Joseph P McElroy
- Center for Biostatistics, Department of Biomedical Informatics, The Ohio State University, Columbus, Ohio
| | - Min-Ae Song
- Comprehensive Cancer Center, The Ohio State University and James Cancer Hospital, and College of Medicine, Columbus, Ohio
| | - Mark D Wewers
- Department of Internal Medicine, The Ohio State University, Columbus, Ohio
| |
Collapse
|
98
|
Rac1 signaling regulates cigarette smoke-induced inflammation in the lung via the Erk1/2 MAPK and STAT3 pathways. Biochim Biophys Acta Mol Basis Dis 2017; 1863:1778-1788. [DOI: 10.1016/j.bbadis.2017.04.013] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Revised: 03/24/2017] [Accepted: 04/16/2017] [Indexed: 02/06/2023]
|
99
|
Intracellular and Extracellular Cytokines in A549 Cells and THP1 Cells Exposed to Cigarette Smoke. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 910:39-45. [PMID: 26987337 DOI: 10.1007/5584_2016_214] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Cigarette smoke (CS) activates inflammatory cells and increases cytokine levels producing local and systemic inflammation. To assess changes in intracellular and extracellular cytokine levels we used human epithelial (A549 cells) and monocyte (THP-1) cell lines grown for 24 h in cigarette smoke-conditioned media. Cytokines were assessed using immunostaining/flow cytometry and ELISA assay. In THP1cells, grown in CS-conditioned media, the intracellular interleukins IL-1β, IL-6, and IL-10 increased by more than tenfold, while less significant increases were found in A549 cells. IL-1α and IL-1β, but not IL-6 or IL-10, were increased in the culture media, while IL-2 was raised by about fivefold only in the culture medium of A549 cells. IL-4, IL-6, IL-8, IL-10, IL-12, and tumor necrosis factor alpha were undetectable, while only a slight increase was observed in extracellular IL-17A (by about 60 %) in the medium of A549 cells and by about 115 % in the medium of THP1 cells. The interferon gamma (IFNγ) was increased by about eightfold, but only in the medium of THP1 cells grown with CS. We conclude that IL-1 and INFγ are the key cytokines responsible for pro-inflammatory signaling in epithelial cells and monocytes, respectively, exposed to cigarette smoke.
Collapse
|
100
|
Jourde-Chiche N, Whalen E, Gondouin B, Speake C, Gersuk V, Dussol B, Burtey S, Pascual V, Chaussabel D, Chiche L. Modular transcriptional repertoire analyses identify a blood neutrophil signature as a candidate biomarker for lupus nephritis. Rheumatology (Oxford) 2017; 56:477-487. [PMID: 28031441 DOI: 10.1093/rheumatology/kew439] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2016] [Indexed: 01/09/2023] Open
Abstract
Objective LN is a severe complication of SLE. Non-invasive biomarkers are needed for identifying patients at risk of a renal flare, for differentiating proliferative from non-proliferative forms and for assessing prognoses for LN. Methods We assessed the link between blood transcriptional signatures and LN using blood samples from patients with biopsy-proven LN, extra-renal SLE flares or quiescent SLE. Healthy controls, and control patients with glomerular diseases or bacterial sepsis were included. Modular repertoire analyses from microarray data were confirmed by PCR. Results A modular neutrophil signature (upregulation of module M5.15) was present in 65% of SLE patients and was strongly associated with LN. M5.15 activity was stronger in LN than in extra-renal flares (88 vs 17%). M5.15 was neither correlated to IFN modules, nor to SLEDAI or anti-dsDNA antibodies, but moderately to CS dose. M5.15 activity was associated with severity of LN, was stronger when proliferative, and decreased in patients responding to treatment. M5.15 activation was not caused by higher CS dose because it correlated only moderately to neutrophil count and was also observed among quiescent patients. Among quiescent patients, those with a past history of LN had higher M5.15 activity (50 vs 8%). M5.15 activation was present in patients with bacterial sepsis or ANCA-associated vasculitis, but not in patients with other glomerular diseases. Overall, M5.15 activation was associated with past, present or future flares of LN. Conclusion Modular neutrophil signature could be a biomarker for stratifying LN risk and for monitoring its response to treatment. Trial registration ClinicalTrials.gov, http://clinicaltrials.gov , NCT00920114.
Collapse
Affiliation(s)
- Noémie Jourde-Chiche
- Department of Nephrology, Aix-Marseille University, AP-HM, Hôpital Conception, UMR_S 1076, Vascular Research Center of Marseille, Marseille, France
| | - Elizabeth Whalen
- Systems Immunology Department, Benaroya Research Institute, Seattle
| | - Bertrand Gondouin
- Department of Nephrology, Aix-Marseille University, AP-HM, Hôpital Conception, UMR_S 1076, Vascular Research Center of Marseille, Marseille, France
| | - Cate Speake
- Systems Immunology Department, Benaroya Research Institute, Seattle
| | - Vivian Gersuk
- Systems Immunology Department, Benaroya Research Institute, Seattle
| | - Bertrand Dussol
- Department of Nephrology, Aix-Marseille University, AP-HM, Hôpital Conception, UMR_S 1076, Vascular Research Center of Marseille, Marseille, France
| | - Stephane Burtey
- Department of Nephrology, Aix-Marseille University, AP-HM, Hôpital Conception, UMR_S 1076, Vascular Research Center of Marseille, Marseille, France
| | - Virginia Pascual
- Immunology, Baylor Institute for Immunology Research, Dallas, TX, USA
| | - Damien Chaussabel
- Systems Biology Department, Sidra Medical and Research Center, Doha, Qatar
| | - Laurent Chiche
- Department of Internal Medicine, Hôpital Européen, Marseille, France
| |
Collapse
|