1
|
Heydemann L, Ciurkiewicz M, Störk T, Zdora I, Hülskötter K, Gregor KM, Michaely LM, Reineking W, Schreiner T, Beythien G, Volz A, Tuchel T, Meyer Zu Natrup C, Schünemann LM, Clever S, Henneck T, von Köckritz-Blickwede M, Schaudien D, Rohn K, Schughart K, Geffers R, Kaneko MK, Kato Y, Gross C, Amanakis G, Pavlou A, Baumgärtner W, Armando F. Respiratory long COVID in aged hamsters features impaired lung function post-exercise with bronchiolization and fibrosis. Nat Commun 2025; 16:2080. [PMID: 40021627 PMCID: PMC11871369 DOI: 10.1038/s41467-025-57267-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Accepted: 02/14/2025] [Indexed: 03/03/2025] Open
Abstract
Long-term consequences of SARS-CoV-2 infection affect millions of people and strain public health systems. The underlying pathomechanisms remain unclear, necessitating further research in appropriate animal models. This study aimed to characterize the trajectory of lung regeneration over 112 days in the male hamster model by combining morphological, transcriptomic and functional readouts. We demonstrate that in the acute phase, SARS-CoV-2 Delta-infected, male, aged hamsters show a severe impairment of lung function at rest. In the chronic phase, similar impairments persisted up to 7 weeks post-infection but were only evident after exercise on a rodent treadmill. The male hamster model recapitulates chronic pulmonary fibrotic changes observed in many patients with respiratory long COVID, but lacks extra-pulmonary long-term lesions. We show that sub-pleural and interstitial pulmonary fibrosis as well as alveolar bronchiolization persist until 112 dpi. Interestingly, CK8+ alveolar differentiation intermediate (ADI) cells are becoming less prominent in the alveolar proliferation areas from 28 dpi on. Instead, CK14+ airway basal cells and SCGB1A1+ club cells, expressing cell proliferation markers, mainly populate alveolar bronchiolization areas at later time-points. We postulate that pulmonary fibrosis and SCGB1A1+ club cell-rich areas of alveolar bronchiolization represent potential risk factors for other diseases in long-COVID survivors.
Collapse
Affiliation(s)
- Laura Heydemann
- Department of Pathology, University of Veterinary Medicine Foundation, Hanover, Germany
| | | | - Theresa Störk
- Department of Pathology, University of Veterinary Medicine Foundation, Hanover, Germany
| | - Isabel Zdora
- Department of Pathology, University of Veterinary Medicine Foundation, Hanover, Germany
| | - Kirsten Hülskötter
- Department of Pathology, University of Veterinary Medicine Foundation, Hanover, Germany
| | | | | | - Wencke Reineking
- Department of Pathology, University of Veterinary Medicine Foundation, Hanover, Germany
| | - Tom Schreiner
- Department of Pathology, University of Veterinary Medicine Foundation, Hanover, Germany
| | - Georg Beythien
- Department of Pathology, University of Veterinary Medicine Foundation, Hanover, Germany
| | - Asisa Volz
- Department of Virology, University of Veterinary Medicine Foundation, Hanover, Germany
- Research Center for Emerging Infections and Zoonoses (RIZ), University of Veterinary Medicine Foundation, Hanover, Germany
| | - Tamara Tuchel
- Department of Virology, University of Veterinary Medicine Foundation, Hanover, Germany
- Research Center for Emerging Infections and Zoonoses (RIZ), University of Veterinary Medicine Foundation, Hanover, Germany
| | - Christian Meyer Zu Natrup
- Department of Virology, University of Veterinary Medicine Foundation, Hanover, Germany
- Research Center for Emerging Infections and Zoonoses (RIZ), University of Veterinary Medicine Foundation, Hanover, Germany
| | - Lisa-Marie Schünemann
- Department of Virology, University of Veterinary Medicine Foundation, Hanover, Germany
- Research Center for Emerging Infections and Zoonoses (RIZ), University of Veterinary Medicine Foundation, Hanover, Germany
| | - Sabrina Clever
- Department of Virology, University of Veterinary Medicine Foundation, Hanover, Germany
- Research Center for Emerging Infections and Zoonoses (RIZ), University of Veterinary Medicine Foundation, Hanover, Germany
| | - Timo Henneck
- Research Center for Emerging Infections and Zoonoses (RIZ), University of Veterinary Medicine Foundation, Hanover, Germany
- Department of Biochemistry, University of Veterinary Medicine Foundation, Hanover, Germany
| | - Maren von Köckritz-Blickwede
- Research Center for Emerging Infections and Zoonoses (RIZ), University of Veterinary Medicine Foundation, Hanover, Germany
- Department of Biochemistry, University of Veterinary Medicine Foundation, Hanover, Germany
| | - Dirk Schaudien
- Fraunhofer Institute for Toxicology and Experimental Medicine (ITEM), Hanover, Germany
| | - Karl Rohn
- Department of Biometry, Epidemiology and Data Management, University of Veterinary Medicine Foundation, Hanover, Germany
| | - Klaus Schughart
- Department of Microbiology, Immunology and Biochemistry, University of Tennessee Health Science Center, Memphis, TN, USA
- Institute of Virology Münster, University of Münster, Münster, Germany
| | - Robert Geffers
- Helmholtz Centre for Infection Research (HZI), Brunswick, Germany
| | - Mika K Kaneko
- Department of antibody drug development, Tohoku University, Sendai, Miyagi, Japan
| | - Yukinari Kato
- Department of antibody drug development, Tohoku University, Sendai, Miyagi, Japan
| | - Carina Gross
- Department of Cardiology and Angiology, Hanover Medical School (MHH), Hanover, Germany
| | - Georgios Amanakis
- Department of Cardiology and Angiology, Hanover Medical School (MHH), Hanover, Germany
| | - Andreas Pavlou
- Institute for Experimental Infection Research, TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Helmholtz Centre for Infection Research and the Hannover Medical School, Hannover, Germany
| | - Wolfgang Baumgärtner
- Department of Pathology, University of Veterinary Medicine Foundation, Hanover, Germany.
| | - Federico Armando
- Pathology Unit, Department of Veterinary Science, University of Parma, Parma, Italy
| |
Collapse
|
2
|
Menson KE, Coleman SRM. Reprint of: Smoking and pulmonary health in women: A narrative review and behavioral health perspective. Prev Med 2024; 188:108113. [PMID: 39198081 PMCID: PMC11563853 DOI: 10.1016/j.ypmed.2024.108113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/01/2024]
Abstract
OBJECTIVE Cigarette smoking prevalence has declined slower among women than men, and smoking-related pulmonary disease (PD) has risen among women. Given these trends, there is a critical need to understand and mitigate PD risk among women who smoke. The purpose of this narrative review and commentary is to highlight important evidence from the literature on smoking and PD among women. METHODS This review focuses broadly on examining cigarette smoking and PD among women within six topic areas: (1) demographic characteristics and prevalence of smoking, (2) smoking behavior, (3) lung cancer, (4) obstructive PD, (5) diagnostic and treatment disparities, and (6) gaps in the literature and potential directions for future research and treatment. RESULTS Growing evidence indicates that compared to men, women are at increased risk for developing smoking-related PD and poorer PD outcomes. Gender disparities in smoking-related PD may be largely accounted for by genetic differences and sex hormones contributing to PD pathogenesis and presentation, smoking behavior, nicotine dependence, and pathogen/carcinogen clearance. Moreover, gender disparities in smoking-related PD may be exacerbated by important social determinants (e.g., women with less formal education and those from minoritized groups may be at especially high risk for poor PD outcomes due to higher rates of smoking). CONCLUSION Rising rates of smoking-related PD among women risk widening diagnostic and treatment disparities. Ongoing research is needed to explore potentially complex relationships between sex, gender, and smoking-related PD processes and outcomes, and to improve smoking-cessation and PD treatment for women.
Collapse
Affiliation(s)
- Katherine E Menson
- Division of Pulmonary and Critical Care Medicine, University of Vermont, Burlington, VT, USA; Larner College of Medicine, University of Vermont, Burlington, VT, USA.
| | - Sulamunn R M Coleman
- Department of Psychiatry, University of Vermont, Burlington, VT, USA; Vermont Center on Behavior and Health, University of Vermont, Burlington, VT, USA
| |
Collapse
|
3
|
Menson KE, Coleman SRM. Smoking and pulmonary health in women: A narrative review and behavioral health perspective. Prev Med 2024; 185:108029. [PMID: 38851402 DOI: 10.1016/j.ypmed.2024.108029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Revised: 05/24/2024] [Accepted: 06/04/2024] [Indexed: 06/10/2024]
Abstract
OBJECTIVE Cigarette smoking prevalence has declined slower among women than men, and smoking-related pulmonary disease (PD) has risen among women. Given these trends, there is a critical need to understand and mitigate PD risk among women who smoke. The purpose of this narrative review and commentary is to highlight important evidence from the literature on smoking and PD among women. METHODS This review focuses broadly on examining cigarette smoking and PD among women within six topic areas: (1) demographic characteristics and prevalence of smoking, (2) smoking behavior, (3) lung cancer, (4) obstructive PD, (5) diagnostic and treatment disparities, and (6) gaps in the literature and potential directions for future research and treatment. RESULTS Growing evidence indicates that compared to men, women are at increased risk for developing smoking-related PD and poorer PD outcomes. Gender disparities in smoking-related PD may be largely accounted for by genetic differences and sex hormones contributing to PD pathogenesis and presentation, smoking behavior, nicotine dependence, and pathogen/carcinogen clearance. Moreover, gender disparities in smoking-related PD may be exacerbated by important social determinants (e.g., women with less formal education and those from minoritized groups may be at especially high risk for poor PD outcomes due to higher rates of smoking). CONCLUSION Rising rates of smoking-related PD among women risk widening diagnostic and treatment disparities. Ongoing research is needed to explore potentially complex relationships between sex, gender, and smoking-related PD processes and outcomes, and to improve smoking-cessation and PD treatment for women.
Collapse
Affiliation(s)
- Katherine E Menson
- Division of Pulmonary and Critical Care Medicine, University of Vermont, Burlington, VT, USA; Larner College of Medicine, University of Vermont, Burlington, VT, USA.
| | - Sulamunn R M Coleman
- Department of Psychiatry, University of Vermont, Burlington, VT, USA; Vermont Center on Behavior and Health, University of Vermont, Burlington, VT, USA
| |
Collapse
|
4
|
Yoo JJ, Park MY, Cho EJ, Yu SJ, Kim SG, Kim YJ, Kim YS, Yoon JH. Smoking Increases the Risk of Hepatocellular Carcinoma and Cardiovascular Disease in Patients with Metabolic-Associated Fatty Liver Disease. J Clin Med 2023; 12:3336. [PMID: 37176776 PMCID: PMC10179445 DOI: 10.3390/jcm12093336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 05/04/2023] [Accepted: 05/06/2023] [Indexed: 05/15/2023] Open
Abstract
The association of smoking with hepatocellular carcinoma (HCC) or cardiovascular disease (CVD) has been reported, but the study of its relationship with metabolic-associated fatty liver disease (MAFLD) is limited. We aimed to investigate the effect of smoking on the incidence of HCC or CVD in MAFLD patients. Using the Korean nationwide health screening database, we analyzed subjects between 2001 and 2015. A total of 283,088 subjects including 110,863 MAFLD patients and 172,225 controls were analyzed. Smoking status was divided by non-smoker, ex-smoker, or current smoker. In the follow-up period, a total of 2903 (1.0%) subjects developed HCC, and the MAFLD group (1723, 1.6%) had a significantly higher incidence than the control group (1180, 0.7%). In the MAFLD group, current smokers showed significantly higher risk of HCC compared to non-smokers (adjusted HR 1.24, 95% CI 1.08-1.41), whereas the control group did not (adjusted HR 1.07, 95% CI 0.89-1.30). A total of 18,984 (6.7%) patients developed CVD, and the incidence was significantly higher in the MAFLD group (8688, 7.8%) than in the control group (10,296, 6.0%), similar to HCC. The risk of CVD in current smokers increased by 22% compared to non-smokers in the MAFLD group (adjusted HR 1.22, 95% CI 1.15-1.30) and by 21% (adjusted HR 1.21, 95% CI 1.13-1.29) in the control group. Based on sex stratification, men showed increased incidence of both HCC and CVD by smoking, whereas women had only increased risk of CVD. Smoking significantly increases the incidence of HCC and CVD in MAFLD patients; thus, it is highly recommended to quit smoking completely in the population with MAFLD.
Collapse
Affiliation(s)
- Jeong-Ju Yoo
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Soonchunhyang University Bucheon Hospital, Gyeonggi-do 14584, Republic of Korea; (J.-J.Y.); (S.G.K.); (Y.S.K.)
| | - Man Young Park
- Korea Institute of Oriental Medicine, Daejeon 34054, Republic of Korea;
| | - Eun Ju Cho
- Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, Seoul 03080, Republic of Korea; (S.J.Y.); (Y.J.K.); (J.-H.Y.)
| | - Su Jong Yu
- Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, Seoul 03080, Republic of Korea; (S.J.Y.); (Y.J.K.); (J.-H.Y.)
| | - Sang Gyune Kim
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Soonchunhyang University Bucheon Hospital, Gyeonggi-do 14584, Republic of Korea; (J.-J.Y.); (S.G.K.); (Y.S.K.)
| | - Yoon Jun Kim
- Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, Seoul 03080, Republic of Korea; (S.J.Y.); (Y.J.K.); (J.-H.Y.)
| | - Young Seok Kim
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Soonchunhyang University Bucheon Hospital, Gyeonggi-do 14584, Republic of Korea; (J.-J.Y.); (S.G.K.); (Y.S.K.)
| | - Jung-Hwan Yoon
- Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, Seoul 03080, Republic of Korea; (S.J.Y.); (Y.J.K.); (J.-H.Y.)
| |
Collapse
|
5
|
Smith R, Yendamuri S, Vedire Y, Rosario S, Zollo R, Washington D, Sass S, Ivanick NM, Reid M, Barbi J. Immunoprofiling bronchoalveolar lavage cells reveals multifaceted smoking-associated immune dysfunction. ERJ Open Res 2023; 9:00688-2022. [PMID: 37342091 PMCID: PMC10277872 DOI: 10.1183/23120541.00688-2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Accepted: 03/22/2023] [Indexed: 06/22/2023] Open
Abstract
Background Bronchoalveolar lavage (BAL) is an underutilised tool in the search for pulmonary disease biomarkers. While leukocytes with effector and suppressor function play important roles in airway immunity and tumours, it remains unclear if frequencies and phenotypes of BAL leukocytes can be useful parameters in lung cancer studies and clinical trials. We therefore explored the utility of BAL leukocytes as a source of biomarkers interrogating the impact of smoking, a major lung cancer risk determinant, on pulmonary immunity. Methods In this "test case" observational study, BAL samples from 119 donors undergoing lung cancer screening and biopsy procedures were evaluated by conventional and spectral flow cytometry to exemplify the comprehensive immune analyses possible with this biospecimen. Proportions of major leukocyte populations and phenotypic markers levels were found. Multivariate linear rank sum analysis considering age, sex, cancer diagnosis and smoking status was performed. Results Significantly increased frequencies of myeloid-derived suppressor cells and PD-L1-expressing macrophages were found in current and former smokers compared to never-smokers. While cytotoxic CD8 T-cells and conventional CD4 helper T-cell frequencies were significantly reduced in current and former smokers, expression of immune checkpoints PD-1 and LAG-3 as well as Tregs proportions were increased. Lastly, the cellularity, viability and stability of several immune readouts under cryostorage suggested BAL samples are useful for correlative end-points in clinical trials. Conclusions Smoking is associated with heightened markers of immune dysfunction, readily assayable in BAL, that may reflect a permissive environment for cancer development and progression in the airway.
Collapse
Affiliation(s)
- Randall Smith
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
- These authors contributed equally
| | - Sai Yendamuri
- Department of Thoracic Surgery, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
- These authors contributed equally
- These authors contributed equally to this article as lead authors and supervised the work
| | - Yeshwanth Vedire
- Department of Thoracic Surgery, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Spencer Rosario
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Robert Zollo
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Deschana Washington
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Stephanie Sass
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Nathaniel M. Ivanick
- Department of Thoracic Surgery, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Mary Reid
- Department of Medicine – Survivorship and Supportive Care, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Joseph Barbi
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
- Department of Thoracic Surgery, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
- These authors contributed equally to this article as lead authors and supervised the work
| |
Collapse
|
6
|
Chen Z, Zhu J, Zhou H, Jia Y, Ruan H, Diao Q, Li M, Zheng L, Yao S, Guo Y, Zhou Y, Jiang Y. The involvement of copper, circular RNAs, and inflammatory cytokines in chronic respiratory disease. CHEMOSPHERE 2022; 303:135005. [PMID: 35605724 DOI: 10.1016/j.chemosphere.2022.135005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Revised: 04/23/2022] [Accepted: 05/15/2022] [Indexed: 06/15/2023]
Abstract
Exposure to high concentrations of copper is associated with pulmonary inflammation and chronic respiratory disease (CRD). Epigenetic modulation of noncoding RNAs contributes to the development of several CRDs. It is unknown whether epigenetic modulation is involved in copper mediated pulmonary inflammation and CRD. We conducted a case-control study of 101 CRD cases and 161 control subjects in Shijiazhuang, China, and evaluated circRNAs and cytokine levels (IL-6 and IL-8) by qPCR and ELISA. Urinary copper concentration was determined by inductively coupled plasma mass spectrometry. Linear mixed models and generalized linear mixed models were used to assess the associations of circRNAs with CRD, urinary copper, and cytokines. We exposed the human bronchial epithelial cell line, 16HBE, to copper and assessed the functional role of a circRNA, circ_0008882, by RNA overexpression. Cellular location of circ_0008882 was assessed by separation of nuclear and cytoplasmic RNAs. Nine circRNAs were associated with an increased risk for CRDs, while the relative expression of circ_0008882 was decreased after copper exposure in vitro and in vivo. Copper exposure stimulated 16HBE cells to release proinflammatory IL-6 and IL-8. The release of the cytokines was inhibited by overexpression of circ_0008882. These results suggest a role for circ_0008882 in the regulation of CRD associated inflammation following copper exposure.
Collapse
Affiliation(s)
- Zehao Chen
- State Key Laboratory of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China; Institute for Chemical Carcinogenesis, Guangzhou Medical University, Guangzhou, 511436, China
| | - Jialu Zhu
- State Key Laboratory of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China; Institute for Chemical Carcinogenesis, Guangzhou Medical University, Guangzhou, 511436, China
| | - Hanyu Zhou
- Institute for Chemical Carcinogenesis, Guangzhou Medical University, Guangzhou, 511436, China
| | - Yangyang Jia
- Institute for Chemical Carcinogenesis, Guangzhou Medical University, Guangzhou, 511436, China
| | - Honglian Ruan
- Institute for Chemical Carcinogenesis, Guangzhou Medical University, Guangzhou, 511436, China
| | - Qinqin Diao
- Institute for Chemical Carcinogenesis, Guangzhou Medical University, Guangzhou, 511436, China
| | - Meizhen Li
- Institute for Chemical Carcinogenesis, Guangzhou Medical University, Guangzhou, 511436, China
| | - Liting Zheng
- Institute for Chemical Carcinogenesis, Guangzhou Medical University, Guangzhou, 511436, China
| | - Shuwei Yao
- Institute for Chemical Carcinogenesis, Guangzhou Medical University, Guangzhou, 511436, China
| | - Yaozheng Guo
- Institute for Chemical Carcinogenesis, Guangzhou Medical University, Guangzhou, 511436, China
| | - Yun Zhou
- State Key Laboratory of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China; Institute for Chemical Carcinogenesis, Guangzhou Medical University, Guangzhou, 511436, China.
| | - Yiguo Jiang
- State Key Laboratory of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China; Institute for Chemical Carcinogenesis, Guangzhou Medical University, Guangzhou, 511436, China.
| |
Collapse
|
7
|
He Y, Patel CJ. Shared exposure liability of type 2 diabetes and other chronic conditions in the UK Biobank. Acta Diabetol 2022; 59:851-860. [PMID: 35348899 PMCID: PMC9085680 DOI: 10.1007/s00592-022-01864-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 01/31/2022] [Indexed: 11/09/2022]
Abstract
AIMS To investigate whether the cumulative exposure risks of incident T2D are shared with other common chronic diseases. RESEARCH DESIGN AND METHODS We first establish and report the cross-sectional prevalence, cross-sectional co-prevalence, and incidence of seven T2D-associated chronic diseases [hypertension, atrial fibrillation, coronary artery disease, obesity, chronic obstructive pulmonary disease (COPD), and chronic kidney and liver diseases] in the UK Biobank. We use published weights of genetic variants and exposure variables to derive the T2D polygenic (PGS) and polyexposure (PXS) risk scores and test their associations to incident diseases. RESULTS PXS was associated with higher levels of clinical risk factors including BMI, systolic blood pressure, blood glucose, triglycerides, and HbA1c in individuals without overt or diagnosed T2D. In addition to predicting incident T2D, PXS and PGS were significantly and positively associated with the incidence of all 7 other chronic diseases. There were 4% and 8% of individuals in the bottom deciles of PXS and PGS, respectively, who were prediabetic at baseline but had low risks of T2D and other chronic diseases. Compared to the remaining population, individuals in the top deciles of PGS and PXS had particularly high risks of developing chronic diseases. For instance, the hazard ratio of COPD and obesity for individuals in the top T2D PXS deciles was 2.82 (95% CI 2.39-3.35, P = 4.00 × 10-33) and 2.54 (95% CI 2.24-2.87, P = 9.86 × 10-50), respectively, compared to the remaining population. We also found that PXS and PGS were both significantly (P < 0.0001) and positively associated with the total number of incident diseases. CONCLUSIONS T2D shares polyexposure risks with other common chronic diseases. Individuals with an elevated genetic and non-genetic risk of T2D also have high risks of cardiovascular, liver, lung, and kidney diseases.
Collapse
Affiliation(s)
- Yixuan He
- Program in Bioinformatics and Integrative Genomics, Harvard Medical School, 10 Shattuck St, Boston, MA, 02215, USA
- Department of Biomedical Informatics, Harvard Medical School, 10 Shattuck St, Boston, MA, USA
| | - Chirag J Patel
- Department of Biomedical Informatics, Harvard Medical School, 10 Shattuck St, Boston, MA, USA.
| |
Collapse
|
8
|
Wang Q, Unwalla H, Rahman I. Dysregulation of mitochondrial complexes and dynamics by chronic cigarette smoke exposure Utilizing MitoQC reporter mice. Mitochondrion 2022; 63:43-50. [PMID: 35032706 PMCID: PMC8885972 DOI: 10.1016/j.mito.2022.01.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 01/06/2022] [Accepted: 01/10/2022] [Indexed: 02/04/2023]
Abstract
Cigarette smoke (CS) is known to cause impaired mitophagy and mitochondrial dysregulation in the pathogenesis of chronic obstructive pulmonary disease (COPD)/emphysema. Mitochondrial complexes and dynamics are affected by acute CS exposure in lung epithelium and mouse lung. We hypothesize that chronic CS exposure (4 months) will induce lung mitochondrial dysregulation and abnormal mitophagy. In this study, we employed the mitoQC reporter mice, a mitochondrial reporter strain, which can reflect the mitophagy based on the fluorescence-tagged mitochondria. Chronic CS exposure induced lung inflammatory cell infiltration, airspace enlargement, and lung cellular senescence. We showed the higher occurrence of mitophagy (GFP/mCherry) in the lung cells by CS exposure, associated with more mitochondrial fluorescence signals (GFP+/mCherry+). After chronic CS exposure, the mitochondrial complexes and function related genes were inhibited, while protein levels of complexes I and III slightly changed. Additionally, chronic CS exposure down-regulated most of the mitochondrial dynamic markers at gene expression level, included mitochondrial fusion/fission and mitochondrial translocate/transfer markers. For the markers related to mitophagy, Pink1 and Parkin, decreased gene and protein levels of Parkin, and decreased gene expression of Pink1, were identified in the CS exposure group. Hence, CS-induced mitophagy is mediated by Pink1-Parkin independent mechanism. Thus, we have shown that the chronic CS epxosure dysregulated mitochondrial complexes and dynamics and induced mitophagy, using the state-of-the art mitoQC reporter mouse model. Our results suggested that dysregulated mitochondrial function and dynamics are associated with CS-induced lung injury and phenotypic development of chronic lung diseases, such as COPD/ emphysema.
Collapse
Affiliation(s)
- Qixin Wang
- Department of Environmental Medicine, School of Medicine and Dentistry, University of Rochester Medical Center, Rochester, NY, USA
| | - Hoshang Unwalla
- Department of Immunology and Nanomedicine, Herbert Wertheim College of medicine, Florida International University, Miami, FL, USA
| | - Irfan Rahman
- Department of Environmental Medicine, School of Medicine and Dentistry, University of Rochester Medical Center, Rochester, NY, USA.
| |
Collapse
|
9
|
Ishak SR, Hassan AM, Kamel TB. Environmental hazards and demographic and clinical data of childhood interstitial lung diseases in a tertiary institute in Egypt. THE EGYPTIAN JOURNAL OF BRONCHOLOGY 2021. [DOI: 10.1186/s43168-020-00048-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
The incidence of childhood interstitial lung diseases increased in the last years in Egypt. Changes in environmental and climatic conditions may be contributing factors. Also, raising birds at home increased in the past years due to financial issues. Other environmental factors include increased industries, traffic, and pollution. Our study aimed to assess the environmental hazards and the severity of childhood interstitial lung diseases.
Results
Sixty-five percent of patients with childhood interstitial lung diseases (chILD) were exposed to cigarette smoke; 45% were exposed to birds, 30% to industrial wastes, 20% to grass and pesticides, and 10% to animals.
Conclusions
Exposure to cigarette smoke and birds increases the risk of development of chILD.
Collapse
|
10
|
Zhang Z, Cao Z, Deng F, Yang Z, Ma S, Guan Q, Liu R, He Z. Infrared Thermal Imaging of Patients With Acute Upper Respiratory Tract Infection: Mixed Methods Analysis. Interact J Med Res 2021; 10:e22524. [PMID: 34420912 PMCID: PMC8414296 DOI: 10.2196/22524] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 08/10/2020] [Accepted: 08/27/2020] [Indexed: 01/30/2023] Open
Abstract
Background Upper respiratory tract infection is a common disease of the respiratory system. Its incidence is very high, and it can even cause pandemics. Infrared thermal imaging (IRTI) can provide an objective and quantifiable reference for the visual diagnosis of people with acute respiratory tract infection, and it can function as an effective indicator of clinical diagnosis. Objective The aims of this study are to observe and analyze the infrared expression location and characteristics of patients with acute upper respiratory tract infection through IRTI technology and to clearly express the quantification of temperature, analyze the role of IRTI in acute upper respiratory tract diagnostic research, and understand the impact of IRTI in qualitative and quantitative research. Methods From December 2018 to February 2019, 154 patients with acute upper respiratory tract infection were randomly selected from the emergency department of the First Affiliated Hospital of Guangzhou Medical University. Among these patients, 73 were men and 81 were women. The subjects were divided into two groups according to the presence of fever, namely, fever and nonfever groups. Qualitative and quantitative analyses of the infrared thermal images were performed to compare the results before and after application of the technology. Results Using the method described in this paper, through the analysis of experimental data, we elucidated the role of IRTI in the diagnosis of acute upper respiratory tract infection, and we found that qualitative and quantitative IRTI analyses play important roles. Through the combination of theory and experimental data, the IRTI analysis showed good results in identifying acute upper respiratory tract infection. Conclusions IRTI technology plays an important role in identifying the infrared expression location and characteristics of patients with acute upper respiratory tract infection as well as in the quantification of clear expression of body temperature, and it provides an objective and quantifiable reference basis for elucidating the pathogenesis of these patients.
Collapse
Affiliation(s)
- Zuopeng Zhang
- Emergency Department, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - ZanFeng Cao
- Emergency Department, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Fangge Deng
- State Key Laboratory of Respiratory Diseases, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Zhanzheng Yang
- Emergency Department, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Sige Ma
- The First Clinical College, Guangzhou Medical University, Guangzhou, China
| | - Qianting Guan
- The First Clinical College, Guangzhou Medical University, Guangzhou, China
| | - Rong Liu
- Emergency Department, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Zhuosen He
- Emergency Department, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
11
|
Lin L, Xuan W, Luckey D, Wang S, Wang F, Lau J, Warrington KJ, Matteson EL, Vassallo R, Taneja V. A novel humanized model of rheumatoid arthritis associated lung disease. Clin Immunol 2021; 230:108813. [PMID: 34333094 DOI: 10.1016/j.clim.2021.108813] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 07/14/2021] [Accepted: 07/26/2021] [Indexed: 02/08/2023]
Abstract
Cigarette smoking has been implicated in the pathogenesis of seropositive rheumatoid arthritis (RA), as well as RA-associated lung disease. Fibrotic interstitial lung disease as well as emphysema occur in RA and cause substantial morbidity. We used arthritis-susceptible HLA-DQ8 transgenic mice to generate RA-associated lung disease. Mice were exposed to cigarette smoke (CS) prior to induction of arthritis, and subsequently injected with a low dose of bleomycin intra-tracheally to induce lung injury. Exposure of arthritic mice to both CS and bleomycin led to a significant reduction in lung compliance consistent with development of diffuse lung disease. Morphologic evaluation of the lung demonstrated areas of emphysematous change and co-existent fibrosis, consistent with a combined pattern of fibrosis and emphysema. These changes were accompanied by inflammatory cell infiltration and upregulation of fibrosis-associated genes. This humanized mouse model can serve as a valuable research tool to understand the pathogenesis of RA associated lung disease.
Collapse
Affiliation(s)
- Li Lin
- Division of Pulmonary and Critical Care Medicine, Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, United States of America
| | - Weixia Xuan
- Division of Pulmonary and Critical Care Medicine, Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, United States of America; Department of Resporatory medicine, Henan Provincial People's Hospital, Zhengzhou, China
| | - David Luckey
- Department of Immunology, Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, United States of America
| | - Shaohua Wang
- Division of Pulmonary and Critical Care Medicine, Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, United States of America
| | - FaPing Wang
- Division of Pulmonary and Critical Care Medicine, Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, United States of America; Department of Respiratory and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Jessica Lau
- Division of Pulmonary and Critical Care Medicine, Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, United States of America; Pulmonary and Critical Care Medicine, The Vancouver Clinic, Vancouver, WA, United States of America
| | - Kenneth J Warrington
- Division of Rheumatology, Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, United States of America
| | - Eric L Matteson
- Division of Rheumatology, Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, United States of America
| | - Robert Vassallo
- Division of Pulmonary and Critical Care Medicine, Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, United States of America; Department of Medicine, Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, United States of America.
| | - Veena Taneja
- Department of Immunology, Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, United States of America; Division of Rheumatology, Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, United States of America.
| |
Collapse
|
12
|
Jung CC, Chen NT, Hsia YF, Hsu NY, Su HJ. Influence of Indoor Temperature Exposure on Emergency Department Visits Due to Infectious and Non-Infectious Respiratory Diseases for Older People. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:ijerph18105273. [PMID: 34063510 PMCID: PMC8156969 DOI: 10.3390/ijerph18105273] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 05/12/2021] [Accepted: 05/13/2021] [Indexed: 12/24/2022]
Abstract
Previous studies have demonstrated that outdoor temperature exposure was an important risk factor for respiratory diseases. However, no study investigates the effect of indoor temperature exposure on respiratory diseases and further assesses cumulative effect. The objective of this study is to study the cumulative effect of indoor temperature exposure on emergency department visits due to infectious (IRD) and non-infectious (NIRD) respiratory diseases among older adults. Subjects were collected from the Longitudinal Health Insurance Database in Taiwan. The cumulative degree hours (CDHs) was used to assess the cumulative effect of indoor temperature exposure. A distributed lag nonlinear model with quasi-Poisson function was used to analyze the association between CDHs and emergency department visits due to IRD and NIRD. For IRD, there was a significant risk at 27, 28, 29, 30, and 31 °C when the CDHs exceeded 69, 40, 14, 5, and 1 during the cooling season (May to October), respectively, and at 19, 20, 21, 22, and 23 °C when the CDHs exceeded 8, 1, 1, 35, and 62 during the heating season (November to April), respectively. For NIRD, there was a significant risk at 19, 20, 21, 22, and 23 °C when the CDHs exceeded 1, 1, 16, 36, and 52 during the heating season, respectively; the CDHs at 1 was only associated with the NIRD at 31 °C during the cooling season. Our data also indicated that the CDHs was lower among men than women. We conclude that the cumulative effects of indoor temperature exposure should be considered to reduce IRD risk in both cooling and heating seasons and NIRD risk in heating season and the cumulative effect on different gender.
Collapse
Affiliation(s)
- Chien-Cheng Jung
- Department of Public Health, China Medical University, Taichung City 406060, Taiwan;
| | - Nai-Tzu Chen
- Research Center of Environmental Trace Toxic Substances, National Cheng Kung University, Tainan City 70403, Taiwan;
| | - Ying-Fang Hsia
- Big Data Center, China Medical University Hospital, Taichung City 404332, Taiwan;
| | - Nai-Yun Hsu
- Department of Environmental and Occupational Health, College of Medicine, National Cheng Kung University, Tainan City 70403, Taiwan;
| | - Huey-Jen Su
- Department of Environmental and Occupational Health, College of Medicine, National Cheng Kung University, Tainan City 70403, Taiwan;
- Correspondence: ; Tel.: +886-6-275-2459; Fax: +886-6-274-3748
| |
Collapse
|
13
|
Charatcharoenwitthaya P, Karaketklang K, Aekplakorn W. Cigarette Smoking Increased Risk of Overall Mortality in Patients With Non-alcoholic Fatty Liver Disease: A Nationwide Population-Based Cohort Study. Front Med (Lausanne) 2020; 7:604919. [PMID: 33365321 PMCID: PMC7750535 DOI: 10.3389/fmed.2020.604919] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Accepted: 11/18/2020] [Indexed: 12/13/2022] Open
Abstract
Background: The evidence suggests a detrimental effect of cigarette smoking on the progression of chronic liver disease. However, the impact of cigarette smoking on mortality among patients with non-alcoholic fatty liver disease (NAFLD) remain unclear. Methods: We used the National Health Examination Survey data collected during 2008-2009 to link the National Death Index to follow-up respondent survival. Diagnosis of NAFLD was based on a lipid accumulation product in participants without significant alcohol use or other liver diseases. Results: During 64,116 person-years of follow-up, 928 of 7,529 participants with NAFLD died, and the cumulative all-cause mortality was 14.5 per 1,000 person-years. In a Cox regression model adjusted for age, body mass index, alcohol intake, exercise, comorbidities, lipid profiles, and handgrip strength, current smoking increased the risk of mortality by 109% (adjusted hazard ratio (aHR): 2.09, 95% confidence interval [CI]: 1.18-3.71) compared with never smoker status in women, but showed only a trend toward harm among men (aHR: 1.41, 95% CI: 0.96-2.08). After controlling for potential confounders, smoking ≥10 pack-years continued to show a significant harmful effect on all-cause mortality among women (aHR: 5.40, 95% CI: 2.19-13.4), but not in men. Among women who drink alcohol ≥10 grams per day, current smoking (aHR: 13.8, 95% CI: 1.66-145) and smoking ≥10 pack-years (aHR: 310, 95% CI: 78-1,296) also significantly increased risk of death. Conclusion: This nationwide population-based study highlight a detrimental effect of cigarette smoking on mortality, with a similar but more definite association in women than in men with NAFLD.
Collapse
Affiliation(s)
- Phunchai Charatcharoenwitthaya
- Division of Gastroenterology, Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Khemajira Karaketklang
- Division of Gastroenterology, Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Wichai Aekplakorn
- Department of Community Medicine, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| |
Collapse
|
14
|
Andres J, Smith LC, Murray A, Jin Y, Businaro R, Laskin JD, Laskin DL. Role of extracellular vesicles in cell-cell communication and inflammation following exposure to pulmonary toxicants. Cytokine Growth Factor Rev 2020; 51:12-18. [PMID: 31901309 PMCID: PMC7052797 DOI: 10.1016/j.cytogfr.2019.12.001] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Accepted: 12/09/2019] [Indexed: 12/22/2022]
Abstract
Extracellular vesicles (EVs) have emerged as key regulators of cell-cell communication during inflammatory responses to lung injury induced by diverse pulmonary toxicants including cigarette smoke, air pollutants, hyperoxia, acids, and endotoxin. Many lung cell types, including epithelial cells and endothelial cells, as well as infiltrating macrophages generate EVs. EVs appear to function by transporting cargo to recipient cells that, in most instances, promote their inflammatory activity. Biologically active cargo transported by EVs include miRNAs, cytokines/chemokines, damage-associated molecular patterns (DAMPs), tissue factor (TF)s, and caspases. Findings that EVs are taken up by target cells such as macrophages, and that this leads to increased proinflammatory functioning provide support for their role in the development of pathologies associated with toxicant exposure. Understanding the nature of EVs responding to toxic exposures and their cargo may lead to the development of novel therapeutic approaches to mitigating lung injury.
Collapse
Affiliation(s)
- Jaclynn Andres
- Department of Pharmacology and Toxicology, Rutgers University Ernest Mario School of Pharmacy, Piscataway, NJ 08854 USA
| | - Ley Cody Smith
- Department of Pharmacology and Toxicology, Rutgers University Ernest Mario School of Pharmacy, Piscataway, NJ 08854 USA
| | - Alexa Murray
- Department of Pharmacology and Toxicology, Rutgers University Ernest Mario School of Pharmacy, Piscataway, NJ 08854 USA
| | - Yang Jin
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Boston University, Boston, MA 02118 USA
| | - Rita Businaro
- Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University of Rome, 04100 Latina, Italy
| | - Jeffrey D Laskin
- Department of Environmental and Occupational Health, Rutgers University School of Public Health, Piscataway, NJ 08854 USA
| | - Debra L Laskin
- Department of Pharmacology and Toxicology, Rutgers University Ernest Mario School of Pharmacy, Piscataway, NJ 08854 USA.
| |
Collapse
|
15
|
Zuo H, Faiz A, van den Berge M, Mudiyanselage SNHR, Borghuis T, Timens W, Nikolaev VO, Burgess JK, Schmidt M. Cigarette smoke exposure alters phosphodiesterases in human structural lung cells. Am J Physiol Lung Cell Mol Physiol 2019; 318:L59-L64. [PMID: 31664853 DOI: 10.1152/ajplung.00319.2019] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Cigarette smoke (CS), a highly complex mixture containing more than 4,000 compounds, causes aberrant cell responses leading to tissue damage around the airways and alveoli, which underlies various lung diseases. Phosphodiesterases (PDEs) are a family of enzymes that hydrolyze cyclic nucleotides. PDE inhibition induces bronchodilation, reduces the activation and recruitment of inflammatory cells, and the release of various cytokines. Currently, the selective PDE4 inhibitor roflumilast is an approved add-on treatment for patients with severe chronic obstructive pulmonary disease with chronic bronchitis and a history of frequent exacerbations. Additional selective PDE inhibitors are being tested in preclinical and clinical studies. However, the effect of chronic CS exposure on the expression of PDEs is unknown. Using mRNA isolated from nasal and bronchial brushes and lung tissues of never smokers and current smokers, we compared the gene expression of 25 PDE coding genes. Additionally, the expression and distribution of PDE3A and PDE4D in human lung tissues was examined. This study reveals that chronic CS exposure modulates the expression of various PDE members. Thus, CS exposure may change the levels of intracellular cyclic nucleotides and thereby impact the efficiency of PDE-targeted therapies.
Collapse
Affiliation(s)
- Haoxiao Zuo
- University of Groningen, Department of Molecular Pharmacology, Groningen, The Netherlands.,University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD, Groningen, The Netherlands.,Institute of Experimental Cardiovascular Research, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - Alen Faiz
- University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD, Groningen, The Netherlands.,University of Groningen, Department of Pulmonary Diseases, University Medical Center Groningen, Groningen, The Netherlands.,Emphysema Center, Woolcock Institute of Medical Research, The University of Sydney, Glebe, New South Wales, Australia.,Faculty of Science, University of Technology Sydney, Respiratory Bioinformatics and Molecular Biology, Ultimo, New South Wales, Australia
| | - Maarten van den Berge
- University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD, Groningen, The Netherlands.,University of Groningen, Department of Pulmonary Diseases, University Medical Center Groningen, Groningen, The Netherlands
| | | | - Theo Borghuis
- University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD, Groningen, The Netherlands.,University of Groningen, University Medical Center Groningen, Department of Pathology and Medical Biology, Groningen, The Netherlands
| | - Wim Timens
- University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD, Groningen, The Netherlands.,University of Groningen, University Medical Center Groningen, Department of Pathology and Medical Biology, Groningen, The Netherlands
| | - Viacheslav O Nikolaev
- Institute of Experimental Cardiovascular Research, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany.,German Center for Cardiovascular Research, Hamburg, Germany
| | - Janette K Burgess
- University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD, Groningen, The Netherlands.,University of Groningen, University Medical Center Groningen, Department of Pathology and Medical Biology, Groningen, The Netherlands
| | - Martina Schmidt
- University of Groningen, Department of Molecular Pharmacology, Groningen, The Netherlands.,University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD, Groningen, The Netherlands
| |
Collapse
|
16
|
Li AL, Shen T, Wang T, Zhou MX, Wang B, Song JT, Zhang PL, Wang XL, Ren DM, Lou HX, Wang XN. Novel diterpenoid-type activators of the Keap1/Nrf2/ARE signaling pathway and their regulation of redox homeostasis. Free Radic Biol Med 2019; 141:21-33. [PMID: 31167117 DOI: 10.1016/j.freeradbiomed.2019.06.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2019] [Revised: 05/15/2019] [Accepted: 06/01/2019] [Indexed: 12/25/2022]
Abstract
Oxidative stress is involved in the onset and progression of many human diseases. Activators of the Keap1/Nrf2/ARE pathway effectively inhibit the progression of oxidative stress-induced diseases. Herein, a small library of diterpenoids was established by means of phytochemical isolation, and chemical modification on naturally occurring molecules. The diterpenoids were subjected to a NAD(P)H: quinone reductase (QR) assay to evaluate its potential inhibition against oxidative stress. Sixteen diterpenoids were found to be novel potential activators of Nrf2-mediated defensive response. Of which, an isopimarane-type diterpenoid, sphaeropsidin A (SA), was identified as a potent activator of the Keap1/Nrf2/ARE pathway, and displayed approximately 5-folds potency than that of sulforaphane (SF). SA activated Nrf2 and its downstream cytoprotective genes through enhancing the stabilization of Nrf2 in a process involving PI3K, PKC, and PERK, as well as potentially interrupting Nrf2-Keap1 protein-protein interaction. In addition, SA conferred protection against sodium arsenite [As(III)]- and cigarette smoke extract (CSE)-induced redox imbalance and cytotoxicity in human lung epithelial cells, as wells as inhibited metronidazole (MTZ)-induced oxidative insult in Tg (krt4: NTR-hKikGR)cy17 transgenic zebrafish and lipopolysaccharide (LPS)-induced oxidative damage in wild-type AB zebrafish. These results imply that SA is a lead compound for therapeutic agent against oxidative stress-induced diseases, and diterpenoid is a good resource for discovering drug candidates and leads of antioxidant therapy.
Collapse
Affiliation(s)
- Ai-Ling Li
- Key Lab of Chemical Biology (MOE), School of Pharmaceutical Sciences, Shandong University, No. 44 Wenhua Xi Road, Jinan, 250012, People's Republic of China
| | - Tao Shen
- Key Lab of Chemical Biology (MOE), School of Pharmaceutical Sciences, Shandong University, No. 44 Wenhua Xi Road, Jinan, 250012, People's Republic of China
| | - Tian Wang
- Key Lab of Chemical Biology (MOE), School of Pharmaceutical Sciences, Shandong University, No. 44 Wenhua Xi Road, Jinan, 250012, People's Republic of China
| | - Ming-Xing Zhou
- Key Lab of Chemical Biology (MOE), School of Pharmaceutical Sciences, Shandong University, No. 44 Wenhua Xi Road, Jinan, 250012, People's Republic of China
| | - Bin Wang
- Key Lab of Chemical Biology (MOE), School of Pharmaceutical Sciences, Shandong University, No. 44 Wenhua Xi Road, Jinan, 250012, People's Republic of China
| | - Jin-Tong Song
- Key Lab of Chemical Biology (MOE), School of Pharmaceutical Sciences, Shandong University, No. 44 Wenhua Xi Road, Jinan, 250012, People's Republic of China
| | - Peng-Liang Zhang
- Key Lab of Chemical Biology (MOE), School of Pharmaceutical Sciences, Shandong University, No. 44 Wenhua Xi Road, Jinan, 250012, People's Republic of China
| | - Xiao-Ling Wang
- The Second Hospital of Shandong University, No. 247 Bei-Yuan Street, Jinan, 250033, People's Republic of China
| | - Dong-Mei Ren
- Key Lab of Chemical Biology (MOE), School of Pharmaceutical Sciences, Shandong University, No. 44 Wenhua Xi Road, Jinan, 250012, People's Republic of China
| | - Hong-Xiang Lou
- Key Lab of Chemical Biology (MOE), School of Pharmaceutical Sciences, Shandong University, No. 44 Wenhua Xi Road, Jinan, 250012, People's Republic of China
| | - Xiao-Ning Wang
- Key Lab of Chemical Biology (MOE), School of Pharmaceutical Sciences, Shandong University, No. 44 Wenhua Xi Road, Jinan, 250012, People's Republic of China.
| |
Collapse
|
17
|
Huang SJ, Xu YM, Lau ATY. Electronic cigarette: A recent update of its toxic effects on humans. J Cell Physiol 2018; 233:4466-4478. [PMID: 29215738 DOI: 10.1002/jcp.26352] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Accepted: 11/29/2017] [Indexed: 02/05/2023]
Abstract
Electronic cigarettes (e-cigarettes), battery-powered and liquid-vaporizing devices, were invented to replace the conventional cigarette (c-cigarette) smoking for the sake of reducing the adverse effects on multiple organ systems that c-cigarettes have induced. Although some of the identified harmful components in e-cigarettes were alleged to be measured in lower quantity than those in c-cigarettes, researchers unveiled that the toxic effects of e-cigarettes should not be understated. This review is sought for an attempt to throw light on several typical types of e-cigarette components (tobacco-specific nitrosamines, carbonyl compounds, and volatile organic compounds) by revealing their possible impacts on human bodies through different action mechanisms characterized by alteration of specific biomarkers on cellular and molecular levels. In addition, this review is intended to draw the limelight that like c-cigarettes, e-cigarettes could also be accompanied with toxic effects on whole human body, which are especially apparent on respiratory system. From head to foot, from physical aspect to chemical aspect, from genotype to phenotype, potential alterations will take place upon the intake of the liquid aerosol.
Collapse
Affiliation(s)
- Shu-Jie Huang
- Laboratory of Cancer Biology and Epigenetics, Department of Cell Biology and Genetics, Shantou University Medical College, Shantou, Guangdong, People's Republic of China
| | - Yan-Ming Xu
- Laboratory of Cancer Biology and Epigenetics, Department of Cell Biology and Genetics, Shantou University Medical College, Shantou, Guangdong, People's Republic of China
| | - Andy T Y Lau
- Laboratory of Cancer Biology and Epigenetics, Department of Cell Biology and Genetics, Shantou University Medical College, Shantou, Guangdong, People's Republic of China
| |
Collapse
|
18
|
Nagashima H, Fujimura I, Nakamura Y, Utsumi Y, Yamauchi K, Takikawa Y, Yokoyama Y, Sakata K, Kobayashi S, Ogawa A. Changes in pulmonary function of residents in Sanriku Seacoast following the tsunami disaster from the Great East Japan Earthquake. Respir Investig 2018; 56:184-188. [PMID: 29548658 DOI: 10.1016/j.resinv.2017.12.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Revised: 12/07/2017] [Accepted: 12/08/2017] [Indexed: 11/18/2022]
Abstract
BACKGROUND Residents in the district struck by the Great East Japan Earthquake Tsunami (GEJET) suffered from adverse living conditions and various pulmonary diseases. OBJECTIVES To evaluate the influence of GEJET, we performed serial assessment of pulmonary function of approximately 10,000 residents in the district struck by GEJET. METHODS Using a spirometer, we assessed the pulmonary function of approximately 10,000 residents older than 18 years in the Sanriku seacoast, which was struck by the tsunami. Measurements were performed in 2011 and 2012. RESULTS We compared FVC (forced vital capacity) % pred. and FEV1 (forced expiratory volume in 1second) % pred. of subjects between 2011 and 2012, by serial spirometry. Of the 7053 subjects studied, including 2611 men and 4442 women, FVC% pred. and FEV1% pred. were significantly higher in 2012 than in 2011. Physical indices including height, body weight and the body mass index (BMI) did not change significantly during this period. Smoking prevalence changed significantly between 2010, 2011, and 2012. Both FVC% pred. and FEV1% pred. of subjects who had quit smoking increased significantly on spirometry carried out in 2012, compared with those in 2011. CONCLUSIONS The pulmonary function expressed as FVC% pred. and FEV1% pred. were significantly higher in 2012 than in 2011 among the subjects studied. The changes in the smoking status may be one of the reasons for the increase in values observed. However, other undetermined factors during recovery from a disaster might have resulted in improved pulmonary function.
Collapse
Affiliation(s)
- Hiromi Nagashima
- Division of Pulmonary Medicine, Allergy and Rheumatology, Department of Internal Medicine, Iwate Medical University School of Medicine, Japan
| | - Itaru Fujimura
- Division of Pulmonary Medicine, Allergy and Rheumatology, Department of Internal Medicine, Iwate Medical University School of Medicine, Japan
| | - Yutaka Nakamura
- Division of Pulmonary Medicine, Allergy and Rheumatology, Department of Internal Medicine, Iwate Medical University School of Medicine, Japan
| | - Yu Utsumi
- Division of Pulmonary Medicine, Allergy and Rheumatology, Department of Internal Medicine, Iwate Medical University School of Medicine, Japan
| | - Kohei Yamauchi
- Division of Pulmonary Medicine, Allergy and Rheumatology, Department of Internal Medicine, Iwate Medical University School of Medicine, Japan.
| | - Yasuhiro Takikawa
- Division of Hepatology, Department of Internal Medicine, Iwate Medical University School of Medicine, Japan
| | - Yukari Yokoyama
- Faculty of Social Welfare, Department of Social Welfare, Nihon Fukushi University, Mihama-cho, Japan
| | - Kiyomi Sakata
- Department of Hygiene and Preventive Medicine, Iwate Medical University School of Medicine, Japan
| | | | - Akira Ogawa
- Iwate Medical University School of Medicine, Morioka, Japan
| |
Collapse
|
19
|
Doyle TJ, Dellaripa PF, Rosas IO. Risk Factors and Biomarkers of RA-ILD. LUNG DISEASE IN RHEUMATOID ARTHRITIS 2018. [DOI: 10.1007/978-3-319-68888-6_5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
|
20
|
Pharmacogenetic study of seven polymorphisms in three nicotinic acetylcholine receptor subunits in smoking-cessation therapies. Sci Rep 2017; 7:16730. [PMID: 29196725 PMCID: PMC5711795 DOI: 10.1038/s41598-017-16946-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Accepted: 11/19/2017] [Indexed: 11/23/2022] Open
Abstract
Smoking-cessation therapy reduces the risk of smoking-related diseases, but is successful only in a fraction of smokers. There is growing evidence that genetic variations in nicotinic acetylcholine receptor (nAChR) subunits influence the risk of nicotine dependence and the ability to quit smoking. To investigate the role of polymorphisms in nAChR genes on smoking quantity and the outcome of smoking-cessation therapies, we carried out an association study on 337 smokers who underwent pharmacotherapy with varenicline, bupropion, nicotine replacement therapy (NRT) alone, or NRT plus bupropion. Smoking habit and abstention were assessed from the number of cigarettes smoked per day (CPD) and the exhaled CO (eCO), at baseline and up to 12 months. We genotyped seven polymorphisms in genes encoding the nAChR subunits CHRNA4, CHRNA5, and CHRNB2. At baseline, both CPD and eCO were associated with polymorphisms in the CHRNA5 locus (rs503464, rs55853698, rs55781567 and rs16969968; P < 0.01). rs503464, a variant in the 5′-UTR of CHRNA5, was also associated with short-, mid- and long-term responses to therapy (P = 0.011, P = 0.0043, P = 0.020, respectively), although after correction for multiple testing only the association at the mid-term assessment remained significant (FDR = 0.03). These data support the role of individual genetic makeup in the ability to quit smoking.
Collapse
|
21
|
MAP3K19 Is Overexpressed in COPD and Is a Central Mediator of Cigarette Smoke-Induced Pulmonary Inflammation and Lower Airway Destruction. PLoS One 2016; 11:e0167169. [PMID: 27935962 PMCID: PMC5147866 DOI: 10.1371/journal.pone.0167169] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Accepted: 11/09/2016] [Indexed: 11/19/2022] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is characterized by persistent airflow limitation and lung inflammation resulting in a progressive decline in lung function whose principle cause is cigarette smoke. MAP3K19 is a novel kinase expressed predominantly by alveolar and interstitial macrophages and bronchial epithelial cells in the lung. We found that MAP3K19 mRNA was overexpressed in a limited sampling of lung tissue from COPD patients, and a closer examination found it to be overexpressed in bronchoalveolar macrophages from COPD patients, as well as the bronchial epithelium and inflammatory cells in the lamina propria. We further found MAP3K19 to be induced in various cell lines upon environmental stress, such as cigarette smoke, oxidative and osmotic stress. Exogenous expression of MAP3K19 in cells caused an upregulation of transcriptionally active NF-κB, and secretion of the chemokines CXCL-8, CCL-20 and CCL-7. Inhibition of MAP3K19 activity by siRNA or small molecular weight inhibitors caused a decrease in cigarette smoke-induced inflammation in various murine models, which included a decrease in pulmonary neutrophilia and KC levels. In a chronic cigarette smoke model, inhibition of MAP3K19 significantly attenuated emphysematous changes in airway parenchyma. Finally, in a viral exacerbation model, mice exposed to cigarette smoke and influenza A virus showed a decrease in pulmonary neutrophilia, pro-inflammatory cytokines and viral load upon inhibition of MAP3K19. Collectively, these results suggest that inhibition of MAP3K19 may represent a novel strategy to target COPD that promises to have a potential therapeutic benefit for patients.
Collapse
|
22
|
Drug-related pneumonitis during mammalian target of rapamycin inhibitor therapy in patients with neuroendocrine tumors: a radiographic pattern-based approach. Eur J Cancer 2016; 53:163-70. [PMID: 26760924 DOI: 10.1016/j.ejca.2015.10.015] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2015] [Accepted: 10/18/2015] [Indexed: 12/22/2022]
Abstract
PURPOSE The purpose of this study was to investigate the incidence of drug-related pneumonitis during mammalian target of rapamycin (mTOR) inhibitor therapy in patients with neuroendocrine tumours (NET) and characterise radiographic patterns of pneumonitis. METHODS Sixty-six patients (39 males, 27 females, age: 22-79 years) with advanced NET treated with mTOR inhibitor, everolimus, were retrospectively studied. Chest computed tomography scans during therapy were reviewed for abnormalities suspicious for drug-related pneumonitis by an independent review of two radiologists. Extent, distributions, and specific findings were evaluated in cases positive for pneumonitis. Radiographic patterns of pneumonitis were classified using the American Thoracic Society/European Respiratory Society classification of interstitial pneumonia. RESULTS Drug-related pneumonitis was radiographically detected in 14 patients (21%). Time from the initiation of therapy to pneumonitis was within 6 months of therapy in 10 patients (71%), while it ranged from 1.0 to 27.7 months. Pneumonitis was more common in patients who had never smoked (p=0.03). Lower lungs were more extensively involved than upper and middle lungs. Peripheral and lower distributions were most common (n=8), followed by peripheral and multifocal distributions (n=3). Ground glass and reticular opacities were present in all cases, with consolidation in eight cases. The radiographic pattern of pneumonitis was classified as cryptogenic organising pneumonia (COP) pattern in eight patients, non-specific interstitial pneumonia (NSIP) pattern in five, and hypersensitivity pneumonitis pattern in one patient. CONCLUSION Drug-related pneumonitis was noted in 21% of the advanced NET patients treated with everolimus. Radiographic pattern of pneumonitis was most commonly COP pattern, followed by NSIP pattern.
Collapse
|
23
|
Khabour O, Alzoubi KH, Abu Thiab TM, Al-Husein BA, Eissenberg T, Shihadeh A. Changes in the expression and protein level of matrix metalloproteinases after exposure to waterpipe tobacco smoke. Inhal Toxicol 2015; 27:689-93. [PMID: 26484568 PMCID: PMC4890709 DOI: 10.3109/08958378.2015.1085471] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Waterpipe smoking has become a worldwide epidemic with health consequences that only now are beginning to be understood fully. Because waterpipe use involves inhaling a large volume of toxicant-laden smoke that can cause inflammation, some health consequences may include inflammation-mediated lung injury. Excess matrix metalloproteinase expression is a key step in the etiology of toxicant exposure-driven inflammation and injury. In this study, changes in the level and mRNA of major matrix metalloproteinases (MMP-1, -9, and -12) in the lungs of mice following exposure to waterpipe smoke were investigated. Balb/c mice were exposed to waterpipe smoke for one hour daily, over a period of 2 or 8 weeks. Control mice were exposed to fresh air only. ELISA and real-time PCR techniques were used to determine the protein and mRNA levels of MMP-1, -9, and -12 in the lungs. Our findings showed that MMP-1, -9, and -12 levels in the lung significantly increased after both 2 (p < 0.05) and 8 weeks (p < 0.01) exposures. Similarly, RT-PCR findings showed that mRNA of those proteinases significantly increased following 2 (p < 0.01) and 8 weeks (p < 0.001) exposures. In conclusion, waterpipe smoking is associated strongly with lung injury as measured by elevation in the expression of MMPs in the lung tissue.
Collapse
Affiliation(s)
- Omar Khabour
- Department of Medical Laboratory Sciences, Jordan University of Science and Technology, Irbid 22110, Jordan
| | - Karem H. Alzoubi
- Department of Clinical Pharmacy, Jordan University of Science and Technology, Irbid 22110, Jordan
| | - Tuqa M. Abu Thiab
- Department of Medical Laboratory Sciences, Jordan University of Science and Technology, Irbid 22110, Jordan
| | - Belal A. Al-Husein
- Department of Clinical Pharmacy, Jordan University of Science and Technology, Irbid 22110, Jordan
| | - Thomas Eissenberg
- Department of Psychology, Virginia Commonwealth University, Richmond, VA, USA
- Center for the Study of Tobacco Products, Virginia Commonwealth University, Richmond, VA, USA
| | - Alan Shihadeh
- Mechanical Engineering Department, American University of Beirut, Beirut, Lebanon
- Center for the Study of Tobacco Products, Virginia Commonwealth University, Richmond, VA, USA
| |
Collapse
|
24
|
Xie J, Broxmeyer HE, Feng D, Schweitzer KS, Yi R, Cook TG, Chitteti BR, Barwinska D, Traktuev DO, Van Demark MJ, Justice MJ, Ou X, Srour EF, Prockop DJ, Petrache I, March KL. Human adipose-derived stem cells ameliorate cigarette smoke-induced murine myelosuppression via secretion of TSG-6. Stem Cells 2015; 33:468-78. [PMID: 25329668 DOI: 10.1002/stem.1851] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2014] [Accepted: 08/20/2014] [Indexed: 01/18/2023]
Abstract
OBJECTIVE Bone marrow-derived hematopoietic stem and progenitor cells (HSC/HPC) are critical to homeostasis and tissue repair. The aims of this study were to delineate the myelotoxicity of cigarette smoking (CS) in a murine model, to explore human adipose-derived stem cells (hASC) as a novel approach to mitigate this toxicity, and to identify key mediating factors for ASC activities. METHODS C57BL/6 mice were exposed to CS with or without i.v. injection of regular or siRNA-transfected hASC. For in vitro experiments, cigarette smoke extract was used to mimic the toxicity of CS exposure. Analysis of bone marrow HPC was performed both by flow cytometry and colony-forming unit assays. RESULTS In this study, we demonstrate that as few as 3 days of CS exposure results in marked cycling arrest and diminished clonogenic capacity of HPC, followed by depletion of phenotypically defined HSC/HPC. Intravenous injection of hASC substantially ameliorated both acute and chronic CS-induced myelosuppression. This effect was specifically dependent on the anti-inflammatory factor TSG-6, which is induced from xenografted hASC, primarily located in the lung and capable of responding to host inflammatory signals. Gene expression analysis within bone marrow HSC/HPC revealed several specific signaling molecules altered by CS and normalized by hASC. CONCLUSION Our results suggest that systemic administration of hASC or TSG-6 may be novel approaches to reverse CS-induced myelosuppression.
Collapse
Affiliation(s)
- Jie Xie
- Department of Cellular & Integrative Physiology, Herman B Wells Center for Pediatric Research, Indiana University, Indianapolis, Indiana, USA; Indiana Center for Vascular Biology and Medicine, VC-CAST Signature Center, Department of Medicine, Herman B Wells Center for Pediatric Research, Indiana University, Indianapolis, Indiana, USA; VA Center for Regenerative Medicine Indianapolis, "Richard L. Roudebush" VA Medical Center, Indianapolis, Indiana, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Heijink IH, Pouwels SD, Leijendekker C, de Bruin HG, Zijlstra GJ, van der Vaart H, ten Hacken NHT, van Oosterhout AJM, Nawijn MC, van der Toorn M. Cigarette Smoke–Induced Damage-Associated Molecular Pattern Release from Necrotic Neutrophils Triggers Proinflammatory Mediator Release. Am J Respir Cell Mol Biol 2015; 52:554-62. [DOI: 10.1165/rcmb.2013-0505oc] [Citation(s) in RCA: 81] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
|
26
|
Korzeniewski K, Nitsch-Osuch A, Konior M, Lass A. Respiratory tract infections in the military environment. Respir Physiol Neurobiol 2014; 209:76-80. [PMID: 25278277 PMCID: PMC7172564 DOI: 10.1016/j.resp.2014.09.016] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2014] [Revised: 09/23/2014] [Accepted: 09/24/2014] [Indexed: 11/08/2022]
Abstract
Soldiers in combat zones are at risk of contracting respiratory infections. Troops deployed to the harsh environment have high rates of respiratory symptoms. Over 40% of soldiers’ respiratory illnesses have been due to unknown causative agents. Limited diagnostic capabilities make it difficult to estimate the number of cases.
Military personnel fighting in contemporary battlefields as well as those participating in combat training are at risk of contracting respiratory infections. Epidemiological studies have demonstrated that soldiers deployed to the harsh environment have higher rates of newly reported respiratory symptoms than non-deployers. Acute respiratory diseases are the principle reason for outpatient treatment and hospitalization among military personnel, with an incidence exceeding that of the adult civilian population by up to three-fold. Adenoviruses, influenza A and B viruses, Streptococcus pneumoniae, Streptococcus pyogenes, coronaviruses and rhinoviruses have been identified as the main causes of acute respiratory infections among the military population. Although infective pathogens have been extensively studied, a significant proportion of illnesses (over 40%) have been due to unknown causative agents. Other health hazards, which can lead to respiratory illnesses among troops, are extreme air temperatures, desert dust, emissions from burn pits, industrial pollutants, and airborne contaminants originating from degraded soil. Limited diagnostic capabilities, especially inside the area of operations, make it difficult to accurately estimate the exact number of respiratory diseases in the military environment. The aim of the study was to discuss the occurrence of respiratory tract infections in army personnel, existing risk factors and preventive measures.
Collapse
Affiliation(s)
- Krzysztof Korzeniewski
- Military Institute of Medicine, Department of Epidemiology and Tropical Medicine, Gdynia, Poland.
| | - Aneta Nitsch-Osuch
- Warsaw Medical University, Department of Family Medicine, Warsaw, Poland
| | - Monika Konior
- Military Institute of Medicine, Department of Epidemiology and Tropical Medicine, Gdynia, Poland
| | - Anna Lass
- Medical University of Gdańsk, Department of Tropical Parasitology, Gdynia, Poland
| |
Collapse
|
27
|
Doyle TJ, Dellaripa PF, Batra K, Frits ML, Iannaccone CK, Hatabu H, Nishino M, Weinblatt ME, Ascherman DP, Washko GR, Hunninghake GM, Choi AMK, Shadick NA, Rosas IO. Functional impact of a spectrum of interstitial lung abnormalities in rheumatoid arthritis. Chest 2014; 146:41-50. [PMID: 24305643 DOI: 10.1378/chest.13-1394] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Approximately 10% of patients with rheumatoid arthritis (RA) have interstitial lung disease (ILD), and one-third have subclinical ILD on chest CT scan. In this study, we aimed to further characterize functional decrements in a spectrum of RA-associated ILD. METHODS All subjects were enrolled in the Brigham and Women's Hospital Rheumatoid Arthritis Sequential Study (BRASS). The presence of interstitial lung abnormalities (ILAs) on clinically indicated chest CT scans was determined using a previously validated sequential reading method. Univariate and multivariate analyses were used to assess the association between degree of ILAs and physiologic, functional, and demographic variables of interest. RESULTS Of 1,145 BRASS subjects, 91 subjects (8%) were included in this study. Twelve had radiologically severe ILAs, 34 had ILAs, and 38 had no ILAs on CT scan. Subjects with radiologically severe ILAs were older (P = .0037), had increased respiratory symptoms (cough, P = .027; dyspnea, P = .010), and more severe RA disease (rheumatoid factor, P = .018; total swollen joints, P = .046) compared with subjects with no ILAs. Participants also had a trend toward having an increased smoking history (P = .16) and having lower FVC % predicted (77% vs 94%, P = .097) and diffusion capacity of carbon monoxide % predicted (52% vs 77%, P = .068). Similar but attenuated increases in respiratory symptoms, functional decrements, and RA disease severity were observed in subjects with ILAs compared with those with no ILAs. CONCLUSIONS We have shown that patients with RA have varying degrees of ILAs that are associated with a spectrum of functional and physiologic decrements. Our findings suggest that improved risk stratification and detection of ILAs will provide a therapeutic window that could improve RA-ILD outcomes.
Collapse
Affiliation(s)
- Tracy J Doyle
- Pulmonary and Critical Care Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Paul F Dellaripa
- Division of Rheumatology, Immunology and Allergy, Brigham and Women's Hospital, Boston, MA
| | - Kerri Batra
- Division of Rheumatology, Rhode Island Hospital, Providence RI
| | - Michelle L Frits
- Division of Rheumatology, Immunology and Allergy, Brigham and Women's Hospital, Boston, MA
| | - Christine K Iannaccone
- Division of Rheumatology, Immunology and Allergy, Brigham and Women's Hospital, Boston, MA
| | - Hiroto Hatabu
- Center for Pulmonary Functional Imaging, Brigham and Women's Hospital, Boston, MA; Department of Radiology, Brigham and Women's Hospital, Boston, MA
| | - Mizuki Nishino
- Center for Pulmonary Functional Imaging, Brigham and Women's Hospital, Boston, MA; Department of Radiology, Brigham and Women's Hospital, Boston, MA
| | - Michael E Weinblatt
- Division of Rheumatology, Immunology and Allergy, Brigham and Women's Hospital, Boston, MA
| | - Dana P Ascherman
- Division of Rheumatology, University of Miami Miller School of Medicine, Miami FL
| | - George R Washko
- Pulmonary and Critical Care Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Gary M Hunninghake
- Pulmonary and Critical Care Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Channing Laboratory, Brigham and Women's Hospital, Boston, MA
| | - Augustine M K Choi
- Pulmonary and Critical Care Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Nancy A Shadick
- Division of Rheumatology, Immunology and Allergy, Brigham and Women's Hospital, Boston, MA
| | - Ivan O Rosas
- Pulmonary and Critical Care Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Lovelace Respiratory Research Institute, Albuquerque, NM.
| |
Collapse
|
28
|
Valiathan R, Miguez MJ, Patel B, Arheart KL, Asthana D. Tobacco smoking increases immune activation and impairs T-cell function in HIV infected patients on antiretrovirals: a cross-sectional pilot study. PLoS One 2014; 9:e97698. [PMID: 24842313 PMCID: PMC4026405 DOI: 10.1371/journal.pone.0097698] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2013] [Accepted: 04/22/2014] [Indexed: 12/20/2022] Open
Abstract
Background The influence of tobacco smoking on the immune system of HIV infected individuals is largely unknown. We investigated the impact of tobacco smoking on immune activation, microbial translocation, immune exhaustion and T-cell function in HIV infected individuals. Method HIV infected smokers and non-smokers (n = 25 each) with documented viral suppression on combination antiretroviral therapy and HIV uninfected smokers and non-smokers (n = 15 each) were enrolled. Markers of immune activation (CD38 and HLA-DR) and immune exhaustion (PD1, Tim3 and CTLA4) were analyzed in peripheral blood mononuclear cells (PBMCs) by flow cytometry. Plasma markers of microbial translocation (soluble-CD14 - sCD14 and lipopolysaccharide - LPS) were measured. Antigen specific functions of CD4+ and CD8+ T-cells were measured, by flow cytometry, in PBMCs after 6 hours stimulation with Cytomegalovirus, Epstein-Barr virus and Influenza Virus (CEF) peptide pool. Results Compared to non-smokers, smokers of HIV infected and uninfected groups showed significantly higher CD4+ and CD8+ T-cell activation with increased frequencies of CD38+HLA-DR+ cells with a higher magnitude in HIV infected smokers. Expressions of immune exhaustion markers (PD1, Tim3 and CTLA4) either alone or in combinations were significantly higher in smokers, especially on CD4+ T-cells. Compared to HIV uninfected non-smokers, microbial translocation (sCD14 and LPS) was higher in smokers of both groups and directly correlated with CD4+ and CD8+ T-cell activation. Antigen specific T-cell function showed significantly lower cytokine response of CD4+ and CD8+ T-cells to CEF peptide-pool stimulation in smokers of both HIV infected and uninfected groups. Conclusions Our results suggest that smoking and HIV infection independently influence T-cell immune activation and function and together they present the worst immune profile. Since smoking is widespread among HIV infected individuals, studies are warranted to further evaluate the cumulative effect of smoking on impairment of the immune system and accelerated disease progression.
Collapse
Affiliation(s)
- Ranjini Valiathan
- Department of Pathology, University of Miami-Miller School of Medicine, Miami, Florida, United States of America
- Laboratory for Clinical and Biological Studies, University of Miami-Miller School of Medicine, Miami, Florida, United States of America
| | - Maria J. Miguez
- School of Integrated Science and Humanities, Florida International University, Miami, Florida, United States of America
| | - Bijal Patel
- Laboratory for Clinical and Biological Studies, University of Miami-Miller School of Medicine, Miami, Florida, United States of America
| | - Kristopher L. Arheart
- Department of Public Health Sciences, Division of Biostatistics, University of Miami-Miller School of Medicine, Miami, Florida, United States of America
| | - Deshratn Asthana
- Department of Pathology, University of Miami-Miller School of Medicine, Miami, Florida, United States of America
- Department of Psychiatry and Behavioral Science, University of Miami-Miller School of Medicine, Miami, Florida, United States of America
- Laboratory for Clinical and Biological Studies, University of Miami-Miller School of Medicine, Miami, Florida, United States of America
- * E-mail:
| |
Collapse
|
29
|
Colombo G, Clerici M, Giustarini D, Portinaro NM, Aldini G, Rossi R, Milzani A, Dalle-Donne I. Pathophysiology of tobacco smoke exposure: recent insights from comparative and redox proteomics. MASS SPECTROMETRY REVIEWS 2014; 33:183-218. [PMID: 24272816 DOI: 10.1002/mas.21392] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/27/2013] [Revised: 05/23/2013] [Accepted: 05/23/2013] [Indexed: 06/02/2023]
Abstract
First-hand and second-hand tobacco smoke are causally linked to a huge number of deaths and are responsible for a broad spectrum of pathologies such as cancer, cardiovascular, respiratory, and eye diseases as well as adverse effects on female reproductive function. Cigarette smoke is a complex mixture of thousands of different chemical species, which exert their negative effects on macromolecules and biochemical pathways, both directly and indirectly. Many compounds can act as oxidants, pro-inflammatory agents, carcinogens, or a combination of these. The redox behavior of cigarette smoke has many implications for smoke related diseases. Reactive oxygen and nitrogen species (both radicals and non-radicals), reactive carbonyl compounds, and other species may induce oxidative damage in almost all the biological macromolecules, compromising their structure and/or function. Different quantitative and redox proteomic approaches have been applied in vitro and in vivo to evaluate, respectively, changes in protein expression and specific oxidative protein modifications induced by exposure to cigarette smoke and are overviewed in this review. Many gel-based and gel-free proteomic techniques have already been used successfully to obtain clues about smoke effects on different proteins in cell cultures, animal models, and humans. The further implementation with other sensitive screening techniques could be useful to integrate the comprehension of cigarette smoke effects on human health. In particular, the redox proteomic approach may also help identify biomarkers of exposure to tobacco smoke useful for preventing these effects or potentially predictive of the onset and/or progression of smoking-induced diseases as well as potential targets for therapeutic strategies.
Collapse
Affiliation(s)
- Graziano Colombo
- Department of Biosciences, Università degli Studi di Milano, Milan, Italy
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Doyle TJ, Lee JS, Dellaripa PF, Lederer JA, Matteson EL, Fischer A, Ascherman DP, Glassberg MK, Ryu JH, Danoff SK, Brown KK, Collard HR, Rosas IO. A roadmap to promote clinical and translational research in rheumatoid arthritis-associated interstitial lung disease. Chest 2014; 145:454-463. [PMID: 24590021 DOI: 10.1378/chest.13-2408] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Rheumatoid arthritis (RA) is a systemic inflammatory disorder affecting approximately 1.3 million adults in the United States. Approximately 10% of these individuals with RA have clinically evident interstitial lung disease (RA-ILD), and an additional one-third demonstrate subclinical ILD on chest CT scan. The risk of death for individuals with RA-ILD is three times higher than for patients with RA without ILD, with a median survival after ILD diagnosis of only 2.6 years. Despite the high prevalence and mortality of RA-ILD, little is known about its molecular features and its natural history. At present, we lack a standard validated approach to the definition, diagnosis, risk stratification, and management of RA-ILD. In this perspective, we discuss the importance of clinical and translational research and how ongoing research efforts can address important gaps in our knowledge over the next few years. Furthermore, recommendations are made to design multicenter collaborative studies that will expedite the development of clinical trials designed to decrease the significant morbidity and mortality associated with RA-ILD.
Collapse
Affiliation(s)
- Tracy J Doyle
- Division of Pulmonary and Critical Care Medicine Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Joyce S Lee
- Division of Pulmonary and Critical Care Medicine, University of California San Francisco School of Medicine, San Francisco, CA
| | - Paul F Dellaripa
- Division of Rheumatology, Immunology, and Allergy Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - James A Lederer
- Department of Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Eric L Matteson
- Division of Rheumatology, Mayo Clinic College of Medicine, Rochester, MN
| | - Aryeh Fischer
- Division of Rheumatology, National Jewish Health and University of Colorado, Denver, CO
| | - Dana P Ascherman
- Division of Rheumatology, University of Miami Miller School of Medicine, Miami, FL
| | - Marilyn K Glassberg
- Division of Pulmonary Medicine, University of Miami Miller School of Medicine, Miami, FL
| | - Jay H Ryu
- Division of Pulmonary and Critical Care Medicine, Mayo Clinic College of Medicine, Rochester, MN
| | - Sonye K Danoff
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Kevin K Brown
- Autoimmune Lung Center and Interstitial Lung Disease Program, National Jewish Health, Denver, CO
| | - Harold R Collard
- Division of Pulmonary and Critical Care Medicine, University of California San Francisco School of Medicine, San Francisco, CA
| | - Ivan O Rosas
- Lovelace Respiratory Research Institute, Albuquerque, NM.
| |
Collapse
|
31
|
Cheng S, Mohammed TLH. Diffuse smoking-related lung disease: emphysema and interstitial lung disease. Semin Roentgenol 2014; 50:16-22. [PMID: 25498399 DOI: 10.1053/j.ro.2014.04.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Stephanie Cheng
- Department of Radiology, Virginia Mason Medical Center, Seattle, WA
| | - Tan Lucien H Mohammed
- Department of Radiology, University of Florida College of Medicine, Gainesville, FL.
| |
Collapse
|
32
|
Agnihotri R, Gaur S. Implications of tobacco smoking on the oral health of older adults. Geriatr Gerontol Int 2014; 14:526-40. [PMID: 24697929 DOI: 10.1111/ggi.12285] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/17/2014] [Indexed: 01/28/2023]
Abstract
Cigarette smoking is the foremost health risk issue affecting individuals of all age groups globally. It specifically influences the geriatric population as a result of chronic exposure to toxins. Its role in various systemic and oral diseases including cancer, premalignant lesions, periodontitis, tooth loss, dental caries and implant failures is well established. Smoking causes immuno-inflammatory imbalances resulting in increased oxidative stress in the body. The latter hastens the immunosenescence and inflammaging process, which increases the susceptibility to infections. Thus, implementation of smoking cessation programs among older adults is imperative to prevent the development and progression of oral and systemic diseases. The present review focuses on smoking-associated oral health problems in older adults, and the steps required for cessation of the habit.
Collapse
Affiliation(s)
- Rupali Agnihotri
- Department of Periodontology, Manipal College of Dental Sciences, Manipal University, Manipal, Karnataka, India
| | | |
Collapse
|
33
|
Papiris SA, Malagari K, Manali ED, Kolilekas L, Triantafillidou C, Baou K, Rontogianni D, Bouros D, Kagouridis K. Bronchiolitis: adopting a unifying definition and a comprehensive etiological classification. Expert Rev Respir Med 2014; 7:289-306. [PMID: 23734650 DOI: 10.1586/ers.13.21] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Bronchiolitis is an inflammatory and potentially fibrosing condition affecting mainly the intralobular conducting and transitional small airways. Secondary bronchiolitis participates in disease process of the airways and/or the surrounding lobular structures in the setting of several already defined clinical entities, mostly of known etiology, and occurs commonly. Primary or idiopathic bronchiolitis dominates and characterizes distinct clinical entities, all of unknown etiology, and occurs rarely. Secondary bronchiolitis regards infections, hypersensitivity disorders, the whole spectrum of smoking-related disorders, toxic fumes and gas inhalation, chronic aspiration, particle inhalation, drug-induced bronchiolar toxicities, sarcoidosis and neoplasms. Idiopathic or primary bronchiolitis defines clinicopathologic entities sufficiently different to be designated as separate disease entities and include cryptogenic constrictive bronchiolitis, diffuse panbronchiolitis, diffuse idiopathic pulmonary neuroendocrine cell hyperplasia, neuroendocrine hyperplasia in infants, bronchiolitis obliterans syndrome in lung and allogeneic hematopoietic cell transplantation, connective tissue disorders, inflammatory bowel disease and bronchiolitis obliterans organizing pneumonia. Most of the above are pathological descriptions used as clinical diagnosis. Acute bronchiolitis, though potentially life threatening, usually regresses. Any etiology chronic bronchiolitis contributes to morbidity and/or mortality if it persists and/or progresses to diffuse airway narrowing and distortion or complete obliteration. Bronchiolitis in specific settings leads to bronchiolectasis, resulting in bronchiectasis.
Collapse
Affiliation(s)
- Spyros A Papiris
- 2nd Pulmonary Medicine Department, Attikon University Hospital, Athens Medical School, National and Kapodistrian University of Athens, Athens, Greece.
| | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Wang H, Mattes WB, Richter P, Mendrick DL. An omics strategy for discovering pulmonary biomarkers potentially relevant to the evaluation of tobacco products. Biomark Med 2013; 6:849-60. [PMID: 23227851 DOI: 10.2217/bmm.12.78] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Smoking is known to cause serious lung diseases including chronic bronchitis, chronic obstructive lung disease, obstruction of small airways, emphysema and cancer. Tobacco smoke is a complex chemical aerosol containing at least 8000 chemical constituents, either tobacco derived or added by tobacco product manufacturers. Identification of all of the toxic agents in tobacco smoke is challenging, and efforts to understand the mechanisms by which tobacco use causes disease will be informed by new biomarkers of exposure and harm. In 2009, President Obama signed into law the Family Smoking Prevention and Tobacco Control Act granting the US FDA the authority to regulate tobacco products to protect public health. This perspective article presents the background, rationale and strategy for using omics technologies to develop new biomarkers, which may be of interest to the FDA when implementing the Family Smoking Prevention and Tobacco Control Act.
Collapse
Affiliation(s)
- Honggang Wang
- Food & Drug Administration, National Center for Toxicological Research, 3900 NCTR Road, Jefferson, AR 72079, USA
| | | | | | | |
Collapse
|
35
|
Mai X, Wactawski-Wende J, Hovey KM, LaMonte MJ, Chen C, Tezal M, Genco RJ. Associations between smoking and tooth loss according to the reason for tooth loss: the Buffalo OsteoPerio Study. J Am Dent Assoc 2013; 144:252-65. [PMID: 23449901 PMCID: PMC3842224 DOI: 10.14219/jada.archive.2013.0112] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
BACKGROUND Smoking is associated with tooth loss. However, smoking's relationship to the specific reason for tooth loss in postmenopausal women is unknown. METHODS Postmenopausal women (n = 1,106) who joined a Women's Health Initiative ancillary study (The Buffalo OsteoPerio Study) underwent oral examinations for assessment of the number of missing teeth, and they reported the reasons for tooth loss. The authors obtained information about smoking status via a self-administered questionnaire. The authors calculated odds ratios (ORs) and 95 percent confidence intervals (CIs) by means of logistic regression to assess smoking's association with overall tooth loss, as well as with tooth loss due to periodontal disease (PD) and with tooth loss due to caries. RESULTS After adjusting for age, education, income, body mass index, history of diabetes diagnosis, calcium supplement use and dental visit frequency, the authors found that heavy smokers (≥ 26 pack-years) were significantly more likely to report having experienced tooth loss compared with never smokers (OR = 1.82; 95 percent CI, 1.10-3.00). Smoking status, packs smoked per day, years of smoking, pack-years and years since quitting smoking were significantly associated with tooth loss due to PD. For pack-years, the association for heavy smokers compared with that for never smokers was OR = 6.83 (95 percent CI, 3.40 -13.72). The study results showed no significant associations between smoking and tooth loss due to caries. CONCLUSIONS AND PRACTICAL IMPLICATIONS Smoking may be a major factor in tooth loss due to PD. However, smoking appears to be a less important factor in tooth loss due to caries. Further study is needed to explore the etiologies by which smoking is associated with different types of tooth loss. Dentists should counsel their patients about the impact of smoking on oral health, including the risk of experiencing tooth loss due to PD.
Collapse
Affiliation(s)
- Xiaodan Mai
- Department of Social and Preventive Medicine, School of Public Health and Health Professions, University at Buffalo, The State University of New York
| | - Jean Wactawski-Wende
- Department of Social and Preventive Medicine, School of Public Health and Health Professions, University at Buffalo, The State University of New York, 270 Farber Hall, Buffalo, N.Y. 14214
| | - Kathleen M. Hovey
- Department of Social and Preventive Medicine, School of Public Health and Health Professions, University at Buffalo, The State University of New York
| | - Michael J. LaMonte
- Department of Social and Preventive Medicine, School of Public Health and Health Professions, University at Buffalo, The State University of New York
| | - Chaoru Chen
- Department of Social and Preventive Medicine, School of Public Health and Health Professions, University at Buffalo, The State University of New York
| | - Mine Tezal
- Department of Oral Biology, School of Dental Medicine, University at Buffalo, The State University of New York
| | - Robert J. Genco
- Department of Oral Biology, School of Dental Medicine, University at Buffalo, The State University of New York
| |
Collapse
|
36
|
Baldi BG, Pereira CADC, Rubin AS, Santana ANDC, Costa AN, Carvalho CRR, Algranti E, Capitani EMD, Bethlem EP, Coletta ENAM, Arakaki JSO, Martinez JAB, Carvalho JFD, Steidle LJM, Rocha MJJ, Lima MS, Soares MR, Caramori ML, Aidé MA, Ferreira RG, Kairalla RA, Oliveira RKFD, Jezler S, Rodrigues SCS, Pimenta SP. Highlights of the Brazilian Thoracic Association guidelines for interstitial lung diseases. J Bras Pneumol 2012; 38:282-91. [PMID: 22782597 DOI: 10.1590/s1806-37132012000300002] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2012] [Accepted: 06/11/2012] [Indexed: 01/30/2023] Open
Abstract
Interstitial lung diseases (ILDs) are heterogeneous disorders, involving a large number of conditions, the approach to which continues to pose an enormous challenge for pulmonologists. The 2012 Brazilian Thoracic Association ILD Guidelines were established in order to provide Brazilian pulmonologists with an instrument that can facilitate the management of patients with ILDs, standardizing the criteria used for the diagnosis of different conditions and offering guidance on the best treatment in various situations. The objective of this article was to briefly describe the highlights of those guidelines.
Collapse
Affiliation(s)
- Bruno Guedes Baldi
- Instituto do Coração Divisão de Pneumologia, Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brazil.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Cyto-genotoxic effects of smoke from commercial filter and non-filter cigarettes on human bronchial and pulmonary cells. Mutat Res 2012; 750:1-11. [PMID: 23010388 DOI: 10.1016/j.mrgentox.2012.06.013] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2011] [Revised: 05/30/2012] [Accepted: 06/26/2012] [Indexed: 01/08/2023]
Abstract
Cigarette smoke is a complex mixture of chemicals, some of which are known as carcinogens. The cyto-genotoxic effects of cigarette-smoke extract (CSE) from commercial cigarettes without (A and B) and with filter (C and D) were evaluated at different CSE concentrations on A549 and BEAS-2B cells. The particle content of the cigarette smoke and the metal composition of the CSE were also analyzed. The cells were exposed to 1-10% of the CSE from one cigarette per experiment. Cytotoxicity was evaluated by use of the MTT assay after 24h, and the lactate dehydrogenase (LDH) assay after 30min and 24h. The Fpg-modified comet assay was used to evaluate direct-oxidative DNA damage on cells exposed for 30min. As expected, unfiltered cigarette smoke (particularly from the B cigarette) contained a higher number of particles than filtered smoke. With smoke extract from the B cigarette we found a decrease in cell viability only in BEAS-2B cells. The results of the LDH test showed membrane damage for B-cigarette smoke extract, particularly in BEAS-2B cells. Extracts from unfiltered cigarette smoke induced significant direct DNA damage, to a larger extent in A549 cells. Filtered cigarette-smoke extract induced a significant direct DNA damage at 5-10%. A significant induction of oxidative DNA damage was found at the highest CSE concentration in both cell types (by smoke extracts from B and C cigarettes in A549 cells, and from A and D cigarettes in BEAS-2B cells). Smoke extracts from filter cigarettes induced less direct DNA damage than those from unfiltered cigarettes in A549 cells, probably due to a protective effect of filter. In BEAS-2B cells the smoke extract from the B-cigarette showed the highest genotoxic effect, with a concentration-dependent trend. These findings show a higher cyto-genotoxicity for smoke extracts from the B-cigarette and oxidative effects for those from the A and D cigarettes, particularly in BEAS-2B cells. Moreover, there was a higher responsiveness of A549 cells to genotoxic insult of CSE, and a cigarette-dependent genotoxicity in BEAS-2B cells. Our experimental model demonstrated to be suitable to sensitively detect early genotoxic response of different lung-cell types to non-cytotoxic concentrations of complex inhalable mixtures.
Collapse
|
38
|
Cigarette smoke-induced accumulation of lung dendritic cells is interleukin-1α-dependent in mice. Respir Res 2012; 13:81. [PMID: 22992200 PMCID: PMC3519608 DOI: 10.1186/1465-9921-13-81] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2012] [Accepted: 08/29/2012] [Indexed: 12/02/2022] Open
Abstract
Background Evidence suggests that dendritic cells accumulate in the lungs of COPD patients and correlate with disease severity. We investigated the importance of IL-1R1 and its ligands IL-1α and β to dendritic cell accumulation and maturation in response to cigarette smoke exposure. Methods Mice were exposed to cigarette smoke using a whole body smoke exposure system. IL-1R1-, TLR4-, and IL-1α-deficient mice, as well as anti-IL-1α and anti-IL-1β blocking antibodies were used to study the importance of IL-1R1 and TLR4 to dendritic cell accumulation and activation. Results Acute and chronic cigarette smoke exposure led to increased frequency of lung dendritic cells. Accumulation and activation of dendritic cells was IL-1R1/IL-1α dependent, but TLR4- and IL-1β-independent. Corroborating the cellular data, expression of CCL20, a potent dendritic cells chemoattractant, was IL-1R1/IL-1α-dependent. Studies using IL-1R1 bone marrow-chimeric mice revealed the importance of IL-1R1 signaling on lung structural cells for CCL20 expression. Consistent with the importance of dendritic cells in T cell activation, we observed decreased CD4+ and CD8+ T cell activation in cigarette smoke-exposed IL-1R1-deficient mice. Conclusion Our findings convey the importance of IL-1R1/IL-1α to the recruitment and activation of dendritic cells in response to cigarette smoke exposure.
Collapse
|
39
|
Caponnetto P, Russo C, Auditore R, Polosa R. The smoker with interstitial lung disease and interventions for successful smoking cessation. REVISTA PORTUGUESA DE PNEUMOLOGIA 2012; 18:285-8. [PMID: 22884276 DOI: 10.1016/j.rppneu.2012.06.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2012] [Revised: 04/27/2012] [Accepted: 06/06/2012] [Indexed: 11/18/2022] Open
Affiliation(s)
- P Caponnetto
- Centro per la Prevenzione e Cura del Tabagismo (CPCT), Azienda Ospedaliero-Universitaria Policlinico-Vittorio Emanuele, Università di Catania, Catania, Italy.
| | | | | | | |
Collapse
|
40
|
Doyle TJ, Hunninghake GM, Rosas IO. Subclinical interstitial lung disease: why you should care. Am J Respir Crit Care Med 2012; 185:1147-53. [PMID: 22366047 DOI: 10.1164/rccm.201108-1420pp] [Citation(s) in RCA: 110] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
The widespread use of high-resolution computed tomography in clinical and research settings has increased the detection of interstitial lung abnormalities (ILA) in asymptomatic and undiagnosed individuals. We reported that in smokers, ILA were present in about 1 of every 12 high-resolution computed tomographic scans; however, the long-term significance of these subclinical changes remains unclear. Studies in families affected with pulmonary fibrosis, smokers with chronic obstructive pulmonary disease, and patients with inflammatory lung disease have shown that asymptomatic and undiagnosed individuals with ILA have reductions in lung volume, functional limitations, increased pulmonary symptoms, histopathologic changes, and molecular profiles similar to those observed in patients with clinically significant interstitial lung disease (ILD). These findings suggest that, in select at-risk populations, ILA may represent early stages of pulmonary fibrosis or subclinical ILD. The growing interest surrounding this topic is motivated by our poor understanding of the inciting events and natural history of ILD, coupled with a lack of effective therapies. In this perspective, we outline past and current research focused on validating radiologic, physiological, and molecular methods to detect subclinical ILD. We discuss the limitations of the available cross-sectional studies and the need for future longitudinal studies to determine the prognostic and therapeutic implications of subclinical ILD in populations at risk of developing clinically significant ILD.
Collapse
Affiliation(s)
- Tracy J Doyle
- Pulmonary and Critical Care Division, Department of Medicine, Brigham and Women's Hospital, 75 Francis Street, Boston, MA 02115, USA
| | | | | |
Collapse
|
41
|
Colombo G, Rossi R, Gagliano N, Portinaro N, Clerici M, Annibal A, Giustarini D, Colombo R, Milzani A, Dalle-Donne I. Red blood cells protect albumin from cigarette smoke-induced oxidation. PLoS One 2012; 7:e29930. [PMID: 22238677 PMCID: PMC3251585 DOI: 10.1371/journal.pone.0029930] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2011] [Accepted: 12/07/2011] [Indexed: 11/18/2022] Open
Abstract
Different studies reported the presence of oxidized (carbonylated) albumin in the extravascular pool, but not in the intravascular one of cigarette smokers. In this study we attempted to explain this apparent discrepancy exposing human serum albumin (HSA) to aqueous cigarette smoke extract (CSE). CSE induces HSA carbonylation and oxidation of the HSA Cys34 sulfhydryl group. An antioxidant action of glutathione, cysteine, and its synthetic derivative N-acetylcysteine was observed only at supra-physiological concentrations, suggesting that physiological (plasma) concentrations of glutathione and cysteine in the low micromolar range are ineffective in preventing cigarette smoke-induced oxidation of HSA. Differently, human erythrocytes resulted to be protective towards CSE-induced oxidation (carbonylation and thiol oxidation) of both HSA and total human plasma proteins.
Collapse
Affiliation(s)
- Graziano Colombo
- Department of Biology, Università degli Studi di Milano, Milan, Italy
| | - Ranieri Rossi
- Department of Evolutionary Biology, University of Siena, Siena, Italy
| | - Nicoletta Gagliano
- Department of Human Morphology and Biomedical Sciences “Città Studi”, Università degli Studi di Milano, Milan, Italy
| | - Nicola Portinaro
- Department of Translational Medicine, Clinica Ortopedica e Traumatologica, Istituto Clinico Humanitas and Università degli Studi di Milano, Rozzano, Milan, Italy
| | - Marco Clerici
- Department of Biology, Università degli Studi di Milano, Milan, Italy
| | - Andrea Annibal
- Department of Biology, Università degli Studi di Milano, Milan, Italy
| | | | - Roberto Colombo
- Department of Biology, Università degli Studi di Milano, Milan, Italy
| | - Aldo Milzani
- Department of Biology, Università degli Studi di Milano, Milan, Italy
| | | |
Collapse
|
42
|
Abstract
Cigarette smoke, a toxic collection of thousands of chemicals generated from combustion of tobacco, is recognized as the primary causative agent of certain diffuse interstitial and bronchiolar lung diseases. Most patients afflicted with these disorders are cigarette smokers, and smoking cessation has been shown to be capable of inducing disease remission and should occupy a pivotal role in the management of all smokers with these diffuse lung diseases. The role of pharmacotherapy with corticosteroids or other immunomodulating agents is not well established but may be considered in patients with progressive forms of smoking-related interstitial lung diseases.
Collapse
Affiliation(s)
- Robert Vassallo
- Division of Pulmonary and Critical Care Medicine, Mayo Clinic, Rochester, MN 55905, USA.
| | | |
Collapse
|
43
|
Botelho FM, Bauer CMT, Finch D, Nikota JK, Zavitz CCJ, Kelly A, Lambert KN, Piper S, Foster ML, Goldring JJP, Wedzicha JA, Bassett J, Bramson J, Iwakura Y, Sleeman M, Kolbeck R, Coyle AJ, Humbles AA, Stämpfli MR. IL-1α/IL-1R1 expression in chronic obstructive pulmonary disease and mechanistic relevance to smoke-induced neutrophilia in mice. PLoS One 2011; 6:e28457. [PMID: 22163019 PMCID: PMC3232226 DOI: 10.1371/journal.pone.0028457] [Citation(s) in RCA: 116] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2011] [Accepted: 11/08/2011] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND Cigarette smoking is the main risk factor for the development of chronic obstructive pulmonary disease (COPD), a major cause of morbidity and mortality worldwide. Despite this, the cellular and molecular mechanisms that contribute to COPD pathogenesis are still poorly understood. METHODOLOGY AND PRINCIPAL FINDINGS The objective of this study was to assess IL-1 α and β expression in COPD patients and to investigate their respective roles in perpetuating cigarette smoke-induced inflammation. Functional studies were pursued in smoke-exposed mice using gene-deficient animals, as well as blocking antibodies for IL-1α and β. Here, we demonstrate an underappreciated role for IL-1α expression in COPD. While a strong correlation existed between IL-1α and β levels in patients during stable disease and periods of exacerbation, neutrophilic inflammation was shown to be IL-1α-dependent, and IL-1β- and caspase-1-independent in a murine model of cigarette smoke exposure. As IL-1α was predominantly expressed by hematopoietic cells in COPD patients and in mice exposed to cigarette smoke, studies pursued in bone marrow chimeric mice demonstrated that the crosstalk between IL-1α+ hematopoietic cells and the IL-1R1+ epithelial cells regulates smoke-induced inflammation. IL-1α/IL-1R1-dependent activation of the airway epithelium also led to exacerbated inflammatory responses in H1N1 influenza virus infected smoke-exposed mice, a previously reported model of COPD exacerbation. CONCLUSIONS AND SIGNIFICANCE This study provides compelling evidence that IL-1α is central to the initiation of smoke-induced neutrophilic inflammation and suggests that IL-1α/IL-1R1 targeted therapies may be relevant for limiting inflammation and exacerbations in COPD.
Collapse
Affiliation(s)
- Fernando M. Botelho
- Department of Pathology and Molecular Medicine, Centre for Gene Therapeutics, McMaster University, Hamilton, Canada
| | - Carla M. T. Bauer
- Medical Sciences Graduate Program, McMaster University, Hamilton, Canada
| | | | - Jake K. Nikota
- Medical Sciences Graduate Program, McMaster University, Hamilton, Canada
| | - Caleb C. J. Zavitz
- Department of Pathology and Molecular Medicine, Centre for Gene Therapeutics, McMaster University, Hamilton, Canada
| | - Ashling Kelly
- Department of Pathology and Molecular Medicine, Centre for Gene Therapeutics, McMaster University, Hamilton, Canada
| | - Kristen N. Lambert
- Department of Pathology and Molecular Medicine, Centre for Gene Therapeutics, McMaster University, Hamilton, Canada
| | - Sian Piper
- MedImmune LTD, Cambridge, United Kingdom
| | - Martyn L. Foster
- Research and Development, AstraZeneca, Charnwood, United Kingdom
| | - James J. P. Goldring
- Academic Unit of Respiratory Medicine, University College London, London, United Kingdom
| | - Jadwiga A. Wedzicha
- Academic Unit of Respiratory Medicine, University College London, London, United Kingdom
| | - Jennifer Bassett
- Department of Pathology and Molecular Medicine, Centre for Gene Therapeutics, McMaster University, Hamilton, Canada
- Medical Sciences Graduate Program, McMaster University, Hamilton, Canada
| | - Jonathan Bramson
- Department of Pathology and Molecular Medicine, Centre for Gene Therapeutics, McMaster University, Hamilton, Canada
| | - Yoichiro Iwakura
- Center for Experimental Medicine and Systems Biology, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | | | - Roland Kolbeck
- MedImmune LLC, Gaithersburg, Maryland, United States of America
| | | | | | - Martin R. Stämpfli
- Department of Pathology and Molecular Medicine, Centre for Gene Therapeutics, McMaster University, Hamilton, Canada
- Department of Medicine, McMaster University, Hamilton, Canada
| |
Collapse
|
44
|
van Eijl S, Mortaz E, Ferreira AF, Kuper F, Nijkamp FP, Folkerts G, Bloksma N. Humic acid enhances cigarette smoke-induced lung emphysema in mice and IL-8 release of human monocytes. Pulm Pharmacol Ther 2011; 24:682-9. [PMID: 21820074 DOI: 10.1016/j.pupt.2011.07.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2010] [Revised: 06/19/2011] [Accepted: 07/02/2011] [Indexed: 01/04/2023]
Abstract
UNLABELLED Tobacco smoke is the main factor in the etiology of lung emphysema. Generally prolonged, substantial exposure is required to develop the disease. Humic acid is a major component of cigarette smoke that accumulates in smokers' lungs over time and induces tissue damage. OBJECTIVES To investigate whether humic acid pre-loading potentiates the development of cigarette smoke-induced lung emphysema in mice and increases IL-8 release by human monocytes. METHODS C57BL/6J mice received humic acid or aqueous vehicle by tracheal installation on day 0 and day 7. From day 21 to day 84, the mice were exposed to cigarette smoke or clean air for 5 days/week. Twenty-four hours after the last exposure we determined leukocytes in lung lavage, heart hypertrophy and alveolar wall destruction. Human monocytes were incubated with cigarette smoke extract (CSE), humic acid or the combination overnight. RESULTS Humic acid nor cigarette smoke caused alveolar wall destruction within two months. Interestingly, the combination did induce lung emphysema. Humic acid, cigarette smoke or the combination did not change leukocyte types and numbers in lung lavage fluid, but the combination caused peribronchiolar and perivascular lymphocyte infiltration. Humic acid treatment resulted in a high proportion of alveolar macrophages heavily loaded with intracellular granula. Humic acid also induces the release of IL-8 from human monocytes and enhances the CSE-induced IL-8 release. CONCLUSIONS Humic acid deposition in the lungs potentiates the development of cigarette smoke-induced interstitial inflammation and lung emphysema. Moreover, humic acid promotes IL-8 release from human monocytes. Since humic acid accumulates steadily in the lungs of smokers, this may provide an explanation for the natural history on late onset of this disease. The model described here offers a novel way to study emphysema and may direct the search for new therapeutic approaches.
Collapse
Affiliation(s)
- Sven van Eijl
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, The Netherlands
| | | | | | | | | | | | | |
Collapse
|
45
|
Didon L, Barton JL, Roos AB, Gaschler GJ, Bauer CMT, Berg T, Stämpfli MR, Nord M. Lung Epithelial CCAAT/Enhancer-binding Protein-β Is Necessary for the Integrity of Inflammatory Responses to Cigarette Smoke. Am J Respir Crit Care Med 2011; 184:233-42. [DOI: 10.1164/rccm.201007-1113oc] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
|
46
|
Frei M, Engel Brügger O, Sendi P, Reichart PA, Ramseier CA, Bornstein MM. Assessment of smoking behaviour in the dental setting. A study comparing self-reported questionnaire data and exhaled carbon monoxide levels. Clin Oral Investig 2011; 16:755-60. [PMID: 21717094 DOI: 10.1007/s00784-011-0583-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2011] [Accepted: 06/16/2011] [Indexed: 11/27/2022]
Abstract
The present study validated the accuracy of data from a self-reported questionnaire on smoking behaviour with the use of exhaled carbon monoxide (CO) level measurements in two groups of patients. Group 1 included patients referred to an oral medicine unit, whereas group 2 was recruited from the daily outpatient service. All patients filled in a standardized questionnaire regarding their current and former smoking habits. Additionally, exhaled CO levels were measured using a monitor. A total of 121 patients were included in group 1, and 116 patients were included in group 2. The mean value of exhaled CO was 7.6 ppm in the first group and 9.2 ppm in the second group. The mean CO values did not statistically significantly differ between the two groups. The two exhaled CO level measurements taken for each patient exhibited very good correlation (Spearman's coefficient of 0.9857). Smokers had a mean difference of exhaled CO values of 13.95 ppm (p < 0.001) compared to non-smokers adjusted for the first or second group. The consumption of one additional pack year resulted in an increase in CO values of 0.16 ppm (p = 0.003). The consumption of one additional cigarette per day elevated the CO measurements by 0.88 ppm (p < 0.001). Based on these results, the correlations between the self-reported smoking habits and exhaled CO values are robust and highly reproducible. CO monitors may offer a non-invasive method to objectively assess current smoking behaviour and to monitor tobacco use cessation attempts in the dental setting.
Collapse
Affiliation(s)
- Marc Frei
- Department of Oral Surgery and Stomatology, School of Dental Medicine, University of Bern, Freiburgstrasse 7, CH-3010 Bern, Switzerland
| | | | | | | | | | | |
Collapse
|
47
|
Jhun BW, Kim DM, Park JH, Jung H, Song L, Han J, Chung MP. A Case of Non-Small Cell Lung Cancer in a Respiratory Bronchiolitis Associated Interstitial Lung Disease Patient. Tuberc Respir Dis (Seoul) 2011. [DOI: 10.4046/trd.2011.71.3.216] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Affiliation(s)
- Byung Woo Jhun
- Department of Internal Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Da Min Kim
- Department of Internal Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Ji Hyeon Park
- Department of Internal Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Hyunae Jung
- Department of Internal Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Limhwa Song
- Department of Internal Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Joungho Han
- Department of Pathology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Man Pyo Chung
- Department of Internal Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| |
Collapse
|
48
|
Bollo de Miguel E, Romero de Tejada JAG, Morales CJ, González FM. Neumonías intersticiales idiopáticas. Arch Bronconeumol 2011; 47 Suppl 8:15-9. [DOI: 10.1016/s0300-2896(11)70061-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
49
|
Abstract
Interstitial lung disease (ILD) is a frequent pulmonary complication of systemic sclerosis (SSc), and nonspecific interstitial pneumonia is the most commonly recognized pattern of lung injury in these patients. In this report, we describe a never-smoker female presenting with Raynaud phenomenon and ILD that demonstrated desquamative interstitial pneumonia (DIP) on surgical lung biopsy. After 8 months, she was diagnosed with pulmonary hypertension at which time clinical examinations and serologic findings established the diagnosis of SSc. This case report expands the spectrum of patterns of ILD seen in association with SSc to include DIP.
Collapse
|
50
|
Patients' awareness of the potential benefit of smoking cessation. A study evaluating self-reported and clinical data from patients referred to an oral medicine unit. Clin Oral Investig 2010; 16:55-62. [PMID: 21120675 DOI: 10.1007/s00784-010-0488-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2010] [Accepted: 11/15/2010] [Indexed: 01/06/2023]
Abstract
The present study analyzed history of smoking and willingness to quit smoking in patients referred for diagnosis and treatment of different oral mucosal lesions. Prior to the initial clinical examination, patients filled in a standardized questionnaire regarding their current and former smoking habits and willingness to quit. Definitive diagnoses were classified into three groups (benign/reactive lesions, premalignant lesions and conditions, and malignant diseases) and correlated with the self-reported data in the questionnaires. Of the 980 patients included, 514 (52%) described themselves as never smokers, 202 (21%) as former smokers, and 264 (27%) as current smokers. In the group of current smokers, 23% thought their premalignant lesions/conditions were related to their smoking habit, but only 15% of the patients with malignant mucosal diseases saw that correlation. Only 14% of the smokers wanted to commence smoking cessation within the next 30 days. Patients with malignant diseases (31%) showed greater willingness to quit than patients diagnosed with benign/reactive lesions (11%). Future clinical studies should attempt (1) to enhance patients' awareness of the negative impact of smoking on the oral mucosa and (2) to increase willingness to quit in smokers referred to a dental/oral medicine setting.
Collapse
|