1
|
Ajenu EO, Seideneck AM, Pandellapalli E, Shinsky EM, Humphries CL, Aparicio NL, Sharma M, Marden JH, Krasilnikova MM. ABCG2 transporter reduces protein aggregation in cigarette smoke condensate-exposed A549 lung cancer cells. PLoS One 2024; 19:e0297661. [PMID: 38442133 PMCID: PMC10914296 DOI: 10.1371/journal.pone.0297661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Accepted: 01/09/2024] [Indexed: 03/07/2024] Open
Abstract
Cigarette smoke-induced protein aggregation damages the lung cells in emphysema and COPD; however, lung cancer cells continue to thrive, evolving to persist in the toxic environment. Here, we showed that upon the cigarette smoke condensate exposure, A549 lung cancer cells exhibit better survival and reduced level of protein aggregation when compared to non-cancerous Beas-2B and H-6053 cells. Our data suggests that upregulation of efflux pumps in cancer cells assists in reducing smoke toxicity. Specifically, we demonstrated that inhibition of the ABCG2 transporter in A549 by febuxostat or its downregulation by shRNA-mediated RNA interference resulted in a significant increase in protein aggregation due to smoke exposure.
Collapse
Affiliation(s)
- Emmanuella O. Ajenu
- Department of Biochemistry and Molecular Biology, Penn State University, University Park, Pennsylvania, United states of America
| | - Ashley M. Seideneck
- Department of Biochemistry and Molecular Biology, Penn State University, University Park, Pennsylvania, United states of America
| | - Esh Pandellapalli
- Department of Biochemistry and Molecular Biology, Penn State University, University Park, Pennsylvania, United states of America
| | - Emily M. Shinsky
- Department of Biochemistry and Molecular Biology, Penn State University, University Park, Pennsylvania, United states of America
| | - Casey L. Humphries
- Department of Biochemistry and Molecular Biology, Penn State University, University Park, Pennsylvania, United states of America
| | - Nicholas L. Aparicio
- Department of Biochemistry and Molecular Biology, Penn State University, University Park, Pennsylvania, United states of America
| | - Mahak Sharma
- Department of Biochemistry and Molecular Biology, Penn State University, University Park, Pennsylvania, United states of America
| | - James H. Marden
- Department of Biology, Penn State University, University Park, Pennsylvania, United states of America
| | - Maria M. Krasilnikova
- Department of Biochemistry and Molecular Biology, Penn State University, University Park, Pennsylvania, United states of America
| |
Collapse
|
2
|
Lai EY, Huang YT. Genome-wide multimediator analyses using the generalized Berk-Jones statistics with the composite test. Bioinformatics 2023; 39:btad544. [PMID: 37665751 PMCID: PMC10500087 DOI: 10.1093/bioinformatics/btad544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 07/18/2023] [Accepted: 09/01/2023] [Indexed: 09/06/2023] Open
Abstract
MOTIVATION Mediation analysis is performed to evaluate the effects of a hypothetical causal mechanism that marks the progression from an exposure, through mediators, to an outcome. In the age of high-throughput technologies, it has become routine to assess numerous potential mechanisms at the genome or proteome scales. Alongside this, the necessity to address issues related to multiple testing has also arisen. In a sparse scenario where only a few genes or proteins are causally involved, conventional methods for assessing mediation effects lose statistical power because the composite null distribution behind this experiment cannot be attained. The power loss hence decreases the true mechanisms identified after multiple testing corrections. To fairly delineate a uniform distribution under the composite null, Huang (Genome-wide analyses of sparse mediation effects under composite null hypotheses. Ann Appl Stat 2019a;13:60-84; AoAS) proposed the composite test to provide adjusted P-values for single-mediator analyses. RESULTS Our contribution is to extend the method to multimediator analyses, which are commonly encountered in genomic studies and also flexible to various biological interests. Using the generalized Berk-Jones statistics with the composite test, we proposed a multivariate approach that favors dense and diverse mediation effects, a decorrelation approach that favors sparse and consistent effects, and a hybrid approach that captures the edges of both approaches. Our analysis suite has been implemented as an R package MACtest. The utility is demonstrated by analyzing the lung adenocarcinoma datasets from The Cancer Genome Atlas and Clinical Proteomic Tumor Analysis Consortium. We further investigate the genes and networks whose expression may be regulated by smoking-induced epigenetic aberrations. AVAILABILITY AND IMPLEMENTATION An R package MACtest is available on https://github.com/roqe/MACtest.
Collapse
Affiliation(s)
- En-Yu Lai
- Institute of Statistical Science, Academia Sinica, Nankang, Taipei 11529, Taiwan
| | - Yen-Tsung Huang
- Institute of Statistical Science, Academia Sinica, Nankang, Taipei 11529, Taiwan
| |
Collapse
|
3
|
Soerensen SBT, Nagy D, Ødum N, Iversen L, Lindahl LM. Chronic Obstructive Pulmonary Disease and Risk of Cutaneous T-cell Lymphoma: A Danish Population-based Cohort Study. Acta Derm Venereol 2023; 103:adv5238. [PMID: 37606154 PMCID: PMC10461176 DOI: 10.2340/actadv.v103.5238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Accepted: 05/09/2023] [Indexed: 08/23/2023] Open
Abstract
Abstract is missing (Short communication)
Collapse
Affiliation(s)
| | - Dávid Nagy
- Department of Clinical Epidemiology, Aarhus University Hospital and Aarhus University, Aarhus, Denmark
| | - Niels Ødum
- Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Lars Iversen
- Department of Dermatology, Aarhus University Hospital, Aarhus, Denmark
| | - Lise M Lindahl
- Department of Dermatology, Aarhus University Hospital, Aarhus, Denmark
| |
Collapse
|
4
|
The Tobacco Smoke Component, Acrolein, as a Major Culprit in Lung Diseases and Respiratory Cancers: Molecular Mechanisms of Acrolein Cytotoxic Activity. Cells 2023; 12:cells12060879. [PMID: 36980220 PMCID: PMC10047238 DOI: 10.3390/cells12060879] [Citation(s) in RCA: 23] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 03/05/2023] [Accepted: 03/08/2023] [Indexed: 03/18/2023] Open
Abstract
Acrolein, a highly reactive unsaturated aldehyde, is a ubiquitous environmental pollutant that seriously threatens human health and life. Due to its high reactivity, cytotoxicity and genotoxicity, acrolein is involved in the development of several diseases, including multiple sclerosis, neurodegenerative diseases such as Alzheimer’s disease, cardiovascular and respiratory diseases, diabetes mellitus and even the development of cancer. Traditional tobacco smokers and e-cigarette users are particularly exposed to the harmful effects of acrolein. High concentrations of acrolein have been found in both mainstream and side-stream tobacco smoke. Acrolein is considered one of cigarette smoke’s most toxic and harmful components. Chronic exposure to acrolein through cigarette smoke has been linked to the development of asthma, acute lung injury, chronic obstructive pulmonary disease (COPD) and even respiratory cancers. This review addresses the current state of knowledge on the pathological molecular mechanisms of acrolein in the induction, course and development of lung diseases and cancers in smokers.
Collapse
|
5
|
Liu C, Li P, Zheng J, Wang Y, Wu W, Liu X. Role of necroptosis in airflow limitation in chronic obstructive pulmonary disease: focus on small-airway disease and emphysema. Cell Death Dis 2022; 8:363. [PMID: 35973987 PMCID: PMC9381515 DOI: 10.1038/s41420-022-01154-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 07/31/2022] [Accepted: 08/01/2022] [Indexed: 11/09/2022]
Abstract
Airflow limitation with intractable progressive mechanisms is the main disease feature of chronic obstructive pulmonary disease (COPD). The pathological process of airflow limitation in COPD involves necroptosis, a form of programmed necrotic cell death with pro-inflammatory properties. In this paper, the correlations of small-airway disease and emphysema with airflow limitation in COPD were firstly reviewed; then, based on this, the effects of necroptosis on small-airway disease and emphysema were analysed, and the possible mechanisms of necroptosis causing airflow limitation in COPD were explored. The results showed that airflow limitation is caused by a combination of small-airway disease and emphysema. In addition, toxic particulate matter stimulates epithelial cells to trigger necroptosis, and necroptosis promotes the expulsion of cell contents, the abnormal hyperplasia of pro-inflammatory mediators and the insufficient clearance of dead cells by macrophages; these processes, coupled with the interaction of necroptosis and oxidative stress, collectively result in small-airway disease and emphysema in COPD.
Collapse
Affiliation(s)
- Chanjing Liu
- Department of Sports Rehabilitation, Shanghai University of Sport, Shanghai, People's Republic of China
| | - Peijun Li
- Department of Sports Rehabilitation, Shanghai University of Sport, Shanghai, People's Republic of China
| | - Jiejiao Zheng
- Department of Rehabilitation Medicine, Huadong Hospital, Shanghai, People's Republic of China
| | - Yingqi Wang
- Department of Sports Rehabilitation, Shanghai University of Sport, Shanghai, People's Republic of China
| | - Weibing Wu
- Department of Sports Rehabilitation, Shanghai University of Sport, Shanghai, People's Republic of China.
| | - Xiaodan Liu
- School of Rehabilitation Science, Shanghai University of Traditional Chinese Medicine, Shanghai, People's Republic of China.
| |
Collapse
|
6
|
He J, Pang Q, Huang C, Xie J, Hu J, Wang L, Wang C, Meng L, Fan R. Environmental dose of 16 priority-controlled PAHs mixture induce damages of vascular endothelial cells involved in oxidative stress and inflammation. Toxicol In Vitro 2021; 79:105296. [PMID: 34896602 DOI: 10.1016/j.tiv.2021.105296] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2021] [Revised: 11/24/2021] [Accepted: 12/06/2021] [Indexed: 12/30/2022]
Abstract
Epidemiological studies have shown that cardiovascular diseases caused by PM2.5 pollution account for the second death rate in China. Polycyclic aromatic hydrocarbons (PAHs) are one important group of persistent organic pollutants absorbed on PM2.5. Though individual PAH is related to vascular disease, the relationship between environmental PAHs exposure and vascular damages is still unclear. To explore the effect of PAHs on blood vessel, human umbilical vein endothelial cells (HUVECs) are treated with 16 priority-controlled PAHs at various concentrations to study their cytotoxicity and morphological alteration. Results showed that, after 48 h treatment, PAHs mixture generally attenuated the ability of wound healing, transwell migration and tube formation of HUVECs (p < 0.01) except for 1 × PAHs in transwell migration. Moreover, PAHs increased the levels of ROS and 8-hydroxy-2'-deoxyguanosine (p < 0.05), indicating that it exceeded the scavenging ability of superoxide dismutase activity. However, PAHs mixture did not increase apoptosis rate, which may be attribute to the difference of PAH concentration and composition between this study and previous reports. Downstream signaling cascades significantly and generally upregulated the relative expression of proteins in Nrf2/HO-1 and NF-ƙB/TNF-α pathway with the activation of oxidative stress, including HO-, TNF-α and Nrf2. In summary, this study suggests that environmental mixture of 16 priority-controlled PAHs can induce the damages of vascular endothelial cells involved in cellular oxidative stress and inflammation.
Collapse
Affiliation(s)
- Jiaying He
- Guangzhou Key Laboratory of Subtropical Biodiversity and Biomonitoring, School of Life Sciences, South China Normal University, Guangzhou 510631, China
| | - Qihua Pang
- Guangzhou Key Laboratory of Subtropical Biodiversity and Biomonitoring, School of Life Sciences, South China Normal University, Guangzhou 510631, China; Guangdong Provincial Engineering Technology Research Center for Drug and Food Biological Resources Processing and Comprehensive Utilization, School of Life Sciences, South China Normal University, Guangzhou 510631, China
| | - Chengmeng Huang
- Guangzhou Key Laboratory of Subtropical Biodiversity and Biomonitoring, School of Life Sciences, South China Normal University, Guangzhou 510631, China
| | - Jiaqi Xie
- Guangzhou Key Laboratory of Subtropical Biodiversity and Biomonitoring, School of Life Sciences, South China Normal University, Guangzhou 510631, China
| | - Jindian Hu
- Guangzhou Key Laboratory of Subtropical Biodiversity and Biomonitoring, School of Life Sciences, South China Normal University, Guangzhou 510631, China
| | - Lei Wang
- Guangzhou Key Laboratory of Subtropical Biodiversity and Biomonitoring, School of Life Sciences, South China Normal University, Guangzhou 510631, China
| | - Congcong Wang
- Guangzhou Key Laboratory of Subtropical Biodiversity and Biomonitoring, School of Life Sciences, South China Normal University, Guangzhou 510631, China
| | - Lingxue Meng
- Guangzhou Key Laboratory of Subtropical Biodiversity and Biomonitoring, School of Life Sciences, South China Normal University, Guangzhou 510631, China
| | - Ruifang Fan
- Guangzhou Key Laboratory of Subtropical Biodiversity and Biomonitoring, School of Life Sciences, South China Normal University, Guangzhou 510631, China; Guangdong Provincial Engineering Technology Research Center for Drug and Food Biological Resources Processing and Comprehensive Utilization, School of Life Sciences, South China Normal University, Guangzhou 510631, China; Guangdong Provincial Key Laboratory of Chemical Pollution and Environmental Safety, South China Normal University, Guangzhou 510006, China.
| |
Collapse
|
7
|
Burgoyne RA, Fisher AJ, Borthwick LA. The Role of Epithelial Damage in the Pulmonary Immune Response. Cells 2021; 10:cells10102763. [PMID: 34685744 PMCID: PMC8534416 DOI: 10.3390/cells10102763] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 10/05/2021] [Accepted: 10/12/2021] [Indexed: 12/14/2022] Open
Abstract
Pulmonary epithelial cells are widely considered to be the first line of defence in the lung and are responsible for coordinating the innate immune response to injury and subsequent repair. Consequently, epithelial cells communicate with multiple cell types including immune cells and fibroblasts to promote acute inflammation and normal wound healing in response to damage. However, aberrant epithelial cell death and damage are hallmarks of pulmonary disease, with necrotic cell death and cellular senescence contributing to disease pathogenesis in numerous respiratory diseases such as idiopathic pulmonary fibrosis (IPF), chronic obstructive pulmonary disease (COPD) and coronavirus disease (COVID)-19. In this review, we summarise the literature that demonstrates that epithelial damage plays a pivotal role in the dysregulation of the immune response leading to tissue destruction and abnormal remodelling in several chronic diseases. Specifically, we highlight the role of epithelial-derived damage-associated molecular patterns (DAMPs) and senescence in shaping the immune response and assess their contribution to inflammatory and fibrotic signalling pathways in the lung.
Collapse
Affiliation(s)
- Rachel Ann Burgoyne
- Fibrosis Research Group, Biosciences Institute, Newcastle University, Newcastle upon Tyne NE2 4HH, UK;
| | - Andrew John Fisher
- Regenerative Medicine, Stem Cells and Transplantation Theme, Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne NE2 4HH, UK;
- Institute of Transplantation, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne NE7 7DN, UK
| | - Lee Anthony Borthwick
- Fibrosis Research Group, Biosciences Institute, Newcastle University, Newcastle upon Tyne NE2 4HH, UK;
- Fibrofind, Medical School, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
- Correspondence: ; Tel.: +44-191-208-3112
| |
Collapse
|
8
|
Impact of cigarette versus electronic cigarette aerosol conditioned media on aortic endothelial cells in a microfluidic cardiovascular model. Sci Rep 2021; 11:4747. [PMID: 33637800 PMCID: PMC7910588 DOI: 10.1038/s41598-021-83511-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Accepted: 02/03/2021] [Indexed: 11/16/2022] Open
Abstract
Atherosclerosis is a complex process involving progressive pathological events, including monocyte adhesion to the luminal endothelial surface. We have developed a functional in vitro adhesion assay using BioFlux microfluidic technology to investigate THP-1 (human acute monocytic leukaemia cell) monocyte adhesion to human aortic endothelial cells (HAECs). The effect of whole smoke conditioned media (WSCM) generated from University of Kentucky reference cigarette 3R4F, electronic cigarette vapour conditioned media (eVCM) from an electronic nicotine delivery system (ENDS) product (Vype ePen) and nicotine on monocyte adhesion to HAECs was evaluated. Endothelial monolayers were grown in microfluidic channels and exposed to 0–1500 ng/mL nicotine or nicotine equivalence of WSCM or eVCM for 24 h. Activated THP-1 cells were perfused through the channels and a perfusion, adhesion period and wash cycle performed four times with increasing adhesion period lengths (10, 20, 30 and 40 min). THP-1 cell adhesion was quantified by counting adherent cells. WSCM induced dose-dependent increases in monocyte adhesion compared to vehicle control. No such increases were observed for eVCM or nicotine. Adhesion regulation was linked to increased ICAM-1 protein expression. Staining of ICAM-1 in HAECs and CD11b (MAC-1) in THP-1 cells demonstrated adhesion molecule co-localisation in BioFlux plates. The ICAM-1 adhesion response to WSCM was downregulated by transfecting HAECs with ICAM-1 siRNA. We conclude that the BioFlux system is able to model human monocyte adhesion to primary human endothelial cells in vitro and WSCM drives the greatest increase in monocyte adhesion via a mechanism involving endothelial ICAM-1 expression.
Collapse
|
9
|
Scharf P, da Rocha GHO, Sandri S, Heluany CS, Pedreira Filho WR, Farsky SHP. Immunotoxic mechanisms of cigarette smoke and heat-not-burn tobacco vapor on Jurkat T cell functions. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2021; 268:115863. [PMID: 33126161 DOI: 10.1016/j.envpol.2020.115863] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 09/29/2020] [Accepted: 10/14/2020] [Indexed: 06/11/2023]
Abstract
Cigarette smoke (CS) affects immune functions, leading to severe outcomes in smokers. Robust evidence addresses the immunotoxic effects of combustible tobacco products. As heat-not-burn tobacco products (HNBT) vaporize lower levels of combustible products, we here compared the effects of cigarette smoke (CS) and HNBT vapor on Jurkat T cells. Cells were exposed to air, conventional cigarettes or heatsticks of HNBT for 30 min and were stimulated or not with phorbol myristate acetate (PMA). Cell viability, proliferation, reactive oxygen species (ROS) production, 8-OHdG, MAP-kinases and nuclear factor κB (NFκB) activation and metallothionein expression (MTs) were assessed by flow cytometry; nitric oxide (NO) and cytokine levels were measured by Griess reaction and ELISA, respectively. Levels of metals in the exposure chambers were quantified by inductively coupled plasma mass spectrometry. MT expressions were quantified by immunohistochemistry in the lungs and liver of C57Bl/6 mice exposed to CS, HNBT or air (1 h, twice a day for five days: via inhalation). While both CS and HBNT exposures increased cell death, CS led to a higher number of necrotic cells, increased the production of ROS, NO, inflammatory cytokines and MTs when compared to HNBT-exposed cells, and led to a higher expression of MTs in mice. CS released higher amounts of metals. CS and HNBT exposures decreased PMA-induced interleukin-2 (IL-2) secretion and impaired Jurkat proliferation, effects also seen in cells exposed to nicotine. Although HNBT vapor does not activate T cells as CS does, exposure to both HNBT and CS suppressed proliferation and IL-2 release, a pivotal cytokine involved with T cell proliferation and tolerance, and this effect may be related to nicotine content in both products.
Collapse
Affiliation(s)
- Pablo Scharf
- Department of Clinical & Toxicological Analyses, School of Pharmaceutical Sciences, University of Sao Paulo, SP, Brazil
| | - Gustavo H O da Rocha
- Department of Clinical & Toxicological Analyses, School of Pharmaceutical Sciences, University of Sao Paulo, SP, Brazil
| | - Silvana Sandri
- Department of Clinical & Toxicological Analyses, School of Pharmaceutical Sciences, University of Sao Paulo, SP, Brazil
| | - Cintia S Heluany
- Department of Clinical & Toxicological Analyses, School of Pharmaceutical Sciences, University of Sao Paulo, SP, Brazil
| | - Walter R Pedreira Filho
- Fundação Jorge Duprat Figueiredo de Segurança e Medicina do Trabalho, Ministério do Trabalho e Previdência Social, Sao Paulo, SP, Brazil
| | - Sandra H P Farsky
- Department of Clinical & Toxicological Analyses, School of Pharmaceutical Sciences, University of Sao Paulo, SP, Brazil.
| |
Collapse
|
10
|
Rodríguez-Fernández P, Gómez AC, Gibert I, Prat-Aymerich C, Domínguez J. Effects of cigarette smoke on the administration of isoniazid and rifampicin to macrophages infected with Mycobacterium tuberculosis. Exp Lung Res 2020; 47:87-97. [PMID: 33305652 DOI: 10.1080/01902148.2020.1854371] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
BACKGROUND Smoking is a cause behind many diseases, including tuberculosis, and it is a risk factor for tuberculosis infection and mortality. Moreover, smoking is associated with a poor tuberculosis treatment outcome. OBJECTIVES In this study, we focus on the effects of cigarette smoke on an infected cell culture treated with anti-tuberculosis drugs. MATERIALS AND METHODS Cytotoxicity on THP-1, J774A.1 and MH-S cell lines and growth of Mycobacterium tuberculosis exposed to a reference or a commercial cigarette was evaluated. THP-1 cell line was exposed to cigarette smoke, infected with Mycobacterium tuberculosis and treated with anti-tuberculosis drugs. Apoptosis and death cell were also tested on M. bovis BCG infected cells. Minimal inhibitory concentrations of anti-tuberculosis drugs were analyzed. RESULTS All cells lines showed viability values higher than 80% when exposed to cigarette smoke extract. However, THP-1 cell line infected with M. bovis BCG and exposed to Marlboro cigarette smoke showed up to a 54% reduction of apoptotic cells than cells unexposed to smoke. M. tuberculosis exposed to Marlboro cigarette smoke for 11 days had an optical density 16% lower than unexposed bacteria. When cells were infected with M. tuberculosis, the intracellular recovery of CFUs showed up to a 0.66 log reduction in cells exposed to cigarette smoke extract because of a potential impairment in the phagocytosis. Macrophages treated with drugs showed up to a 2.55 log reduction in the intracellular load burden compared with non-treated ones. Despite poor treatment outcome on TB smoker patients, minimal inhibitory concentration of rifampicin increased only 2-fold in M. tuberculosis exposed to cigarette smoke. CONCLUSION Smoking interferes with tuberculosis treatment impairing the immunity of the host.
Collapse
Affiliation(s)
- Pablo Rodríguez-Fernández
- Servei de Microbiologia, Hospital Universitari Germans Trias i Pujol, Institut d'Investigació en Ciències de la Salut Germans Trias i Pujol, Badalona, Spain.,CIBER Enfermedades Respiratorias (CIBERES), Madrid, Spain.,Institut de Biotecnologia i de Biomedicina (IBB), Universitat Autònoma de Barcelona (UAB), Barcelona, Spain.,Departament de Genètica i de Microbiologia, Universitat Autònoma de Barcelona (UAB), Barcelona, Spain
| | - Andromeda-Celeste Gómez
- Servei de Microbiologia, Hospital Universitari Germans Trias i Pujol, Institut d'Investigació en Ciències de la Salut Germans Trias i Pujol, Badalona, Spain.,CIBER Enfermedades Respiratorias (CIBERES), Madrid, Spain.,Institut de Biotecnologia i de Biomedicina (IBB), Universitat Autònoma de Barcelona (UAB), Barcelona, Spain.,Departament de Genètica i de Microbiologia, Universitat Autònoma de Barcelona (UAB), Barcelona, Spain
| | - Isidre Gibert
- Institut de Biotecnologia i de Biomedicina (IBB), Universitat Autònoma de Barcelona (UAB), Barcelona, Spain.,Departament de Genètica i de Microbiologia, Universitat Autònoma de Barcelona (UAB), Barcelona, Spain
| | - Cristina Prat-Aymerich
- Servei de Microbiologia, Hospital Universitari Germans Trias i Pujol, Institut d'Investigació en Ciències de la Salut Germans Trias i Pujol, Badalona, Spain.,CIBER Enfermedades Respiratorias (CIBERES), Madrid, Spain.,Departament de Genètica i de Microbiologia, Universitat Autònoma de Barcelona (UAB), Barcelona, Spain
| | - Jose Domínguez
- Servei de Microbiologia, Hospital Universitari Germans Trias i Pujol, Institut d'Investigació en Ciències de la Salut Germans Trias i Pujol, Badalona, Spain.,CIBER Enfermedades Respiratorias (CIBERES), Madrid, Spain.,Departament de Genètica i de Microbiologia, Universitat Autònoma de Barcelona (UAB), Barcelona, Spain
| |
Collapse
|
11
|
De Cunto G, Cavarra E, Bartalesi B, Lucattelli M, Lungarella G. Innate Immunity and Cell Surface Receptors in the Pathogenesis of COPD: Insights from Mouse Smoking Models. Int J Chron Obstruct Pulmon Dis 2020; 15:1143-1154. [PMID: 32547002 PMCID: PMC7246326 DOI: 10.2147/copd.s246219] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Accepted: 05/03/2020] [Indexed: 12/23/2022] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is mainly associated with smoking habit. Inflammation is the major initiating process whereby neutrophils and monocytes are attracted into the lung microenvironment by external stimuli present in tobacco leaves and in cigarette smoke, which promote chemotaxis, adhesion, phagocytosis, release of superoxide anions and enzyme granule contents. A minority of smokers develops COPD and different molecular factors, which contribute to the onset of the disease, have been put forward. After many years of research, the pathogenesis of COPD is still an object of debate. In vivo models of cigarette smoke-induced COPD may help to unravel cellular and molecular mechanisms underlying the pathogenesis of COPD. The mouse represents the most favored animal choice with regard to the study of immune mechanisms due to its genetic and physiological similarities to humans, the availability of a large variability of inbred strains, the presence in the species of several genetic disorders analogous to those in man, and finally on the possibility to create models “made-to-measure” by genetic manipulation. The review outlines the different response of mouse strains to cigarette smoke used in COPD studies while retaining a strong focus on their relatability to human patients. These studies reveal the importance of innate immunity and cell surface receptors in the pathogenesis of pulmonary injury induced by cigarette smoking. They further advance the way in which we use wild type or genetically manipulated strains to improve our overall understanding of a multifaceted disease such as COPD. The structural and functional features, which have been found in the different strains of mice after chronic exposure to cigarette smoke, can be used in preclinical studies to develop effective new therapeutic agents for the different phenotypes in human COPD.
Collapse
Affiliation(s)
- Giovanna De Cunto
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - Eleonora Cavarra
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - Barbara Bartalesi
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - Monica Lucattelli
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - Giuseppe Lungarella
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| |
Collapse
|
12
|
Peng H, Guo T, Chen Z, Zhang H, Cai S, Yang M, Chen P, Guan C, Fang X. Hypermethylation of mitochondrial transcription factor A induced by cigarette smoke is associated with chronic obstructive pulmonary disease. Exp Lung Res 2019; 45:101-111. [PMID: 31198067 DOI: 10.1080/01902148.2018.1556748] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Revised: 11/21/2018] [Accepted: 12/04/2018] [Indexed: 02/08/2023]
Abstract
Purpose of the study: Cigarette smoking is a leading environmental contributor to chronic obstructive pulmonary disease (COPD), but its epigenetic regulation of mtTFA gene remains elusive. This study aims to explore the relationship of DNA methylation of mtTFA and cigarette smoking in COPD. Materials and Methods: We analyzed DNA methylation on mtTFA promoters in clinical samples from COPD patients and subjects with normal pulmonary function. Expression of mtTFA mRNA in the clinical samples and mtTFA mRNA and protein in human umbilical vein endothelial cells(HUVECs) treated with cigarette smoke extract (CSE) was evaluated. mtTFA mRNA and protein levels were measured to determine effects of demethylation agents on CSE-treated HUVECs. Results: The DNA methylation level of the mtTFA promoter was significantly increased in COPD group. Expression of mtTFA mRNA was downregulated in the lungs as a consequence of hypermethylation of mtTFA promoter. Expression of mtTFA mRNA and protein was downregulated in CSE-treated HUVECs as a consequence of hypermethylation of the mtTFA promoter. mtTFA expression in CSE-treated HUVECs was restored by the methylation inhibitor, 5-aza-2'-deoxycytidine(AZA). Conclusions: Cigarette smoke-induced hypermethylation of the mtTFA promoter is related to the initiation and progression of COPD. Our finding may provide a new strategy for the intervention of COPD by developing demethylation agents targeting mtTFA hypermethylation.
Collapse
Affiliation(s)
- Hong Peng
- a Department of Respiratory and Critical Care Medicine , The Second Xiangya Hospital of Central-South University , Changsha , PR China
- b b The Respiratory Disease Research Institute of Central South University , Changsha , PR China
- c c The Respiratory Disease Diagnosis and Treatment Center of Hunan Province , Changsha , PR China
| | - Ting Guo
- a Department of Respiratory and Critical Care Medicine , The Second Xiangya Hospital of Central-South University , Changsha , PR China
- b b The Respiratory Disease Research Institute of Central South University , Changsha , PR China
- c c The Respiratory Disease Diagnosis and Treatment Center of Hunan Province , Changsha , PR China
| | - Zhiyong Chen
- d d Department of Urology , Xiangya Hospital of Central-South University , Changsha , PR China
| | - Hongliang Zhang
- a Department of Respiratory and Critical Care Medicine , The Second Xiangya Hospital of Central-South University , Changsha , PR China
- b b The Respiratory Disease Research Institute of Central South University , Changsha , PR China
- c c The Respiratory Disease Diagnosis and Treatment Center of Hunan Province , Changsha , PR China
| | - Shan Cai
- a Department of Respiratory and Critical Care Medicine , The Second Xiangya Hospital of Central-South University , Changsha , PR China
- b b The Respiratory Disease Research Institute of Central South University , Changsha , PR China
- c c The Respiratory Disease Diagnosis and Treatment Center of Hunan Province , Changsha , PR China
| | - Min Yang
- a Department of Respiratory and Critical Care Medicine , The Second Xiangya Hospital of Central-South University , Changsha , PR China
- b b The Respiratory Disease Research Institute of Central South University , Changsha , PR China
- c c The Respiratory Disease Diagnosis and Treatment Center of Hunan Province , Changsha , PR China
| | - Ping Chen
- a Department of Respiratory and Critical Care Medicine , The Second Xiangya Hospital of Central-South University , Changsha , PR China
- b b The Respiratory Disease Research Institute of Central South University , Changsha , PR China
- c c The Respiratory Disease Diagnosis and Treatment Center of Hunan Province , Changsha , PR China
| | - Chaxiang Guan
- e Physiological Research Center , Xiangya Medical School of Central-South University , Changsha , PR China
| | - Xiang Fang
- f Department of Neurology , University of Texas Medical Branch , Galveston , Texas, USA
| |
Collapse
|
13
|
Mechanisms of whole smoke conditioned media induced cytotoxicity to human aortic endothelial cells. Toxicol In Vitro 2019; 58:239-244. [PMID: 30885593 DOI: 10.1016/j.tiv.2019.03.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Revised: 01/22/2019] [Accepted: 03/07/2019] [Indexed: 11/22/2022]
Abstract
Chronic exposure to cigarette smoke can lead to endothelial dysfunction and potentially endothelial cell death. Here, we exposed Human Aortic Endothelial Cells (HAECs) to whole smoke conditioned media (WSCM) over a range of nicotine equivalence (n.e.) concentrations (0-8000 ng/mL n.e.). After 24 h, Neutral Red Uptake (NRU) and reduction of 3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2H-tetrazolium bromide (MTT) to formazan was determined for each exposure concentration and compared to control. IC50 values in the NRU assay were: 4582 ng/mL n.e. ± 1074, 4587 ng/mL n.e. ± 951, 4993 ng/mL n.e. ± 1239 and 4691 ng/mL n.e. ± 402 for four HAEC donors. IC50 values in the MTT assay were: 4885 ng/mL n.e. ± 1341, 4584 ng/mL n.e. ± 806, 5749 ng/mL n.e. ± 783 and 5228 ng/mL n.e. ± 593 for the four donors. To examine the mechanism responsible for WSCM-induced cytotoxicity in HAECs, flow cytometry using necrosis (Propidium Iodide) and apoptosis (Annexin V) markers were used. Annexin V-positive cell populations increased in a dose dependent manner while increases in PI-positive cell populations occurred at the highest doses of WSCM (5000-8000 ng/mL n.e.). Western blotting for cleaved caspase-3 confirmed that apoptosis occurs at >5000 ng/mL n.e. WSCM, coinciding with reduced HAEC survival.
Collapse
|
14
|
Somborac-Bačura A, Rumora L, Novak R, Rašić D, Dumić J, Čepelak I, Žanić-Grubišić T. Differential expression of heat shock proteins and activation of mitogen-activated protein kinases in A549 alveolar epithelial cells exposed to cigarette smoke extract. Exp Physiol 2018; 103:1666-1678. [PMID: 30242929 DOI: 10.1113/ep087038] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Accepted: 09/20/2018] [Indexed: 01/24/2023]
Abstract
NEW FINDINGS What is the central question of this study? What is the effect of cigarette smoke on cell death, oxidative damage, expression of heat shock proteins (HSPs) and activation of mitogen-activated protein kinases (MAPKs) in A549 alveolar epithelial cells? What is the main finding and its importance? Cigarette smoke induces cytotoxicity and oxidative damage to A549 cells, increases expression of different HSPs and activates MAPK signalling pathways. This could be related to inflammatory response and apoptosis observed in lungs of patients with smoking-related diseases. ABSTRACT Cigarette smoking is one of the main risk factors for development of chronic obstructive pulmonary disease (COPD). We previously reported that cigarette smoke (CS) induces damage to proteins and their ineffective degradation. Here, we hypothesize that CS could induce oxidative stress and cytotoxicity in lung epithelial cells through alterations of heat shock protein (HSP) expression and mitogen-activated protein kinase (MAPK) signalling pathways. We exposed A549 alveolar epithelial cells to various concentrations of cigarette smoke extract (CSE). Higher concentrations of CSE caused apoptosis of A549 cells after 4 h, while after 24 h cell viability was decreased, and lactate dehydrogenase in cell culture medium was increased as well as the number of necrotic cells. Concentrations of malondialdehyde (MDA) were elevated, while total thiol groups were decreased. Changes in the expression of HSPs (HSP70, HSP32 and HSP27) were time-dependent. After 6 h, CSE caused an increase in the expression of HSP70 and HSP32, while after 8 h all examined HSPs were up-regulated and remained increased up to 48 h. Treatment of A549 cells with CSE stimulated phosphorylation of extracellular signal-regulated kinase and p38 in a dose-dependent manner, while c-Jun N-terminal kinase activation was not detected. By using specific inhibitors, we demonstrated that MAPKs and HSPs interplay in CSE effects. In conclusion, our results show that MAPKs and HSPs are involved in the mechanism underlying CSE-induced cytotoxicity and oxidative damage to A549 alveolar epithelial cells. These processes could be related to inflammatory response and apoptosis observed in lungs of patients with smoking-related diseases, such as COPD.
Collapse
Affiliation(s)
- Anita Somborac-Bačura
- Faculty of Pharmacy and Biochemistry, Department of Medical Biochemistry and Hematology, University of Zagreb, Kneza Domagoja, Zagreb, Croatia
| | - Lada Rumora
- Faculty of Pharmacy and Biochemistry, Department of Medical Biochemistry and Hematology, University of Zagreb, Kneza Domagoja, Zagreb, Croatia
| | - Ruđer Novak
- Faculty of Pharmacy and Biochemistry, Department of Biochemistry and Molecular Biology, University of Zagreb, Ante Kovačića, Zagreb, Croatia
| | - Dubravka Rašić
- Unit of Toxicology, Institute for Medical Research and Occupational Health, Ksaverska cesta, Zagreb, Croatia
| | - Jerka Dumić
- Faculty of Pharmacy and Biochemistry, Department of Biochemistry and Molecular Biology, University of Zagreb, Ante Kovačića, Zagreb, Croatia
| | - Ivana Čepelak
- Faculty of Pharmacy and Biochemistry, Department of Medical Biochemistry and Hematology, University of Zagreb, Kneza Domagoja, Zagreb, Croatia
| | - Tihana Žanić-Grubišić
- Faculty of Pharmacy and Biochemistry, Department of Medical Biochemistry and Hematology, University of Zagreb, Kneza Domagoja, Zagreb, Croatia
| |
Collapse
|
15
|
Shermatov K, Kazanasmaz H, Guzel B, Cebi N. LEVELS OF SERUM M30 AND M65 PROTEINS AS BIOMARKERS OF APOPTOSIS IN CHILDREN EXPOSED TO PASSIVE SMOKING. KONURALP TIP DERGISI 2018. [DOI: 10.18521/ktd.435349] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
|
16
|
Bai X, Aerts SL, Verma D, Ordway DJ, Chan ED. Epidemiologic Evidence of and Potential Mechanisms by Which Second-Hand Smoke Causes Predisposition to Latent and Active Tuberculosis. Immune Netw 2018; 18:e22. [PMID: 29984040 PMCID: PMC6026693 DOI: 10.4110/in.2018.18.e22] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Revised: 06/15/2018] [Accepted: 06/16/2018] [Indexed: 12/13/2022] Open
Abstract
Many studies have linked cigarette smoke (CS) exposure and tuberculosis (TB) infection and disease although much fewer have studied second-hand smoke (SHS) exposure. Our goal is to review the epidemiologic link between SHS and TB as well as to summarize the effects SHS and direct CS on various immune cells relevant for TB. PubMed searches were performed using the key words "tuberculosis" with "cigarette," "tobacco," or "second-hand smoke." The bibliography of relevant papers were examined for additional relevant publications. Relatively few studies associate SHS exposure with TB infection and active disease. Both SHS and direct CS can alter various components of host immunity resulting in increased vulnerability to TB. While the epidemiologic link of these 2 health maladies is robust, more definitive, mechanistic studies are required to prove that SHS and direct CS actually cause increased susceptibility to TB.
Collapse
Affiliation(s)
- Xiyuan Bai
- Department of Medicine, Denver Veterans Affairs Medical Center, University of Colorado Anschutz Medical Center, Denver, CO 80045, USA
- Department of Medicine and Office of Academic Affairs, National Jewish Health, Denver, CO 80206, USA
- Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Anschutz Medical Campus, Denver, CO 80045, USA
| | - Shanae L. Aerts
- Department of Medicine and Office of Academic Affairs, National Jewish Health, Denver, CO 80206, USA
| | - Deepshikha Verma
- Department of Microbiology, Immunology, and Pathology, Mycobacteria Research Laboratories, Colorado State University, Fort Collins, CO 80523, USA
| | - Diane J. Ordway
- Department of Microbiology, Immunology, and Pathology, Mycobacteria Research Laboratories, Colorado State University, Fort Collins, CO 80523, USA
| | - Edward D. Chan
- Department of Medicine, Denver Veterans Affairs Medical Center, University of Colorado Anschutz Medical Center, Denver, CO 80045, USA
- Department of Medicine and Office of Academic Affairs, National Jewish Health, Denver, CO 80206, USA
- Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Anschutz Medical Campus, Denver, CO 80045, USA
| |
Collapse
|
17
|
Lu Q, Gottlieb E, Rounds S. Effects of cigarette smoke on pulmonary endothelial cells. Am J Physiol Lung Cell Mol Physiol 2018; 314:L743-L756. [PMID: 29351435 DOI: 10.1152/ajplung.00373.2017] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Cigarette smoking is the leading cause of preventable disease and death in the United States. Cardiovascular comorbidities associated with both active and secondhand cigarette smoking indicate the vascular toxicity of smoke exposure. Growing evidence supports the injurious effect of cigarette smoke on pulmonary endothelial cells and the roles of endothelial cell injury in development of acute respiratory distress syndrome (ARDS), emphysema, and pulmonary hypertension. This review summarizes results from studies of humans, preclinical animal models, and cultured endothelial cells that document toxicities of cigarette smoke exposure on pulmonary endothelial cell functions, including barrier dysfunction, endothelial activation and inflammation, apoptosis, and vasoactive mediator production. The discussion is focused on effects of cigarette smoke-induced endothelial injury in the development of ARDS, emphysema, and vascular remodeling in chronic obstructive pulmonary disease.
Collapse
Affiliation(s)
- Qing Lu
- Vascular Research Laboratory, Providence Veterans Affairs Medical Center , Providence, Rhode Island.,Department of Medicine, Alpert Medical School of Brown University , Providence, Rhode Island
| | - Eric Gottlieb
- Department of Medicine, Alpert Medical School of Brown University , Providence, Rhode Island
| | - Sharon Rounds
- Vascular Research Laboratory, Providence Veterans Affairs Medical Center , Providence, Rhode Island.,Department of Medicine, Alpert Medical School of Brown University , Providence, Rhode Island
| |
Collapse
|
18
|
Peixoto MS, de Oliveira Galvão MF, Batistuzzo de Medeiros SR. Cell death pathways of particulate matter toxicity. CHEMOSPHERE 2017; 188:32-48. [PMID: 28865791 DOI: 10.1016/j.chemosphere.2017.08.076] [Citation(s) in RCA: 99] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Revised: 08/14/2017] [Accepted: 08/16/2017] [Indexed: 06/07/2023]
Abstract
Humans are exposed to various complex mixtures of particulate matter (PM) from different sources. Long-term exposure to high levels of these particulates has been linked to a diverse range of respiratory and cardiovascular diseases that have resulted in hospital admission. The evaluation of the effects of PM exposure on the mechanisms related to cell death has been a challenge for many researchers. Therefore, in this review, we have discussed the effects of airborne PM exposure on mechanisms related to cell death. For this purpose, we have compiled literature data on PM sources, the effects of exposure, and the assays and models used for evaluation, in order to establish comparisons between various studies. The analysis of this collected data suggested divergent responses to PM exposure that resulted in different cell death types (apoptosis, autophagy, and necrosis). In addition, PM induced oxidative stress within cells, which appeared to be an important factor in the determination of cell fate. When the levels of reactive oxygen species were overpowering, the cellular fate was directed toward cell death. This may be the underlying mechanism of the development or exacerbation of respiratory diseases, such as emphysema and chronic obstructive pulmonary diseases. In addition, PM was shown to cause DNA damage and the resulting mutations increased the risk of cancer. Furthermore, several conditions should be considered in the assessment of cell death in PM-exposed models, including the cell culture line, PM composition, and the interaction of the different cells types in in vivo models.
Collapse
Affiliation(s)
- Milena Simões Peixoto
- Graduate Program in Biochemistry, Biosciences Center, Federal University of Rio Grande do Norte, Natal, RN, Brazil.
| | - Marcos Felipe de Oliveira Galvão
- Graduate Program in Biochemistry, Biosciences Center, Federal University of Rio Grande do Norte, Natal, RN, Brazil; Department of Cell Biology and Genetics, Biosciences Center, Federal University of Rio Grande do Norte, Natal, RN, Brazil.
| | | |
Collapse
|
19
|
Joshua V, Chatzidionisyou K, Catrina AI. Role of the lung in individuals at risk of rheumatoid arthritis. Best Pract Res Clin Rheumatol 2017; 31:31-41. [DOI: 10.1016/j.berh.2017.08.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Revised: 03/31/2017] [Accepted: 06/12/2017] [Indexed: 10/18/2022]
|
20
|
Torshabi M, Esfahrood ZR, Gholamin P, Karami E. Effects of nicotine in the presence and absence of vitamin E on morphology, viability and osteogenic gene expression in MG-63 osteoblast-like cells. J Basic Clin Physiol Pharmacol 2016; 27:595-602. [PMID: 27180340 DOI: 10.1515/jbcpp-2015-0143] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Accepted: 04/15/2016] [Indexed: 06/05/2023]
Abstract
BACKGROUND Evidence shows that oxidative stress induced by nicotine plays an important role in bone loss. Vitamin E with its antioxidative properties may be able to reverse the effects of nicotine on bone. This study aimed to assess the effects of nicotine in the presence and absence of vitamin E on morphology, viability and osteogenic gene expression in MG-63 (osteosarcoma) human osteoblast-like cells. METHODS We treated the cells with 5 mM nicotine. The viability and morphology of cells were evaluated respectively using the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium (MTT) and crystal violet assays. The effect of nicotine on osteogenic gene expression in MG-63 cells was assessed by real-time reverse-transcription polymerase chain reaction of osteoblast markers, namely, alkaline phosphatase, osteocalcin and bone sialoprotein. RESULTS The results revealed that survival and proliferation of MG-63 cells were suppressed following exposure to nicotine, and cytoplasm vacuolization occurred in the cells. Nicotine significantly down-regulated the expression of osteogenic marker genes. Such adverse effects on morphology, viability and osteogenic gene expression of MG-63 cells were reversed by vitamin E therapy. CONCLUSIONS In conclusion, vitamin E supplementation may play a role in proliferation and differentiation of osteoblasts, and vitamin E can be considered as an anabolic agent to treat nicotine-induced bone loss.
Collapse
|
21
|
Lin J, Zhang Y, Wang H, Chang J, Wei L, Cao L, Zhang Z, Zhang X. Genetic Polymorphisms in the Apoptosis-Associated Gene CASP3 and the Risk of Lung Cancer in Chinese Population. PLoS One 2016; 11:e0164358. [PMID: 27723786 PMCID: PMC5056705 DOI: 10.1371/journal.pone.0164358] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Accepted: 09/24/2016] [Indexed: 11/30/2022] Open
Abstract
Caspase-3 (CASP3) plays a central role in executing cell apoptosis and thus in carcinogenesis. We previously investigated the relationship between functional polymorphisms in CAPS3 829 A>C and 20541 C>T and risk of esophageal squamous cell carcinoma. However little is known about the role of CASP3 variants in susceptibility to lung cancer. To figure out the contribution of CASP3 polymorphisms to lung cancer risk, genotypes of 1000 lung cancer patients and 1000 controls were conducted by RFLP-PCR (restriction fragment length polymorphism PCR). The transcriptional activity of CASP3 829 A>C was examined by dual luciferase reporter assay. Logistic regression was applied to calculate Odds ratios (OR) and 95% confidence intervals (95%CI). Compared with CASP3 829 AA genotype, AC and CC genotype had significantly increased risk of lung cancer with OR (95% CI) of 1.33 (1.09–1.63) and 1.55 (1.19–2.01), respectively. To further explore the possible impact of 829 A>C SNP on CASP3 transcriptional activity, we detected the dual luciferase activity of PGL3-promoter vectors containing 829A or 829C alleles in lung cancer cell lines and found that report gene expressions driven by 829A containing CASP3 promoter were 1.64-fold, 1.94-fold greater than those driven by CASP3 829C containing counterparts in A549 and NCI-H1975 cells (P<0.001). When stratified by sex, the significantly increased risk associated with CASP3 829 AC or CC genotype was obviousl in males with OR (95% CI) of 1.42 (1.11–1.81) and 1.51 (1.11–2.05), but not in females. When stratified by age, we found that CASP3 829 AC or CC genotype contributed to the risk of lung cancer in youngers with OR (95% CI) of 2.73 (1.71–4.34) and 4.02 (2.20–7.32), but not in elder group. We also found that 829AC or 829CC genotype increased adenocarcinoma risk compared with the AA genotype with OR (95%CI) of 1.33 (1.04–1.70) and 1.51(1.09–2.07). CASP3 polymorphism and smoking interaction was demonstrated related with higher risk of lung cancer. We achieved that the CASP3 829AC or 829CC genotypes was associated with increased risk of lung cancer in both non-smoker and smoker group, with OR (95%CI) of 1.48 (1.08–2.02) and OR (95%CI) of 1.64 (1.09–2.48) among non-smokers and OR (95%CI) of 2.68 (1.89–3.81) and OR (95%CI) of 3.23 (2.21–4.92) among smokers, respectively. Among carriers with 20541CT genotype, the ORs (95%CI) of risk with lung cancer for smoking <16, 16–28, or > 28 pack-years were 1.16(0.65–2.07), 1.66(0.98–2.82) and 5.01(3.31–7.58) compared with the 20541CC carriers. And among carriers with 20541CT genotype, the ORs (95%CI) were 0.86(0.33–2.20), 2.12(0.83–5.41) and 5.71(2.68–12.16). These results highlight apoptosis-related CASP3 as an important gene in human carcinogenesis and further support the CASP3 polymorphisms confer to the lung cancer susceptibility.
Collapse
Affiliation(s)
- Jia Lin
- Department of Molecular Genetics, College of Life Science, North China University of Science and Technology, Tangshan, China
| | - Yanyan Zhang
- Department of Molecular Genetics, College of Life Science, North China University of Science and Technology, Tangshan, China
- Department of Epidemiology, School of Public Health, North China University of Science and Technology, Tangshan, China
| | - Hongge Wang
- Department of Molecular Genetics, College of Life Science, North China University of Science and Technology, Tangshan, China
| | - Jiang Chang
- Department of Epidemiology and Biostatistics, and State Key Laboratory of Environment Health (Incubation), MOE (Ministry of Education) Key Laboratory of Environment & Health, Ministry of Environmental Protection Key Laboratory of Environment and Health (Wuhan), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lixuan Wei
- Department of Etiology and Carcinogenesis, Cancer Institute and Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Lei Cao
- Department of Molecular Genetics, College of Life Science, North China University of Science and Technology, Tangshan, China
| | - Zhi Zhang
- Department of Chemotherapy and Radiotherapy, Tangshan Gongren Hospital, Tangshan, China
| | - Xuemei Zhang
- Department of Molecular Genetics, College of Life Science, North China University of Science and Technology, Tangshan, China
- * E-mail:
| |
Collapse
|
22
|
Catrina AI, Joshua V, Klareskog L, Malmström V. Mechanisms involved in triggering rheumatoid arthritis. Immunol Rev 2016; 269:162-74. [PMID: 26683152 DOI: 10.1111/imr.12379] [Citation(s) in RCA: 105] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Rheumatoid arthritis (RA) is a chronic inflammatory syndrome with a strong autoimmune component. The autoantigens in RA are neither tissue nor organ-specific, but comprise a broad collection of post-translational modified proteins, such as citrullinated proteins. These modifications are likely to be triggered by innate stimuli. In genetically susceptible hosts, they can lead to a more substantiated secondary autoimmune reaction targeting the joints and precipitating the clinical onset of RA. Both innate and adaptive mechanisms will then closely interplay to promote chronic joint inflammation in the several absence of appropriate treatment. This scenario, is shared with other autoimmune diseases where potentially pathogenic immune responses are present already before disease onset. Better understanding of these processes will allow both earlier diagnosis of RA and identification of those healthy individuals that are at risk of developing disease, opening possibilities for disease prevention. In this review, we discuss the iterative processes of innate and adaptive immunity responsible for the (longitudinal) development of immune reactions that may contribute to the development of RA.
Collapse
Affiliation(s)
- Anca I Catrina
- Rheumatology Unit, Department of Medicine, Solna, Karolinska Institutet, Stockholm, Sweden
| | - Vijay Joshua
- Rheumatology Unit, Department of Medicine, Solna, Karolinska Institutet, Stockholm, Sweden
| | - Lars Klareskog
- Rheumatology Unit, Department of Medicine, Solna, Karolinska Institutet, Stockholm, Sweden
| | - Vivianne Malmström
- Rheumatology Unit, Department of Medicine, Solna, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
23
|
Kim MS, Yun JW, Park JH, Park BW, Kang YH, Hah YS, Hwang SC, Woo DK, Byun JH. Autophagy Has a Beneficial Role in Relieving Cigarette Smoke-Induced Apoptotic Death in Human Gingival Fibroblasts. Int J Med Sci 2016; 13:357-64. [PMID: 27226776 PMCID: PMC4879768 DOI: 10.7150/ijms.14592] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Accepted: 04/12/2016] [Indexed: 01/11/2023] Open
Abstract
The deleterious role of cigarette smoke has long been documented in various human diseases including periodontal complications. In this report, we examined this adverse effect of cigarette smoke on human gingival fibroblasts (HGFs) which are critical not only in maintaining gingival tissue architecture but also in mediating immune responses. As well documented in other cell types, we also observed that cigarette smoke promoted cellular reactive oxygen species in HGFs. And we found that this cigarette smoke-induced oxidative stress reduced HGF viability through inducing apoptosis. Our results indicated that an increased Bax/Bcl-xL ratio and resulting caspase activation underlie the apoptotic death in HGFs exposed to cigarette smoke. Furthermore, we detected that cigarette smoke also triggered autophagy, an integrated cellular stress response. Interesting, a pharmacological suppression of the cigarette smoke-induced autophagy led to a further reduction in HGF viability while a pharmacological promotion of autophagy increased the viability of HGFs with cigarette smoke exposures. These findings suggest a protective role for autophagy in HGFs stressed with cigarette smoke, highlighting that modulation of autophagy can be a novel therapeutic target in periodontal complications with cigarette smoke.
Collapse
Affiliation(s)
- Moon-Soo Kim
- 1. Department of Oral and Maxillofacial Surgery, Gyeongsang National University School of Medicine and Gyeongsang National University Hospital, Institute of Health Sciences, Gyeongsang National University, Chilam-dong, Jinju, 660-702, Republic of Korea
| | - Jeong-Won Yun
- 1. Department of Oral and Maxillofacial Surgery, Gyeongsang National University School of Medicine and Gyeongsang National University Hospital, Institute of Health Sciences, Gyeongsang National University, Chilam-dong, Jinju, 660-702, Republic of Korea
| | - Jin-Ho Park
- 1. Department of Oral and Maxillofacial Surgery, Gyeongsang National University School of Medicine and Gyeongsang National University Hospital, Institute of Health Sciences, Gyeongsang National University, Chilam-dong, Jinju, 660-702, Republic of Korea
| | - Bong-Wook Park
- 1. Department of Oral and Maxillofacial Surgery, Gyeongsang National University School of Medicine and Gyeongsang National University Hospital, Institute of Health Sciences, Gyeongsang National University, Chilam-dong, Jinju, 660-702, Republic of Korea
| | - Young-Hoon Kang
- 1. Department of Oral and Maxillofacial Surgery, Gyeongsang National University School of Medicine and Gyeongsang National University Hospital, Institute of Health Sciences, Gyeongsang National University, Chilam-dong, Jinju, 660-702, Republic of Korea
| | - Young-Sool Hah
- 2. Clinical Research Institutue of Gyeongsang National University Hospital, Jinju, Republic of Korea
| | - Sun-Chul Hwang
- 3. Department of Orthopaedic Surgery, Institute of Health Sciences, Gyeongsang National University School of Medicine, Jinju, Republic of Korea
| | - Dong Kyun Woo
- 4. College of Pharmacy and Research Institute of Pharmaceutical Sciences, Gyeongsang National University, Jinju, Republic of Korea
| | - June-Ho Byun
- 1. Department of Oral and Maxillofacial Surgery, Gyeongsang National University School of Medicine and Gyeongsang National University Hospital, Institute of Health Sciences, Gyeongsang National University, Chilam-dong, Jinju, 660-702, Republic of Korea
| |
Collapse
|
24
|
Zhang X, Xiao X, Duan H, Gao F, Li Y, Niu Y, Gao W, Wang H, Yu S, Zheng Y. Cytotoxicity of diesel engine exhaust among the Chinese occupational population: a complement of cytokinesis-block micronucleus cytome. Inhal Toxicol 2016; 28:274-80. [DOI: 10.3109/08958378.2016.1162233] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
25
|
Pouwels SD, Zijlstra GJ, van der Toorn M, Hesse L, Gras R, Ten Hacken NHT, Krysko DV, Vandenabeele P, de Vries M, van Oosterhout AJM, Heijink IH, Nawijn MC. Cigarette smoke-induced necroptosis and DAMP release trigger neutrophilic airway inflammation in mice. Am J Physiol Lung Cell Mol Physiol 2016; 310:L377-86. [PMID: 26719146 DOI: 10.1152/ajplung.00174.2015] [Citation(s) in RCA: 113] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Accepted: 12/18/2015] [Indexed: 11/22/2022] Open
Abstract
Recent data indicate a role for airway epithelial necroptosis, a regulated form of necrosis, and the associated release of damage-associated molecular patterns (DAMPs) in the development of chronic obstructive pulmonary disease (COPD). DAMPs can activate pattern recognition receptors (PRRs), triggering innate immune responses. We hypothesized that cigarette smoke (CS)-induced epithelial necroptosis and DAMP release initiate airway inflammation in COPD. Human bronchial epithelial BEAS-2B cells were exposed to cigarette smoke extract (CSE), and necrotic cell death (membrane integrity by propidium iodide staining) and DAMP release (i.e., double-stranded DNA, high-mobility group box 1, heat shock protein 70, mitochondrial DNA, ATP) were analyzed. Subsequently, BEAS-2B cells were exposed to DAMP-containing supernatant of CS-induced necrotic cells, and the release of proinflammatory mediators [C-X-C motif ligand 8 (CXCL-8), IL-6] was evaluated. Furthermore, mice were exposed to CS in the presence and absence of the necroptosis inhibitor necrostatin-1, and levels of DAMPs and inflammatory cell numbers were determined in bronchoalveolar lavage fluid. CSE induced a significant increase in the percentage of necrotic cells and DAMP release in BEAS-2B cells. Stimulation of BEAS-2B cells with supernatant of CS-induced necrotic cells induced a significant increase in the release of CXCL8 and IL-6, in a myeloid differentiation primary response gene 88-dependent fashion. In mice, exposure of CS increased the levels of DAMPs and numbers of neutrophils in bronchoalveolar lavage fluid, which was statistically reduced upon treatment with necrostatin-1. Together, we showed that CS exposure induces necrosis of bronchial epithelial cells and subsequent DAMP release in vitro, inducing the production of proinflammatory cytokines. In vivo, CS exposure induces neutrophilic airway inflammation that is sensitive to necroptosis inhibition.
Collapse
Affiliation(s)
- Simon D Pouwels
- Department of Pathology and Medical Biology, Experimental Pulmonology and Inflammation Research, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands; GRIAC Research Institute, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - G Jan Zijlstra
- Department of Pathology and Medical Biology, Experimental Pulmonology and Inflammation Research, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands; GRIAC Research Institute, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands; Department of Pulmonology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Marco van der Toorn
- Department of Pathology and Medical Biology, Experimental Pulmonology and Inflammation Research, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands; GRIAC Research Institute, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Laura Hesse
- Department of Pathology and Medical Biology, Experimental Pulmonology and Inflammation Research, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands; GRIAC Research Institute, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Renee Gras
- Department of Pathology and Medical Biology, Experimental Pulmonology and Inflammation Research, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands; GRIAC Research Institute, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Nick H T Ten Hacken
- GRIAC Research Institute, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands; Department of Pulmonology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Dmitri V Krysko
- Molecular Signaling and Cell Death Unit, Inflammation Research Center, VIB, Ghent, Belgium; and Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Peter Vandenabeele
- Molecular Signaling and Cell Death Unit, Inflammation Research Center, VIB, Ghent, Belgium; and Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Maaike de Vries
- Department of Pathology and Medical Biology, Experimental Pulmonology and Inflammation Research, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands; GRIAC Research Institute, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Antoon J M van Oosterhout
- Department of Pathology and Medical Biology, Experimental Pulmonology and Inflammation Research, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands; GRIAC Research Institute, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Irene H Heijink
- Department of Pathology and Medical Biology, Experimental Pulmonology and Inflammation Research, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands; GRIAC Research Institute, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Martijn C Nawijn
- Department of Pathology and Medical Biology, Experimental Pulmonology and Inflammation Research, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands; GRIAC Research Institute, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands;
| |
Collapse
|
26
|
Esakky P, Hansen DA, Drury AM, Cusumano A, Moley KH. Cigarette smoke-induced cell death of a spermatocyte cell line can be prevented by inactivating the Aryl hydrocarbon receptor. Cell Death Discov 2015; 1:15050. [PMID: 27551479 PMCID: PMC4979462 DOI: 10.1038/cddiscovery.2015.50] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2015] [Revised: 09/10/2015] [Accepted: 09/12/2015] [Indexed: 11/29/2022] Open
Abstract
Cigarette smoke exposure causes germ cell death during spermatogenesis. Our earlier studies demonstrated that cigarette smoke condensate (CSC) causes spermatocyte cell death in vivo and growth arrest of the mouse spermatocyte cell line (GC-2spd(ts)) in vitro via the aryl hydrocarbon receptor (AHR). We hypothesize here that inactivation of AHR could prevent the CSC-induced cell death in spermatocytes. We demonstrate that CSC exposure generates oxidative stress, which differentially regulates mitochondrial apoptosis in GC-2spd(ts) and wild type (WT) and AHR knockout (AHR-KO) mouse embryonic fibroblasts (MEFs). SiRNA-mediated silencing of Ahr augments the extent of CSC-mediated cellular damage while complementing the AHR-knockout condition. Pharmacological inhibition using the AHR-antagonist (CH223191) modulates the CSC-altered expression of apoptotic proteins and significantly abrogates DNA fragmentation though the cleavage of PARP appears AHR independent. Pretreatment with CH223191 at concentrations above 50 μM significantly prevents the CSC-induced activation of caspase-3/7 and externalization of phosphatidylserine in the plasma membrane. However, MAPK inhibitors alone or together with CH223191 could not prevent the membrane damage upon CSC addition and the caspase-3/7 activation and membrane damage in AHR-deficient MEF indicates the interplay of multiple cell signaling and cytoprotective ability of AHR. Thus the data obtained on one hand signifies the protective role of AHR in maintaining normal cellular homeostasis and the other, could be a potential prophylactic therapeutic target to promote cell survival and growth under cigarette smoke exposed environment by receptor antagonism via CH223191-like mechanism. Antagonist-mediated inactivation of the aryl hydrocarbon receptor blocks downstream events leading to cigarette smoke-induced cell death of a spermatocyte cell line.
Collapse
Affiliation(s)
- P Esakky
- Department of Veterans Affairs Medical Center, Washington University School of Medicine, St. Louis, MO, USA
- Department of Obstetrics and Gynecology, Washington University School of Medicine, St. Louis, MO, USA
| | - D A Hansen
- Department of Veterans Affairs Medical Center, Washington University School of Medicine, St. Louis, MO, USA
| | - A M Drury
- Department of Veterans Affairs Medical Center, Washington University School of Medicine, St. Louis, MO, USA
- Department of Obstetrics and Gynecology, Washington University School of Medicine, St. Louis, MO, USA
| | - A Cusumano
- Department of Obstetrics and Gynecology, Washington University School of Medicine, St. Louis, MO, USA
| | - K H Moley
- Department of Obstetrics and Gynecology, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
27
|
Yang M, Chen P, Peng H, Zhang H, Chen Y, Cai S, Lu Q, Guan C. Cigarette smoke extract induces aberrant cytochrome-c oxidase subunit II methylation and apoptosis in human umbilical vascular endothelial cells. Am J Physiol Cell Physiol 2015; 308:C378-84. [PMID: 25500741 DOI: 10.1152/ajpcell.00197.2014] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Cigarette smoke-induced apoptosis of vascular endothelial cells contributes to the pathogenesis of chronic obstructive pulmonary disease. However, the mechanisms responsible for endothelial apoptosis remain poorly understood. We conducted an in vitro study to investigate whether DNA methylation is involved in smoking-induced endothelial apoptosis. Human umbilical vascular endothelial cells (HUVECs) were exposed to cigarette smoke extract (CSE) at a range of concentrations (0-10%). HUVECs were also incubated with a demethylating reagent, 5-aza-2'-deoxycytidinem (AZA), with and without CSE. Apoptosis was assessed by terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling assay and flow cytometry using annexin V-FITC/propidium iodide staining. We found that CSE treatment significantly increased HUVEC apoptosis in a dose- and time-dependent manner. Quantitative real-time RT-PCR and immunoblot revealed that CSE treatment decreased cytochrome-c oxidase subunit II (COX II) mRNA and protein levels and decreased COX activity. Methylation-specific PCR and direct bisulfite sequencing revealed positive COX II gene methylation. AZA administration partly increased mRNA and protein expressions of COX II, and COX activity decreased by CSE and attenuated the toxic effects of CSE. Our results showed that CSE induced aberrant COX II methylation and apoptosis in HUVECs.
Collapse
Affiliation(s)
- Min Yang
- Department of Respiratory Medicine, The Second Xiangya Hospital of Central-South University, Hunan, Changsha, China
| | - Ping Chen
- Department of Respiratory Medicine, The Second Xiangya Hospital of Central-South University, Hunan, Changsha, China;
| | - Hong Peng
- Department of Respiratory Medicine, The Second Xiangya Hospital of Central-South University, Hunan, Changsha, China
| | - Hongliang Zhang
- Emergency Department, The Second Xiangya Hospital of Central-South University, Hunan, Changsha, China
| | - Yan Chen
- Department of Respiratory Medicine, The Second Xiangya Hospital of Central-South University, Hunan, Changsha, China
| | - Shan Cai
- Department of Respiratory Medicine, The Second Xiangya Hospital of Central-South University, Hunan, Changsha, China
| | - Qianjin Lu
- Dermatological Department, The Second Xiangya Hospital of Central-South University, Hunan, Changsha, China
| | - Chaxiang Guan
- Department of Physiology, Central-South University, Hunan, Changsha, China
| |
Collapse
|
28
|
Abstract
Chronic obstructive pulmonary disease (COPD) is one of the most common chronic illnesses in the world. The disease encompasses emphysema, chronic bronchitis, and small airway obstruction and can be caused by environmental exposures, primarily cigarette smoking. Since only a small subset of smokers develop COPD, it is believed that host factors interact with the environment to increase the propensity to develop disease. The major pathogenic factors causing disease include infection and inflammation, protease and antiprotease imbalance, and oxidative stress overwhelming antioxidant defenses. In this review, we will discuss the major environmental and host sources for oxidative stress; discuss how oxidative stress regulates chronic bronchitis; review the latest information on genetic predisposition to COPD, specifically focusing on oxidant/antioxidant imbalance; and review future antioxidant therapeutic options for COPD. The complexity of COPD will necessitate a multi-target therapeutic approach. It is likely that antioxidant supplementation and dietary antioxidants will have a place in these future combination therapies.
Collapse
Affiliation(s)
- Bernard M Fischer
- Department of Pediatrics, Duke University Medical Center, Durham, NC, USA
| | - Judith A Voynow
- Department of Pediatrics, Children’s Hospital of Richmond at Virginia Commonwealth University, Richmond, VA, USA
| | - Andrew J Ghio
- National Health and Environmental Effects Research Laboratory, US Environmental Protection Agency, Chapel Hill, NC, USA
| |
Collapse
|
29
|
Chan ED, Kinney WH, Honda JR, Bishwakarma R, Gangavelli A, Mya J, Bai X, Ordway DJ. Tobacco exposure and susceptibility to tuberculosis: is there a smoking gun? Tuberculosis (Edinb) 2014; 94:544-50. [PMID: 25305002 DOI: 10.1016/j.tube.2014.08.010] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2014] [Revised: 08/14/2014] [Accepted: 08/19/2014] [Indexed: 12/22/2022]
Abstract
In many regions of the world, there is a great overlap between the prevalence of cigarette smoke exposure and tuberculosis. Despite the large body of epidemiologic evidence that tobacco smoke exposure is associated with increased tuberculosis infection, active disease, severity of disease, and mortality from tuberculosis, these studies cannot distinguish whether the mechanism is principally through direct impairment of anti-tuberculosis immunity by cigarette smoke or due to potential confounders that increase risk for tuberculosis and are commonly associated with smoking--such as poverty, malnutrition, and crowded living conditions. While there are several in vivo murine and in vitro macrophage studies showing cigarette smoke impairs control of tuberculous infection, little is known of the molecular and cellular mechanisms by which this impairment occurs. Herein, we highlight the key findings of these studies. Additionally, we review key immune cells that play critical roles in host-defense or pathogenesis of tuberculosis and generate a hypothesis-driven discussion of the possible mechanisms by which cigarette smoke impairs or enhances their functions, respectively, ultimately resulting in compromised immunity against tuberculosis.
Collapse
Affiliation(s)
- Edward D Chan
- Department of Medicine, Denver Veterans Affairs Medical Center, 1055 Clermont St, Denver, CO 80220, USA; Departments of Medicine and Academic Affairs, D509, Neustadt Building, National Jewish Health, 1400 Jackson St, Denver, CO 80206, USA; Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Anschutz Medical Campus, Research 2, Box C-272, 9th Floor, 12700 East 19th Avenue, Aurora, CO 80045, USA.
| | - William H Kinney
- Departments of Medicine and Academic Affairs, D509, Neustadt Building, National Jewish Health, 1400 Jackson St, Denver, CO 80206, USA
| | - Jennifer R Honda
- Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Anschutz Medical Campus, Research 2, Box C-272, 9th Floor, 12700 East 19th Avenue, Aurora, CO 80045, USA
| | - Raju Bishwakarma
- Departments of Medicine and Academic Affairs, D509, Neustadt Building, National Jewish Health, 1400 Jackson St, Denver, CO 80206, USA
| | - Avani Gangavelli
- Departments of Medicine and Academic Affairs, D509, Neustadt Building, National Jewish Health, 1400 Jackson St, Denver, CO 80206, USA
| | - Jenny Mya
- Departments of Medicine and Academic Affairs, D509, Neustadt Building, National Jewish Health, 1400 Jackson St, Denver, CO 80206, USA
| | - Xiyuan Bai
- Departments of Medicine and Academic Affairs, D509, Neustadt Building, National Jewish Health, 1400 Jackson St, Denver, CO 80206, USA
| | - Diane J Ordway
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, 1682 Campus Delivery, Fort Collins, CO 80523, USA
| |
Collapse
|
30
|
Le V, Kim YH, Min J. The dependence of nitric oxide synthase inhibition caused by cigarette smoking extract on the cellular aging of bovine aortic endothelial cells. ENVIRONMENTAL HEALTH AND TOXICOLOGY 2014; 29:e2014010. [PMID: 25262772 PMCID: PMC4178539 DOI: 10.5620/eht.e2014010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/21/2014] [Accepted: 08/12/2014] [Indexed: 06/03/2023]
Abstract
OBJECTIVES Cigarette smoking had been recorded as the main cause of impaired endothelium- dependent vasodilation in smokers by reducing nitric oxide (NO), a production of endothelial nitric oxide synthase (eNOS). However, the mechanism of NO impairment via eNOS activity is unclear until now. In this study, cell passage is suggested to be a relevant factor to eNOS expression under cigarette smoking stress. METHODS Bovine aortic endothelial cells (BAECs) were chosen as the research subject with passages ranking from 6 to 9 (6P to 9P). After exposure of cigarette smoking extract (CSE) solution, MTT assay and Western blot method were performed to check the cell viability as well as eNOS protein concentration. In these experiments, four concentrations of CSE at 0.5, 1, 2, and 4% were selected for treatment. RESULTS Our results showed that cells almost died at 4% of CSE. Besides, eNOS protein mass had a linear decrease under the increase of CSE concentration. In addition, the effect of CSE on eNOS expression was dissimilar between different passages. CONCLUSIONS This study indicated that CSE had effect on both cell viability and eNOS expression. Besides, a reduction in protein mass was matched with the decrease of cell viability due to CSE tress. Last but not least, the response of eNOS protein to different concentration of CSE at different passages was disparate, making the hypothesis about cell passage related inhibition of eNOS caused by CSE solution.
Collapse
Affiliation(s)
- VuQuynhAnh Le
- Department of Bioprocess Engineering, Chonbuk National University, Jeonju, Korea
| | - Yang-Hoon Kim
- Department of Microbiology, Chungbuk National University, Cheongju, Korea
| | - Jiho Min
- Department of Bioprocess Engineering, Chonbuk National University, Jeonju, Korea
| |
Collapse
|
31
|
Gao Z, Nissen JC, Ji K, Tsirka SE. The experimental autoimmune encephalomyelitis disease course is modulated by nicotine and other cigarette smoke components. PLoS One 2014; 9:e107979. [PMID: 25250777 PMCID: PMC4176721 DOI: 10.1371/journal.pone.0107979] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2014] [Accepted: 08/16/2014] [Indexed: 12/12/2022] Open
Abstract
Epidemiological studies have reported that cigarette smoking increases the risk of developing multiple sclerosis (MS) and accelerates its progression. However, the molecular mechanisms underlying these effects remain unsettled. We have investigated here the effects of the nicotine and the non-nicotine components in cigarette smoke on MS using the experimental autoimmune encephalomyelitis (EAE) model, and have explored their underlying mechanism of action. Our results show that nicotine ameliorates the severity of EAE, as shown by reduced demyelination, increased body weight, and attenuated microglial activation. Nicotine administration after the development of EAE symptoms prevented further disease exacerbation, suggesting that it might be useful as an EAE/MS therapeutic. In contrast, the remaining components of cigarette smoke, delivered as cigarette smoke condensate (CSC), accelerated and increased adverse clinical symptoms during the early stages of EAE, and we identify a particular cigarette smoke compound, acrolein, as one of the potential mediators. We also show that the mechanisms underlying the opposing effects of nicotine and CSC on EAE are likely due to distinct effects on microglial viability, activation, and function.
Collapse
Affiliation(s)
- Zhen Gao
- Department of Pharmacological Sciences, School of Medicine, Stony Brook University, Stony Brook, New York, United States of America
- Program in Neuroscience, School of Medicine, Stony Brook University, Stony Brook, New York, United States of America
| | - Jillian C. Nissen
- Department of Pharmacological Sciences, School of Medicine, Stony Brook University, Stony Brook, New York, United States of America
| | - Kyungmin Ji
- Department of Pharmacological Sciences, School of Medicine, Stony Brook University, Stony Brook, New York, United States of America
| | - Stella E. Tsirka
- Department of Pharmacological Sciences, School of Medicine, Stony Brook University, Stony Brook, New York, United States of America
- Program in Neuroscience, School of Medicine, Stony Brook University, Stony Brook, New York, United States of America
| |
Collapse
|
32
|
de Vries M, Heijink IH, Gras R, den Boef LE, Reinders-Luinge M, Pouwels SD, Hylkema MN, van der Toorn M, Brouwer U, van Oosterhout AJM, Nawijn MC. Pim1 kinase protects airway epithelial cells from cigarette smoke-induced damage and airway inflammation. Am J Physiol Lung Cell Mol Physiol 2014; 307:L240-51. [DOI: 10.1152/ajplung.00156.2013] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Exposure to cigarette smoke (CS) is the main risk factor for developing chronic obstructive pulmonary disease and can induce airway epithelial cell damage, innate immune responses, and airway inflammation. We hypothesized that cell survival factors might decrease the sensitivity of airway epithelial cells to CS-induced damage, thereby protecting the airways against inflammation upon CS exposure. Here, we tested whether Pim survival kinases could protect from CS-induced inflammation. We determined expression of Pim kinases in lung tissue, airway inflammation, and levels of keratinocyte-derived cytokine (KC) and several damage-associated molecular patterns in bronchoalveolar lavage in mice exposed to CS or air. Human bronchial epithelial BEAS-2B cells were treated with CS extract (CSE) in the presence or absence of Pim1 inhibitor and assessed for loss of mitochondrial membrane potential, induction of cell death, and release of heat shock protein 70 (HSP70). We observed increased expression of Pim1, but not of Pim2 and Pim3, in lung tissue after exposure to CS. Pim1-deficient mice displayed a strongly enhanced neutrophilic airway inflammation upon CS exposure compared with wild-type controls. Inhibition of Pim1 activity in BEAS-2B cells increased the loss of mitochondrial membrane potential and reduced cell viability upon CSE treatment, whereas release of HSP70 was enhanced. Interestingly, we observed release of S100A8 but not of double-strand DNA or HSP70 in Pim1-deficient mice compared with wild-type controls upon CS exposure. In conclusion, we show that expression of Pim1 protects against CS-induced cell death in vitro and neutrophilic airway inflammation in vivo. Our data suggest that the underlying mechanism involves CS-induced release of S100A8 and KC.
Collapse
Affiliation(s)
- M. de Vries
- University of Groningen, University Medical Center Groningen, Laboratory of Allergology and Pulmonary Diseases, Department of Pathology and Medical Biology, Groningen, The Netherlands
- University of Groningen, University Medical Center Groningen, GRIAC Research Institute, Groningen, The Netherlands
| | - I. H. Heijink
- University of Groningen, University Medical Center Groningen, Laboratory of Allergology and Pulmonary Diseases, Department of Pathology and Medical Biology, Groningen, The Netherlands
- University of Groningen, University Medical Center Groningen, GRIAC Research Institute, Groningen, The Netherlands
| | - R. Gras
- University of Groningen, University Medical Center Groningen, Laboratory of Allergology and Pulmonary Diseases, Department of Pathology and Medical Biology, Groningen, The Netherlands
- University of Groningen, University Medical Center Groningen, GRIAC Research Institute, Groningen, The Netherlands
| | - L. E. den Boef
- University of Groningen, University Medical Center Groningen, Laboratory of Allergology and Pulmonary Diseases, Department of Pathology and Medical Biology, Groningen, The Netherlands
- University of Groningen, University Medical Center Groningen, GRIAC Research Institute, Groningen, The Netherlands
| | - M. Reinders-Luinge
- University of Groningen, University Medical Center Groningen, Pathology Section, Department of Pathology and Medical Biology, Groningen, The Netherlands
- University of Groningen, University Medical Center Groningen, GRIAC Research Institute, Groningen, The Netherlands
| | - S. D. Pouwels
- University of Groningen, University Medical Center Groningen, Laboratory of Allergology and Pulmonary Diseases, Department of Pathology and Medical Biology, Groningen, The Netherlands
- University of Groningen, University Medical Center Groningen, GRIAC Research Institute, Groningen, The Netherlands
| | - M. N. Hylkema
- University of Groningen, University Medical Center Groningen, Pathology Section, Department of Pathology and Medical Biology, Groningen, The Netherlands
- University of Groningen, University Medical Center Groningen, GRIAC Research Institute, Groningen, The Netherlands
| | - M. van der Toorn
- University of Groningen, University Medical Center Groningen, Laboratory of Allergology and Pulmonary Diseases, Department of Pathology and Medical Biology, Groningen, The Netherlands
- University of Groningen, University Medical Center Groningen, GRIAC Research Institute, Groningen, The Netherlands
| | - U. Brouwer
- University of Groningen, University Medical Center Groningen, Laboratory of Allergology and Pulmonary Diseases, Department of Pathology and Medical Biology, Groningen, The Netherlands
- University of Groningen, University Medical Center Groningen, GRIAC Research Institute, Groningen, The Netherlands
| | - A. J. M. van Oosterhout
- University of Groningen, University Medical Center Groningen, Laboratory of Allergology and Pulmonary Diseases, Department of Pathology and Medical Biology, Groningen, The Netherlands
- University of Groningen, University Medical Center Groningen, GRIAC Research Institute, Groningen, The Netherlands
| | - M. C. Nawijn
- University of Groningen, University Medical Center Groningen, Laboratory of Allergology and Pulmonary Diseases, Department of Pathology and Medical Biology, Groningen, The Netherlands
- University of Groningen, University Medical Center Groningen, GRIAC Research Institute, Groningen, The Netherlands
| |
Collapse
|
33
|
Comer DM, Elborn JS, Ennis M. Inflammatory and cytotoxic effects of acrolein, nicotine, acetylaldehyde and cigarette smoke extract on human nasal epithelial cells. BMC Pulm Med 2014; 14:32. [PMID: 24581246 PMCID: PMC3945717 DOI: 10.1186/1471-2466-14-32] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2013] [Accepted: 02/25/2014] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Cigarette smoke induces a pro-inflammatory response in airway epithelial cells but it is not clear which of the various chemicals contained within cigarette smoke (CS) should be regarded as predominantly responsible for these effects. We hypothesised that acrolein, nicotine and acetylaldehyde, important chemicals contained within volatile cigarette smoke in terms of inducing inflammation and causing addiction, have immunomodulatory effects in primary nasal epithelial cell cultures (PNECs). METHODS PNECs from 19 healthy subjects were grown in submerged cultures and were incubated with acrolein, nicotine or acetylaldehyde prior to stimulation with Pseudomonas aeruginosa lipopolysaccharide (PA LPS). Experiments were repeated using cigarette smoke extract (CSE) for comparison. IL-8 was measured by ELISA, activation of NF-κB by ELISA and Western blotting, and caspase-3 activity by Western blotting. Apoptosis was evaluated using Annexin-V staining and the terminal transferase-mediated dUTP nick end-labeling (TUNEL) method. RESULTS CSE was pro-inflammatory after a 24 h exposure and 42% of cells were apoptotic or necrotic after this exposure time. Acrolein was pro-inflammatory for the PNEC cultures (30 μM exposure for 4 h inducing a 2.0 fold increase in IL-8 release) and also increased IL-8 release after stimulation with PA LPS. In contrast, nicotine had anti-inflammatory properties (0.6 fold IL-8 release after 50 μM exposure to nicotine for 24 h), and acetylaldehyde was without effect. Acrolein and nicotine had cellular stimulatory and anti-inflammatory effects respectively, as determined by NF-κB activation. Both chemicals increased levels of cleaved caspase 3 and induced cell death. CONCLUSIONS Acrolein is pro-inflammatory and nicotine anti-inflammatory in PNEC cultures. CSE induces cell death predominantly by apoptotic mechanisms.
Collapse
Affiliation(s)
- David M Comer
- Centre for Infection and Immunity, Health Sciences Building, School of Medicine, Dentistry and Biomedical Sciences, Queen’s University of Belfast, 97 Lisburn Road, Belfast BT9 7BL, UK
- Respiratory Department, Belfast City Hospital, Lisburn Road, Belfast BT9 7AB, UK
| | - Joseph Stuart Elborn
- Centre for Infection and Immunity, Health Sciences Building, School of Medicine, Dentistry and Biomedical Sciences, Queen’s University of Belfast, 97 Lisburn Road, Belfast BT9 7BL, UK
- Respiratory Department, Belfast City Hospital, Lisburn Road, Belfast BT9 7AB, UK
| | - Madeleine Ennis
- Centre for Infection and Immunity, Health Sciences Building, School of Medicine, Dentistry and Biomedical Sciences, Queen’s University of Belfast, 97 Lisburn Road, Belfast BT9 7BL, UK
| |
Collapse
|
34
|
Bozinovski S, Anthony D, Anderson GP, Irving LB, Levy BD, Vlahos R. Treating neutrophilic inflammation in COPD by targeting ALX/FPR2 resolution pathways. Pharmacol Ther 2013; 140:280-9. [PMID: 23880288 DOI: 10.1016/j.pharmthera.2013.07.007] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2013] [Accepted: 07/09/2013] [Indexed: 12/19/2022]
Abstract
Neutrophilic inflammation persists in COPD despite best current therapies and it is particularly resistant to inhaled glucocorticosteroids. Persistent neutrophil activation not only contributes to matrix breakdown, but can maintain inflammation through the release of endogenous damage associated molecule patterns (DAMPs). Inhibiting excessive neutrophilic inflammation is challenging as many pathogen recognition receptors can initiate migration and the targeting of downstream signaling molecules may compromise essential host defense mechanisms. Here, we discuss new strategies to combat this inflammation in COPD by focusing on the anti-inflammatory role of ALX/FPR2 receptors. ALX/FPR2 is a promiscuous G-protein coupled receptor (GPCR) responding to lipid and peptide agonists that can either switch on acute inflammation or promote resolution of inflammation. We highlight this receptor as an emerging target in the pathogenesis of COPD because known ALX/FPR2 endogenous agonists are enriched in COPD. Serum Amyloid A (SAA) has recently been discovered to be abundantly expressed in COPD and is a potent ALX/FPR2 agonist that unlike almost all other inflammatory chemoattractants, is induced by glucocorticosteroids. SAA not only initiates lung inflammation via ALX/FPR2 but can allosterically modify this receptor so that it no longer transduces pro-resolving signals from endogenous lipoxins that would otherwise promote tissue healing. We propose that there is an imbalance in endogenous and microbial ALX/FPR2 receptor agonists in the inflamed COPD lung environment that oppose protective anti-inflammatory and pro-resolution pathways. These insights open the possibility of targeting ALX/FPR2 receptors using synthetic agonists to resolve persistent neutrophilic inflammation without compromising essential host defense mechanisms.
Collapse
Affiliation(s)
- Steven Bozinovski
- Department of Pharmacology and Therapeutics, The University of Melbourne, Victoria, Australia.
| | | | | | | | | | | |
Collapse
|
35
|
Mato N, Bando M, Kusano A, Hirano T, Nakayama M, Uto T, Nakaya T, Yamasawa H, Sugiyama Y. Clinical significance of interleukin 33 (IL-33) in patients with eosinophilic pneumonia. Allergol Int 2013; 62:45-52. [PMID: 23000728 DOI: 10.2332/allergolint.12-oa-0439] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2012] [Accepted: 05/31/2012] [Indexed: 01/17/2023] Open
Abstract
BACKGROUND Interleukin 33 (IL-33) works as a functional mediator in allergic disease by enhancing the activity of eosinophils and inducing expression of T helper 2 (Th2)-associated cytokines. However, the role of IL-33 in pulmonary eosinophilia has not been elucidated. We investigated the levels of IL-33 in eosinophilic pneumonia (EP) together with associated cytokines, and discussed the clinical significance of IL-33 in EP. METHODS Sera and bronchoalveolar lavage fluid (BALF) were obtained from 16 patients with EP, including acute eosinophilic pneumonia (AEP) and chronic eosinophilic pneumonia (CEP). Twelve patients with acute respiratory distress syndrome (ARDS) were also included for comparison. The concentration of IL-33 and Th2 cytokines (IL-4, IL-5, IL-13) were measured by enzyme-linked immunosorbent assay (ELISA). RESULTS The concentration of serum IL-33 was significantly higher in patients with AEP than in CEP. In CEP, only patients with atopic factors showed mild increase of serum IL-33. The concentration of BALF IL-33 was also significantly elevated in AEP, however, it remained quite low in CEP. Among Th2 cytokines, IL-5 was significantly increased in both serum and BALF in AEP, and the level of IL-5 was positively correlated with that of IL-33. ARDS showed no increase of serum and BALF IL-33. CONCLUSIONS The remarkable increase of BALF IL-33 in AEP indicated the local production of IL-33 in lungs. IL-33 is considered to be a local key molecule for triggering pulmonary eosinophilia, together with IL-5. BALF IL-33 appears to be a useful marker for discriminating AEP from CEP and ARDS.
Collapse
Affiliation(s)
- Naoko Mato
- Division of Pulmonary Medicine, Department of Medicine, Jichi Medical University, Tochigi, Japan.
| | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Hernandez CP, Morrow K, Velasco C, Wyczechowska DD, Naura A, Rodriguez PC. Effects of cigarette smoke extract on primary activated T cells. Cell Immunol 2013; 282:38-43. [PMID: 23665673 PMCID: PMC3676722 DOI: 10.1016/j.cellimm.2013.04.005] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2012] [Revised: 02/15/2013] [Accepted: 04/14/2013] [Indexed: 12/29/2022]
Abstract
Tobacco smoking predisposes the development of diseases characterized by chronic inflammation and T cell dysfunction. In this study, we aimed to determine the direct effects of cigarette smoke on primary T cells and to identify the corresponding molecular mediators. Activated T cells cultured in the presence of cigarette smoke extract (CSE) displayed a dose-dependent decrease in cell proliferation, which associated with the induction of cellular apoptosis. T cell apoptosis by CSE was independent of caspases and mediated through reactive oxygen and nitrogen species endogenously contained within CSE. Additional results showed that exposure of T cells to CSE induced phosphorylation of the stress mediator eukaryotic-translation-initiation-factor 2 alpha (eIF2α). Inhibition of the phosphorylation of eIF2α in T cells prevented the cellular apoptosis induced by CSE. Altogether, the results show the direct effects of CSE on T cells, which advance in the understanding of how cigarette smoking promotes chronic inflammation and immune dysfunction.
Collapse
Affiliation(s)
- Claudia P. Hernandez
- Stanley S. Scott Cancer Center, Louisiana State University-Health Sciences Center, New Orleans, LA, 70112
| | - Kevin Morrow
- Stanley S. Scott Cancer Center, Louisiana State University-Health Sciences Center, New Orleans, LA, 70112
| | - Cruz Velasco
- Stanley S. Scott Cancer Center, Louisiana State University-Health Sciences Center, New Orleans, LA, 70112
| | - Dorota D. Wyczechowska
- Stanley S. Scott Cancer Center, Louisiana State University-Health Sciences Center, New Orleans, LA, 70112
| | - Amarjit Naura
- Stanley S. Scott Cancer Center, Louisiana State University-Health Sciences Center, New Orleans, LA, 70112
| | - Paulo C. Rodriguez
- Stanley S. Scott Cancer Center, Louisiana State University-Health Sciences Center, New Orleans, LA, 70112
- Department of Microbiology Immunology and Parasitology, Louisiana State University-Health Sciences Center, New Orleans, LA, 70112
| |
Collapse
|
37
|
Abstract
Cigarette smoke is an aerosol that contains >4,000 chemicals, including nicotine, carbon monoxide, acrolein, and oxidant compounds. Exposure to cigarette smoke induces multiple pathological effects in the endothelium, several of which are the result of oxidative stress initiated by reactive oxygen species, reactive nitrogen species, and other oxidant constituents of cigarette smoke. Cigarette-smoke exposure interferes adversely with the control of all stages of plaque formation and development and pathological thrombus formation. The reactive oxygen species in cigarette smoke contribute to oxidative stress, upregulation of inflammatory cytokines, and endothelial dysfunction, by reducing the bioavailability of nitric oxide. Plaque formation and the development of vulnerable plaques also result from exposure to cigarette smoke via the enhancement of inflammatory processes and the activation of matrix metalloproteases. Moreover, exposure to cigarette smoke results in platelet activation, stimulation of the coagulation cascade, and impairment of anticoagulative fibrinolysis. Many cigarette-smoke-mediated prothrombotic changes are quickly reversible upon smoking cessation. Public health efforts should urgently promote our understanding of current cigarette-smoke-induced cardiovascular pathology to encourage individuals to reduce their exposure to cigarette smoke and, therefore, the detrimental consequences of associated atherothrombotic disease.
Collapse
Affiliation(s)
- Adam Csordas
- Division of Cardiovascular Surgery, University Hospital Zürich, Raemistrasse 100, CH-8091 Zürich, Switzerland
| | | |
Collapse
|
38
|
Arimilli S, Damratoski BE, Bombick B, Borgerding MF, Prasad G. Evaluation of cytotoxicity of different tobacco product preparations. Regul Toxicol Pharmacol 2012; 64:350-60. [DOI: 10.1016/j.yrtph.2012.09.004] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2012] [Revised: 08/20/2012] [Accepted: 09/11/2012] [Indexed: 01/01/2023]
|
39
|
Tosiek MJ, Bader SR, Gruber AD, Buer J, Gereke M, Bruder D. CD8(+) T cells responding to alveolar self-antigen lack CD25 expression and fail to precipitate autoimmunity. Am J Respir Cell Mol Biol 2012; 47:869-78. [PMID: 22984087 DOI: 10.1165/rcmb.2011-0387oc] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Although the contribution of CD8(+) T cells to the pathogenesis of noncommunicable lung diseases has become increasingly appreciated, our knowledge about the mechanisms controlling self-reactive CD8(+) T cells in the respiratory tract remains largely elusive. The outcome of the encounter between pulmonary self-antigen and naive CD8(+) T cells, in the presence or absence of inflammation, was traced after adoptive transfer of fluorescence-labeled CD8(+) T cells specific for the neo-self-antigen influenza A hemagglutinin into transgenic mice expressing hemagglutinin specifically in alveolar type II epithelial cells in order: to study the outcome of alveolar antigen encounter in the steady state and under inflammatory conditions; to define the phenotype and fate of CD8(+) T cells primed in the respiratory tract; and, finally, to correlate these findings with the onset of autoimmunity in the lung. We found that CD8(+) T cells remain ignorant in the steady state, whereas transient proliferation of self-reactive CD8(+) T cells is induced by forced maturation or licensing of dendritic cells, increases in the antigenic threshold, and targeted release of alveolar self-antigen by epithelial injury. However, these cells fail to acquire effector functions, lack the expression of the high-affinity IL-2 receptor CD25, and do not precipitate autoimmunity in the lung. We conclude that inadvertent activation of CD8(+) T cells in the lung is prevented in the absence of "danger signals," whereas tissue damage after infection or noninfectious inflammation creates an environment that allows the priming of previously ignorant T cells. Failure in effector cell differentiation after abortive priming, however, precludes the establishment of self-perpetuating autoimmunity in the lung.
Collapse
Affiliation(s)
- Milena J Tosiek
- Immune Regulation Group, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | | | | | | | | | | |
Collapse
|
40
|
Gualerzi A, Sciarabba M, Tartaglia G, Sforza C, Donetti E. Acute effects of cigarette smoke on three-dimensional cultures of normal human oral mucosa. Inhal Toxicol 2012; 24:382-9. [PMID: 22564096 DOI: 10.3109/08958378.2012.679367] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
CONTEXT Human oral mucosa is the combustion chamber of cigarette, but scanty evidence is available about the early smoke effects. OBJECTIVE The present work aimed at evaluating from a morphological point of view whole smoke early effects on epithelial intercellular adhesion and keratinocyte terminal differentiation in a three-dimensional model of human oral mucosa. MATERIALS AND METHODS Biopsies of keratinized oral mucosa of healthy nonsmoking women (n = 5) were collected. After culturing in a Transwell system, one fragment of each biopsy was exposed to the smoke of one single cigarette; the remnant represented the internal control. The distribution of epithelial differentiation markers (keratin-10, K10, and keratin-14, K14, for suprabasal and basal cells respectively), desmosomes (desmoglein-1, desmoglein-3), tight junctions (occludin), adherens junctions (E-cadherin, β-catenin), and apoptotic cells (p53, caspase 3) were evaluated by immunofluorescence. RESULTS Quantitative analysis of K14 immunolabeling revealed an overexpression in the suprabasal layers as early as 3 h after smoke exposure, without impairment of the epithelial junctional apparatus and apoptosis induction. DISCUSSION AND CONCLUSION These results suggested that the first significant response to cigarette smoke came from the basal and suprabasal layers of the human oral epithelium. The considered model maintained the three-dimensional arrangement of the human mucosa in the oral cavity and mimicked the inhalation/exhalation cycle during the exposure to cigarette smoke, offering a good possibility to extrapolate the reported observations to humans.
Collapse
Affiliation(s)
- Alice Gualerzi
- Dipartimento di Morfologia Umana e Scienze Biomediche - Città Studi, Italy
| | | | | | | | | |
Collapse
|
41
|
Reddy NM, Vegiraju S, Irving A, Paun BC, Luzina IG, Atamas SP, Biswal S, Ana NA, Mitzner W, Reddy SP. Targeted deletion of Jun/AP-1 in alveolar epithelial cells causes progressive emphysema and worsens cigarette smoke-induced lung inflammation. THE AMERICAN JOURNAL OF PATHOLOGY 2012; 180:562-74. [PMID: 22265050 DOI: 10.1016/j.ajpath.2011.10.029] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/24/2011] [Revised: 10/12/2011] [Accepted: 10/27/2011] [Indexed: 10/14/2022]
Abstract
Chronic obstructive pulmonary disease appears to occur slowly and progressively over many years, with both genetic factors and environmental modifiers contributing to its pathogenesis. Although the c-Jun/activator protein 1 transcriptional factor regulates cell proliferation, apoptosis, and inflammatory responses, its role in lung pathogenesis is largely unknown. In this study, we report decreased expression levels of c-Jun mRNA and protein in the lung tissues of patients with advanced chronic obstructive pulmonary disease, and the genetic deletion of c-Jun specifically in alveolar epithelial cells causes progressive emphysema with lung inflammation and alveolar air space enlargement, which are cardinal features of emphysema. Although mice lacking c-Jun specifically in lung alveolar epithelial cells appear normal at the age of 6 weeks, when exposed to long-term cigarette smoke, c-Jun-mutant mice display more lung inflammation with perivascular and peribronchiolar infiltrates compared with controls. These results demonstrate that the c-Jun/activator protein 1 pathway is critical for maintaining lung alveolar cell homeostasis and that loss of its expression can contribute to lung inflammation and progressive emphysema.
Collapse
Affiliation(s)
- Narsa M Reddy
- Department of Environmental Health Sciences, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Zhang HM, Nie JS, Li X, Niu Q. Characteristic analysis of peripheral blood mononuclear cell apoptosis in coke oven workers. J Occup Health 2011; 54:44-50. [PMID: 22186298 DOI: 10.1539/joh.11-0155-oa] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
OBJECTIVES The aim of the present study was to determine the peripheral blood mononuclear cell (PBMC) apoptosis in coke oven workers so that we can take effective measures to protect coke oven workers. METHODS The subjects, 129 coke oven workers and 37 warehouse workers (controls), were investigated using a questionnaire to collect information about their age, working years, smoking and drinking habits, vocational history and other general information. The coke oven workers were divided into the oven-bottom group (34), oven-side group (48) and oven-top group (47) according to their working sites and environmental monitoring data. The concentration of benzo[a]pyrene (B[a]P) and the subjects' urinary 1-hydroxypyrene (1-OH-Py) levels were determined by HPLC. Additionally, the PBMCs were separated from blood samples, and the early and late apoptosis rates were determined by flow cytometry. RESULTS The airborne B[a]P concentrations were 19.5 ± 13.2, 185.9 ± 38.6 and 1,623.5 ± 435.8 ng/m(3) at the bottom, side and top of the oven, respectively, and were higher than in the controls' workplaces 10.2 ± 7.6 ng/m(3). Urinary 1-OH-Py, indicating the B[a]P's internal exposure level, was significantly higher in the exposed groups than in the controls (p<0.05). Compared with the controls, the coke oven workers' PBMC apoptosis rates were significantly increased and increased in association with the B[a]P level. PBMC apoptosis increased in association with the 1-OH-Py level and coking operation years and decreased in association with years of alcohol consumption. CONCLUSIONS PBMC apoptosis in the coke oven workers was associated with the 1-OH-Py level, coke operation years and years of alcohol consumption and may be induced by B[a]P.
Collapse
Affiliation(s)
- Hong Mei Zhang
- Department of Occupational and Environmental Health, Shanxi Medical University
| | | | | | | |
Collapse
|
43
|
Kim BS, Kim SJ, Kim HJ, Lee SJ, Park YJ, Lee J, You HK. Effects of nicotine on proliferation and osteoblast differentiation in human alveolar bone marrow-derived mesenchymal stem cells. Life Sci 2011; 90:109-15. [PMID: 22115820 DOI: 10.1016/j.lfs.2011.10.019] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2011] [Revised: 09/20/2011] [Accepted: 10/11/2011] [Indexed: 01/13/2023]
Abstract
AIMS Nicotine is a risk factor for various diseases, including osteoporosis, oral cancer, and periodontal disease. Numerous studies have elucidated the effects of nicotine on cell proliferation and differentiation. The purpose of this study was to determine the effects of nicotine on the proliferation and osteoblast differentiation of human alveolar bone marrow-derived mesenchymal stem cells (hABMMSCs). MAIN METHODS In this study, we treated hABMMSCs with different doses (1 μM to 5 mM) of nicotine. The survival and proliferation of hABMMSCs were evaluated by the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium (MTT) assay and crystal violet assay. TUNEL and propidium iodide (PI) double staining assay were also performed. The effect of nicotine on osteoblast differentiation of hABMMSCs was determined by measuring calcium accumulation using alizarin red-sulfate (AR-S) staining, measurement of alkaline phosphatase (ALP) activity, and semi-quantitative PCR of osteoblast markers. KEY FINDINGS The survival and proliferation of hABMMSCs did not differ when they were exposed to nicotine at concentrations ranging from 1 μM to 100 μM; however, cell proliferation increased when the cells were exposed to nicotine at concentrations of 1-2 mM and decreased significantly when exposed to 5mM of nicotine. A number of cells were stained by PI but not by TUNEL, and membrane vacuolization was observed in hABMMSCs treated with 5mM nicotine. Calcium accumulation; ALP activity; and mRNA levels of ALP, bone sialoprotein (BSP), collagen type I α 1 (Col1αI), and runt-related transcription factor 2 (Runx2) were significantly decreased by treatment with 2mM of nicotine, while osteocalcin transcripts decreased by treatment with 1 to 2 mM of nicotine. SIGNIFICANCE These results suggest that nicotine has a bimodal effect on the proliferation and osteoblast differentiation in hABMMSCs.
Collapse
Affiliation(s)
- Beom-Su Kim
- Wonkwang Bone Regeneration Research Institute, Wonkwang University, Iksan 570-749, Republic of Korea
| | | | | | | | | | | | | |
Collapse
|
44
|
Karavitis J, Kovacs EJ. Macrophage phagocytosis: effects of environmental pollutants, alcohol, cigarette smoke, and other external factors. J Leukoc Biol 2011; 90:1065-78. [PMID: 21878544 DOI: 10.1189/jlb.0311114] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
The ability of a pathogen to evade host immunity successfully, in contrast to the host's capacity to defend itself against a foreign invader, is a complex struggle, in which eradication of infection is dictated by a robust immunologic response. Often, there are external factors that can alter the outcome by tipping the scale to benefit pathogen establishment rather than resolution by the host's defense system. These external sources, such a cigarettes, alcohol, or environmental pollutants, can negatively influence the effectiveness of the immune system's response to a pathogen. The observed suppression of immune function can be attributed to dysregulated cytokine and chemokine production, the loss of migratory potential, or the inability to phagocytose pathogens by immune cells. This review will focus on the mechanisms involved during the toxin-induced suppression of phagocytosis. The accumulated data support the importance of studying the mechanisms of phagocytosis following exposure to these factors, in that this effect alone cannot only leave the host susceptible to infection but also promote alterations in many other macrophage functions necessary for pathogen clearance and restoration of homeostasis.
Collapse
Affiliation(s)
- John Karavitis
- Program of Cell Biology, Neurobiology and Anatomy, Loyola University Medical Center, Maywood, Illinois, USA
| | | |
Collapse
|
45
|
Chen ZL, Tao J, Yang J, Yuan ZL, Liu XH, Jin M, Shen ZQ, Wang L, Li HF, Qiu ZG, Wang JF, Wang XW, Li JW. Vitamin E modulates cigarette smoke extract-induced cell apoptosis in mouse embryonic cells. Int J Biol Sci 2011; 7:927-36. [PMID: 21850202 PMCID: PMC3157267 DOI: 10.7150/ijbs.7.927] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2011] [Accepted: 07/24/2011] [Indexed: 01/09/2023] Open
Abstract
Vitamin E (VE) can effectively prevent occurrence of lung cancer caused by passive smoking in mice. However, whether VE prevents smoking-induced cytotoxicity remains unclear. In this study, a primary culture of embryonic lung cells (ELCs) was used to observe the cytotoxic effects of cigarette smoke extract (CSE), including its influence on cell survival, cell cycle, apoptosis, and DNA damage, and also to examine the effects of VE intervention on CSE-induced cytotoxicity. Our results showed that CSE could significantly inhibit the survival of ELCs with dose- and time-dependent effects. Furthermore, CSE clearly disturbed the cell cycle of ELCs by decreasing the proportion of cells at the S and G₂/M phases and increasing the proportion of cells at the G₀/G₁ phase. CSE promoted cell apoptosis, with the highest apoptosis rate reaching more than 40%. CSE also significantly caused DNA damage of ELCs. VE supplementation could evidently inhibit or reverse the cytotoxic effects of CSE in a dose- and time-dependent manner. The mechanism of CSE effects on ELCs and that of VE intervention might involve the mitochondrial pathway of cytochrome c-mediated caspase activation. Our study validate that VE plays a clearly protective effect against CSE-induced cytotoxicity in mouse embryonic lung cells.
Collapse
Affiliation(s)
- Zhao-Li Chen
- 1. Department of Health and Environment, Institute of Health and Environmental Medicine; Key Laboratory of Risk Assessment and Control for Environment & Food Safety, No.1, Dali Road, Tianjin, 300050, P. R. China
| | - Jian Tao
- 2. Food Sci-Eng College, Northwest A & F University, Yangling, Shanxi Province, 712100, P. R. China
| | - Jie Yang
- 3. Department of Chronic Disease, Chinese Center for Disease Control and Prevention, Beijing, 100050, P. R. China
| | - Zhen-Li Yuan
- 4. School of Public Health, Jilin University, Changchun, 130021, P. R. China
| | - Xing-Hua Liu
- 2. Food Sci-Eng College, Northwest A & F University, Yangling, Shanxi Province, 712100, P. R. China
| | - Min Jin
- 1. Department of Health and Environment, Institute of Health and Environmental Medicine; Key Laboratory of Risk Assessment and Control for Environment & Food Safety, No.1, Dali Road, Tianjin, 300050, P. R. China
| | - Zhi-Qiang Shen
- 1. Department of Health and Environment, Institute of Health and Environmental Medicine; Key Laboratory of Risk Assessment and Control for Environment & Food Safety, No.1, Dali Road, Tianjin, 300050, P. R. China
| | - Lu Wang
- 3. Department of Chronic Disease, Chinese Center for Disease Control and Prevention, Beijing, 100050, P. R. China
| | - Hai-Feng Li
- 1. Department of Health and Environment, Institute of Health and Environmental Medicine; Key Laboratory of Risk Assessment and Control for Environment & Food Safety, No.1, Dali Road, Tianjin, 300050, P. R. China
| | - Zhi-Gang Qiu
- 1. Department of Health and Environment, Institute of Health and Environmental Medicine; Key Laboratory of Risk Assessment and Control for Environment & Food Safety, No.1, Dali Road, Tianjin, 300050, P. R. China
| | - Jing-Feng Wang
- 1. Department of Health and Environment, Institute of Health and Environmental Medicine; Key Laboratory of Risk Assessment and Control for Environment & Food Safety, No.1, Dali Road, Tianjin, 300050, P. R. China
| | - Xin-Wei Wang
- 1. Department of Health and Environment, Institute of Health and Environmental Medicine; Key Laboratory of Risk Assessment and Control for Environment & Food Safety, No.1, Dali Road, Tianjin, 300050, P. R. China
| | - Jun-Wen Li
- 1. Department of Health and Environment, Institute of Health and Environmental Medicine; Key Laboratory of Risk Assessment and Control for Environment & Food Safety, No.1, Dali Road, Tianjin, 300050, P. R. China
| |
Collapse
|
46
|
Chang X, Ravi R, Pham V, Bedi A, Chatterjee A, Sidransky D. Adenylate kinase 3 sensitizes cells to cigarette smoke condensate vapor induced cisplatin resistance. PLoS One 2011; 6:e20806. [PMID: 21698293 PMCID: PMC3115955 DOI: 10.1371/journal.pone.0020806] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2010] [Accepted: 05/12/2011] [Indexed: 11/24/2022] Open
Abstract
BACKGROUND The major established etiologic risk factor for bladder cancer is cigarette smoking and one of the major antineoplastic agents used for the treatment of advanced bladder cancer is cisplatin. A number of reports have suggested that cancer patients who smoke while receiving treatment have lower rates of response and decreased efficacy of cancer therapies. METHODOLOGY/PRINCIPAL FINDINGS In this study, we investigated the effect of cigarette smoke condensate (CSC) vapor on cisplatin toxicity in urothelial cell lines SV-HUC-1 and SCaBER cells. We showed that chronic exposure to CSC vapor induced cisplatin resistance in both cell lines. In addition, we found that the expression of mitochondrial-resident protein adenylate kinase-3 (AK3) is decreased by CSC vapor. We further observed that chronic CSC vapor-exposed cells displayed decreased cellular sensitivity to cisplatin, decreased mitochondrial membrane potential (ΔΨm) and increased basal cellular ROS levels compared to unexposed cells. Re-expression of AK3 in CSC vapor-exposed cells restored cellular sensitivity to cisplatin. Finally, CSC vapor increased the growth of the tumors and also curtail the response of tumor cells to cisplatin chemotherapy in vivo. CONCLUSIONS/SIGNIFICANCE The current study provides evidence that chronic CSC vapor exposure affects AK3 expression and renders the cells resistant to cisplatin.
Collapse
Affiliation(s)
- Xiaofei Chang
- Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Rajani Ravi
- Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Vui Pham
- Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Atul Bedi
- Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Aditi Chatterjee
- Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - David Sidransky
- Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| |
Collapse
|
47
|
Misaka S, Sato H, Aoki Y, Mizumoto T, Onoue S, Yamada S. Novel vasoactive intestinal peptide derivatives with improved stability protect rat alveolar L2 cells from cigarette smoke-induced cytotoxicity and apoptosis. Peptides 2011; 32:401-7. [PMID: 20977915 DOI: 10.1016/j.peptides.2010.10.021] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2010] [Revised: 10/17/2010] [Accepted: 10/18/2010] [Indexed: 11/24/2022]
Abstract
Vasoactive intestinal peptide (VIP) has been thought to be a promising candidate for asthma/chronic obstructive pulmonary disease (COPD), and our group previously developed several long-lasting VIP derivatives. The objective of the present study was to clarify the therapeutic potential of new VIP derivatives with improved chemical and metabolic stability. Exposure of rat alveolar L2 cells to cigarette smoke extract (CSE) for 1h led to release of lactate dehydrogenase (LDH) and decreased viability in a CSE concentration-dependent manner. There appeared to be marked induction of apoptosis after CSE exposure, as demonstrated by 59% elevation of caspase-3 activity and TUNEL staining. In contrast, a stabilized VIP derivative, [R(15,20,21), L(17)]-VIP-GRR (IK312532), at a concentration of 10(-7)M, exhibited 71% attenuation of LDH release and 85% decrease of the number of apoptotic cells. In addition to IK312532, new VIP derivatives also showed anti-apoptotic effects against CSE toxicity and marked reduction of nitric oxide production. In terms of cytoprotective effects, [R(15,20,21), L(17), A(24,25), des-N(28)]-VIP-GRR was more effective than VIP and IK312532, possibly due to the improved stability. Thus, the present study is the first to demonstrate that novel stabilized VIP derivatives exert anti-apoptotic and cytoprotective effects on CSE-induced cytotoxicity.
Collapse
Affiliation(s)
- Shingen Misaka
- Department of Pharmacokinetics, University of Shizuoka, Suruga-ku, Shizuoka 422-8526, Japan
| | | | | | | | | | | |
Collapse
|
48
|
Fujiwara M, Hamatake Y, Arimoto S, Okamoto K, Suzuki T, Negishi T. Exposure to Cigarette Smoke Increases Urate Level and Decreases Glutathione Level in Larval Drosophila melanogaster. Genes Environ 2011. [DOI: 10.3123/jemsge.33.89] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
|
49
|
Abstract
It is becoming evident that failure in the removal of dying cells causes and/or promotes the onset of chronic diseases. Impairment of phagocytosis of apoptotic cells can be due not only to genetic or molecular malfunctioning but also to external/environmental factors. Two of these environmental factors have been recently reported to down regulate the clearance of apoptotic cells: cigarette smoke and static magnetic fields. Cigarette smoke contains highly reactive carbonyls that modify proteins which directly/indirectly affects cellular function. Human macrophages interacting with carbonyl or cigarette smoke modified extracellular matrix (ECM) proteins dramatically down regulated their ability to phagocytose apoptotic neutrophils. It was postulated that changes in the ECM environment as a result of cigarette smoke affect the ability of macrophages to remove apoptotic cells. This decreased phagocytic activity was as a result of sequestration of receptors involved in the uptake of apoptotic cells towards that of recognition of carbonyl adducts on the modified ECM proteins leading to increased macrophage adhesion. Downregulation of the phagocytosis of apoptotic cells was also described when performed in presence of static magnetic fields (SMFs) of moderate intensity. SMFs have been reported to perturb distribution of membrane proteins and glycoproteins, receptors, cytoskeleton and trans-membrane fluxes of different ions, especially calcium [Ca(2+)]i, that in turn, interfere with many different physiological activities, including phagocytosis. The effects of cigarette smoke and SMF on the phagocytosis of dying cells will be here discussed.
Collapse
Affiliation(s)
- Luciana Dini
- Department Biological and Environmental Science and Technology, University of the Salento, Lecce, Italy.
| |
Collapse
|
50
|
Hwang JW, Chung S, Sundar IK, Yao H, Arunachalam G, McBurney MW, Rahman I. Cigarette smoke-induced autophagy is regulated by SIRT1-PARP-1-dependent mechanism: implication in pathogenesis of COPD. Arch Biochem Biophys 2010; 500:203-9. [PMID: 20493163 DOI: 10.1016/j.abb.2010.05.013] [Citation(s) in RCA: 125] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2010] [Revised: 05/14/2010] [Accepted: 05/14/2010] [Indexed: 12/19/2022]
Abstract
Autophagy is a fundamental cellular process that eliminates long-lived proteins and damaged organelles through lysosomal degradation pathway. Cigarette smoke (CS)-mediated oxidative stress induces cytotoxic responses in lung cells. However, the role of autophagy and its mechanism in CS-mediated cytotoxic responses is not known. We hypothesized that NAD(+)-dependent deacetylase, sirtuin 1 (SIRT1) plays an important role in regulating autophagy in response to CS. CS exposure resulted in induction of autophagy in lung epithelial cells, fibroblasts and macrophages. Pretreatment of cells with SIRT1 activator resveratrol attenuated CS-induced autophagy whereas SIRT1 inhibitor, sirtinol, augmented CS-induced autophagy. Elevated levels of autophagy were induced by CS in the lungs of SIRT1 deficient mice. Inhibition of poly(ADP-ribose)-polymerase-1 (PARP-1) attenuated CS-induced autophagy via SIRT1 activation. These data suggest that the SIRT1-PARP-1 axis plays a critical role in the regulation of CS-induced autophagy and have important implications in understanding the mechanisms of CS-induced cell death and senescence.
Collapse
Affiliation(s)
- Jae-woong Hwang
- Department of Environmental Medicine, University of Rochester Medical Center, NY, USA
| | | | | | | | | | | | | |
Collapse
|