1
|
Liu Y, Li H, Zhao X. Sinomenine attenuates lipopolysaccharide-induced inflammation and apoptosis of WI-38 cells by reducing glutathione S-transferase M1 expression. Chem Biol Drug Des 2023; 102:434-443. [PMID: 36303295 DOI: 10.1111/cbdd.14161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Revised: 10/05/2022] [Accepted: 10/22/2022] [Indexed: 12/01/2022]
Abstract
Pediatric pneumonia is an infectious lung disease with high morbidity and mortality. Sinomenine, an alkaloid extracted from Caulis Sinomenii, exerts anti-inflammatory and anti-apoptotic activities. Lipopolysaccharide (LPS) is widely used for the establishment of an inflammatory model. This research aimed to explore the influences of sinomenine on LPS-caused inflammatory injuries in fetal lung WI-38 cells. WI-38 cells were treated with LPS to establish a cellular model of pediatric pneumonia. Cell viability was evaluated using CCK-8 assay. Apoptosis was evaluated using TUNEL staining and caspase-3 activity assays. Inflammatory cytokines and NF-κB p65 phosphorylation levels were measured by Enzyme-Linked Immunosorbent Assay. Glutathione S-transferase M1 (GSTM1) expression was detected by western blotting. Results showed that LPS reduced WI-38 cell viability, and sinomenine protected cells against LPS-induced viability reduction. Sinomenine concentration-dependently attenuated LPS-induced inflammation by reducing TNF-α, IL-1β and MCP-1, and increasing IL-10 levels. Sinomenine mitigated LPS-induced apoptosis. GSTM1 was screened by matching the targets of sinomenine and pediatric pneumonia. GSTM1 was upregulated in LPS-treated WI-38 cells, and this effect was attenuated after sinomenine treatment. GSTM1 was upstream of NF-κB pathway. Overexpression of GSTM1 reversed the suppressive functions of sinomenine on LPS-stimulated inflammation and apoptosis. Overall, sinomenine attenuates inflammation and apoptosis in WI-38 cells stimulated by LPS via inhibiting GSTM1 expression, indicating the therapeutic potential of sinomenine in pediatric pneumonia.
Collapse
Affiliation(s)
- Yan Liu
- Department of Paediatrics, The First Hospital of Yulin, Yulin, China
| | - Huilin Li
- Department of Nuclear Medicine, The First Hospital of Yulin, Yulin, China
| | - Xiao Zhao
- Outpatient Department of Pediatrics, Qingdao Municipal Hospital (Group), Qingdao, China
| |
Collapse
|
2
|
Park SY, Kim KS, Lee WY, Kim CE, Lee S. Integrative Approach to Identifying System-Level Mechanisms of Chung-Sang-Bo-Ha-Hwan's Influence on Respiratory Tract Diseases: A Network Pharmacological Analysis with Experimental Validation. PLANTS (BASEL, SWITZERLAND) 2023; 12:3024. [PMID: 37687271 PMCID: PMC10489874 DOI: 10.3390/plants12173024] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 08/19/2023] [Accepted: 08/21/2023] [Indexed: 09/10/2023]
Abstract
Chung-Sang-Bo-Ha-Hwan (CSBHH) is an herbal prescription widely used to treat various chronic respiratory diseases. To investigate the system-level treatment mechanisms of CSBHH in respiratory tract diseases, we identified 56 active ingredients of CSBHH and evaluated the degree of overlap between their targets and respiratory tract disease-associated proteins. We then investigated the respiratory tract disease-related signaling pathways associated with CSBHH targets. Enrichment analysis showed that the CSBHH targets were significantly associated with various signaling pathways related to inflammation, alveolar structure, and tissue fibrosis. Experimental validation was conducted using phorbol-12-myristate-13-acetate (PMA)-stimulated NCI-H292 cells by analyzing the mRNA expression levels of biomarkers (IL-1β and TNF-α for inflammation; GSTP1, GSTM1, and PTEN for apoptosis) derived from network pharmacological analysis, in addition to the mucin genes MUC5AC and MUC2, to investigate the phlegm-expelling effect of CSBHH. The mRNA expression levels of these genes were consistent with network pharmacological predictions in a concentration-dependent manner. These results suggest that the therapeutic mechanisms of CSBHH in respiratory tract diseases could be attributed to the simultaneous action of multiple active ingredients in the herbal prescription.
Collapse
Affiliation(s)
- Sa-Yoon Park
- Department of Physiology, College of Korean Medicine, Gachon University, Seongnam 13120, Republic of Korea; (S.-Y.P.); (W.-Y.L.)
| | - Kang-Sub Kim
- College of Korean Medicine, Gachon University, Seongnam 13120, Republic of Korea;
| | - Won-Yung Lee
- Department of Physiology, College of Korean Medicine, Gachon University, Seongnam 13120, Republic of Korea; (S.-Y.P.); (W.-Y.L.)
| | - Chang-Eop Kim
- Department of Physiology, College of Korean Medicine, Gachon University, Seongnam 13120, Republic of Korea; (S.-Y.P.); (W.-Y.L.)
| | - Sullim Lee
- Department of Life Science, College of Bio-Nano Technology, Gachon University, Seongnam 13120, Republic of Korea
| |
Collapse
|
3
|
Jangala M, Manche SK, Katika MM, Koralla RM, Akka J. Association of CYP1A2 and GST gene variants with asthma in cases presenting with allergic chronic rhinosinusitis. EGYPTIAN JOURNAL OF MEDICAL HUMAN GENETICS 2023. [DOI: 10.1186/s43042-023-00397-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/09/2023] Open
Abstract
Abstract
Background
Inter-individual differences in regulation and activity of xenobiotic metabolizing enzymes (XMEs) CYP1A and GST might cause distinct susceptibility to chronic rhinosinusitis (CRS) phenotypes that need to be explored. Therefore, the present study aimed to evaluate the role and risk of CYP1A and GST gene variants in allergic CRS subjects with and without asthma. A total of 224 allergic CRS cases with asthma, 252 allergic CRS cases without asthma, and 350 healthy control subjects were subjected to genetic analysis. Gene variants of cytochrome P450 (CYP1A1 T3801 rs4646903, A2455G rs1048943, C2453A rs1799814 and CYP1A2 G3858A rs2069514, T739G rs2069526, C163A rs762551) and glutathione S-transferase P (GSTP1 A313G rs1605 & C341T rs1799811) were investigated by polymerase chain reaction-restriction fragment length polymorphism and GSTM1null, and GSTT1null by multiplex PCR methods.
Results
TG genotype of CYP1A2 rs2069526 (OR 1.73, 95% CI 1.20–2.50, p < 0.002), TC genotype of CYP1A1 rs4646903 (OR 1.43, 95% CI 1.03–1.98, p < 0.031) and GSTM1del (OR 1.87, 95% CI 1.24–2.81, p < 0.003) and were found to be significantly associated with only allergic CRS cases. CYP1A2 rs2069526 (OR 2.33, 95% CI 1.61–3.37, p < 0.001), GG genotype of GSTP1 rs1605 (OR 4.75, 95% CI 2.62–8.63, p < 0.001), GSTM1del (OR 1.82, 95% CI 1.19–2.78, p < 0.006), GSTM1/GSTT1 double null (OR 2.58, 95% CI 1.36–4.87, p < 0.004) and were found to be significantly associated with asthma in allergic CRS cases. Further, G-G-C haplotype of CYP1A2 rs2069514, rs2069526 and rs762551 gene variants was found to increase the risk for asthma by 5 folds in allergic CRS subjects (OR 5.53, 95% CI 1.76–17.31, p < 0.003) while T-G-C haplotype of CYP1A1 rs4646903, rs1048943, rs1799814 (OR 0.11, 95% CI (0.01–0.95, p < 0.045) and A-T haplotype of GSTP1 rs1605, rs1799811 (OR 0.27, 95% CI (0.08–0.89, p < 0.032) showed protective effect in allergic CRS group.
Conclusion
The present study reports the significantly increased association of CYP1A2, GSTM, and GSTP gene variants with asthma in allergic CRS.
Collapse
|
4
|
Zeng X, Tian G, Zhu J, Yang F, Zhang R, Li H, An Z, Li J, Song J, Jiang J, Liu D, Wu W. Air pollution associated acute respiratory inflammation and modification by GSTM1 and GSTT1 gene polymorphisms: a panel study of healthy undergraduates. Environ Health 2023; 22:14. [PMID: 36703205 PMCID: PMC9881318 DOI: 10.1186/s12940-022-00954-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Accepted: 12/28/2022] [Indexed: 06/18/2023]
Abstract
Epidemiological evidence has linked air pollution with adverse respiratory outcomes, but the mechanisms underlying susceptibility to air pollution remain unclear. This study aimed to investigate the role of glutathione S-transferase (GST) polymorphism in the association between air pollution and lung function levels. A total of 75 healthy young volunteers aged 18-20 years old were recruited for six follow-up visits and examinations. Spirometry was conducted to obtain lung function parameters such as forced vital capacity (FVC), and forced expiratory volume in 1 s (FEV1). Nasal fluid concentrations of interleukin-6 (IL-6), interleukin-8 (IL-8), tumor necrosis factor-α (TNF-α), and 8-epi-prostaglandin F2α (8-epi-PGF2a) were measured using ELISA kits. Linear mixed-effect models were used to evaluate the association of air pollutants with respiratory outcomes. Additionally, polymorphisms of glutathione S-transferase mu 1 (GSTM1) and glutathione S-transferase theta 1 (GSTT1) were estimated to explore its role in the association between air pollutants and lung function. We found that short-term exposure to atmospheric particulates such as PM2.5 and PM10 can cause an increase in nasal biomarkers of inflammation, oxidative stress, and lung function, while air gaseous pollutant exposure is linked with decreased lung function, except for CO. Stratification analyses showed that an increase in nasal inflammatory cytokines caused by exposure to atmospheric particulates is more obvious in subjects with GSTM1-sufficient (GSTM1+) than GSTM1-null (GSTM1-), while elevated lung function levels due to air particles are more significant in subjects with the genotype of GSTM1- when compared to GSTM1+. As for air gaseous pollutants, decreased lung function levels caused by O3, SO2, and NO2 exposure is more manifest in subjects with the genotype of GSTM1- compared to GSTM1+. Taken together, short-term exposure to air pollutants is associated with alterations in nasal biomarkers and lung function levels in young healthy adults, and susceptible genotypes play an important mediation role in the association between exposure to air pollutants and inflammation, oxidative stress, and lung function levels.
Collapse
Affiliation(s)
- Xiang Zeng
- Henan International Collaborative Laboratory for Health Effects and Intervention of Air Pollution, School of Public Health, Xinxiang Medical University, 601 Jinsui Road, Xinxiang, Henan, 453003, China
- School of Public Health, Zhejiang Chinese Medical University, 548 Binwen Road, Hangzhou, 310053, Zhejiang Province, China
| | - Ge Tian
- Henan International Collaborative Laboratory for Health Effects and Intervention of Air Pollution, School of Public Health, Xinxiang Medical University, 601 Jinsui Road, Xinxiang, Henan, 453003, China
| | - Jingfang Zhu
- Henan International Collaborative Laboratory for Health Effects and Intervention of Air Pollution, School of Public Health, Xinxiang Medical University, 601 Jinsui Road, Xinxiang, Henan, 453003, China
| | - Fuyun Yang
- Henan International Collaborative Laboratory for Health Effects and Intervention of Air Pollution, School of Public Health, Xinxiang Medical University, 601 Jinsui Road, Xinxiang, Henan, 453003, China
| | - Rui Zhang
- Henan International Collaborative Laboratory for Health Effects and Intervention of Air Pollution, School of Public Health, Xinxiang Medical University, 601 Jinsui Road, Xinxiang, Henan, 453003, China
| | - Huijun Li
- Henan International Collaborative Laboratory for Health Effects and Intervention of Air Pollution, School of Public Health, Xinxiang Medical University, 601 Jinsui Road, Xinxiang, Henan, 453003, China
| | - Zhen An
- Henan International Collaborative Laboratory for Health Effects and Intervention of Air Pollution, School of Public Health, Xinxiang Medical University, 601 Jinsui Road, Xinxiang, Henan, 453003, China
| | - Juan Li
- Henan International Collaborative Laboratory for Health Effects and Intervention of Air Pollution, School of Public Health, Xinxiang Medical University, 601 Jinsui Road, Xinxiang, Henan, 453003, China
| | - Jie Song
- Henan International Collaborative Laboratory for Health Effects and Intervention of Air Pollution, School of Public Health, Xinxiang Medical University, 601 Jinsui Road, Xinxiang, Henan, 453003, China
| | - Jing Jiang
- Henan International Collaborative Laboratory for Health Effects and Intervention of Air Pollution, School of Public Health, Xinxiang Medical University, 601 Jinsui Road, Xinxiang, Henan, 453003, China
| | - Dongling Liu
- School of Basic Medical Science, Zhejiang Chinese Medical University, 548 Binwen Road, Hangzhou, 310053, Zhejiang Province, China
| | - Weidong Wu
- Henan International Collaborative Laboratory for Health Effects and Intervention of Air Pollution, School of Public Health, Xinxiang Medical University, 601 Jinsui Road, Xinxiang, Henan, 453003, China.
| |
Collapse
|
5
|
Alexis NE, Zhou LY, Burbank AJ, Almond M, Hernandez ML, Mills KH, Noah TL, Wells H, Zhou H, Peden DB. Development of a screening protocol to identify persons who are responsive to wood smoke particle-induced airway inflammation with pilot assessment of GSTM1 genotype and asthma status as response modifiers. Inhal Toxicol 2022; 34:329-339. [PMID: 35968917 PMCID: PMC10519374 DOI: 10.1080/08958378.2022.2110334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 07/28/2022] [Indexed: 11/05/2022]
Abstract
BACKGROUND We are currently screening human volunteers to determine their sputum polymorphonuclear neutrophil (PMN) response 6- and 24-hours following initiation of exposure to wood smoke particles (WSP). Inflammatory responders (≥10% increase in %PMN) are identified for their subsequent participation in mitigation studies against WSP-induced airways inflammation. In this report we compared responder status (<i>N</i> = 52) at both 6 and 24 hr time points to refine/expand its classification, assessed the impact of the GSTM1 genotype, asthma status and sex on responder status, and explored whether sputum soluble phase markers of inflammation correlate with PMN responsiveness to WSP. RESULTS Six-hour responders tended to be 24-hour responders and vice versa, but 24-hour responders also had significantly increased IL-1beta, IL-6, IL-8 at 24 hours post WSP exposure. The GSTM1 null genotype significantly (<i>p</i> < 0.05) enhanced the %PMN response by 24% in the 24-hour responders and not at all in the 6 hours responders. Asthma status enhanced the 24 hour %PMN response in the 6- and 24-hour responders. In the entire cohort (not stratified by responder status), we found a significant, but very small decrease in FVC and systolic blood pressure immediately following WSP exposure and sputum %PMNs were significantly increased and associated with sputum inflammatory markers (IL-1beta, IL-6, IL-8, and PMN/mg) at 24 but not 6 hours post exposure. Blood endpoints in the entire cohort showed a significant increase in %PMN and PMN/mg at 6 but not 24 hours. Sex had no effect on %PMN response. CONCLUSIONS The 24-hour time point was more informative than the 6-hour time point in optimally and expansively defining airway inflammatory responsiveness to WSP exposure. GSTM1 and asthma status are significant effect modifiers of this response. These study design and subject parameters should be considered before enrolling volunteers for proof-of-concept WSP mitigation studies.
Collapse
Affiliation(s)
- Neil E Alexis
- Center for Environmental Medicine, Asthma and Lung Biology, School of Medicine, University of North Carolina, Chapel Hill, NC, USA
- Division of Allergy & Immunology, Department of Pediatrics, School of Medicine, University of North Carolina, Chapel Hill, NC, USA
| | - Laura Y Zhou
- Center for Environmental Medicine, Asthma and Lung Biology, School of Medicine, University of North Carolina, Chapel Hill, NC, USA
- Department of Biostatistics, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC, USA
| | - Allison J Burbank
- Division of Allergy & Immunology, Department of Pediatrics, School of Medicine, University of North Carolina, Chapel Hill, NC, USA
- Children's Research Institute, Department of Pediatrics, School of Medicine, University of North Carolina, Chapel Hill, NC, USA
| | - Martha Almond
- Center for Environmental Medicine, Asthma and Lung Biology, School of Medicine, University of North Carolina, Chapel Hill, NC, USA
| | - Michelle L Hernandez
- Division of Allergy & Immunology, Department of Pediatrics, School of Medicine, University of North Carolina, Chapel Hill, NC, USA
- Children's Research Institute, Department of Pediatrics, School of Medicine, University of North Carolina, Chapel Hill, NC, USA
| | - Katherine H Mills
- Division of Allergy & Immunology, Department of Pediatrics, School of Medicine, University of North Carolina, Chapel Hill, NC, USA
| | - Terry L Noah
- Center for Environmental Medicine, Asthma and Lung Biology, School of Medicine, University of North Carolina, Chapel Hill, NC, USA
- Division of Pulmonology, Department of Pediatrics, School of Medicine, University of North Carolina, Chapel Hill, NC, USA
| | - Heather Wells
- Center for Environmental Medicine, Asthma and Lung Biology, School of Medicine, University of North Carolina, Chapel Hill, NC, USA
| | - Haibo Zhou
- Center for Environmental Medicine, Asthma and Lung Biology, School of Medicine, University of North Carolina, Chapel Hill, NC, USA
- Children's Research Institute, Department of Pediatrics, School of Medicine, University of North Carolina, Chapel Hill, NC, USA
| | - David B Peden
- Center for Environmental Medicine, Asthma and Lung Biology, School of Medicine, University of North Carolina, Chapel Hill, NC, USA
- Division of Allergy & Immunology, Department of Pediatrics, School of Medicine, University of North Carolina, Chapel Hill, NC, USA
| |
Collapse
|
6
|
Genetic Variants Associated with Chronic Obstructive Pulmonary Disease Risk: Cumulative Epidemiological Evidence from Meta-Analyses and Genome-Wide Association Studies. Can Respir J 2022; 2022:3982335. [PMID: 35721789 PMCID: PMC9203202 DOI: 10.1155/2022/3982335] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 05/26/2022] [Indexed: 12/03/2022] Open
Abstract
Background Last two decades, many association studies on genetic variants and chronic obstructive pulmonary disease (COPD) risk have been published. But results from different studies are inconsistent. Therefore, we performed this article to systematically evaluate results from previous meta-analyses and genome-wide association studies (GWASs). Material and Methods. Firstly, we retrieved meta-analyses in PubMed, Embase, and China National Knowledge Infrastructure and GWASs in PubMed and GWAS catalog on or before April 7th, 2022. Then, data were extracted and screened. Finally, two main methods—Venice criteria and false-positive report probability test—were used to evaluate significant associations. Results As a result, eighty-eight meta-analyses and 5 GWASs were deemed eligible for inclusion. Fifty variants in 26 genes obtained from meta-analyses were significantly associated with COPD risk. Cumulative epidemiological evidence of an association was graded as strong for 10 variants in 8 genes (GSTM1, CHRNA, ADAM33, SP-D, TNF-α, VDBP, HMOX1, and HHIP), moderate for 6 variants in 5 genes (PI, GSTM1, ADAM33, TNF-α, and VDBP), and weak for 40 variants in 23 genes. Five variants in 4 genes showed convincing evidence of no association with COPD risk in meta-analyses. Additionally, 29 SNPs identified in GWASs were proved to be noteworthy based on the FPRP test. Conclusion In summary, more than half (52.38%) of genetic variants reported in previous meta-analyses showed no association with COPD risk. However, 13 variants in 9 genes had moderate to strong evidence for an association. This article can serve as a useful reference for further studies.
Collapse
|
7
|
Antioxidant Genetic Profile Modifies Probability of Developing Neurological Sequelae in Long-COVID. Antioxidants (Basel) 2022; 11:antiox11050954. [PMID: 35624818 PMCID: PMC9138155 DOI: 10.3390/antiox11050954] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 05/06/2022] [Accepted: 05/08/2022] [Indexed: 02/06/2023] Open
Abstract
Understanding the sequelae of COVID-19 is of utmost importance. Neuroinflammation and disturbed redox homeostasis are suggested as prevailing underlying mechanisms in neurological sequelae propagation in long-COVID. We aimed to investigate whether variations in antioxidant genetic profile might be associated with neurological sequelae in long-COVID. Neurological examination and antioxidant genetic profile (SOD2, GPXs and GSTs) determination, as well as, genotype analysis of Nrf2 and ACE2, were conducted on 167 COVID-19 patients. Polymorphisms were determined by the appropriate PCR methods. Only polymorphisms in GSTP1AB and GSTO1 were independently associated with long-COVID manifestations. Indeed, individuals carrying GSTP1 Val or GSTO1 Asp allele exhibited lower odds of long-COVID myalgia development, both independently and in combination. Furthermore, the combined presence of GSTP1 Ile and GSTO1 Ala alleles exhibited cumulative risk regarding long-COVID myalgia in carriers of the combined GPX1 LeuLeu/GPX3 CC genotype. Moreover, individuals carrying combined GSTM1-null/GPX1LeuLeu genotype were more prone to developing long-COVID “brain fog”, while this probability further enlarged if the Nrf2 A allele was also present. The fact that certain genetic variants of antioxidant enzymes, independently or in combination, affect the probability of long-COVID manifestations, further emphasizes the involvement of genetic susceptibility when SARS-CoV-2 infection is initiated in the host cells, and also months after.
Collapse
|
8
|
Long E, Carlsten C. Controlled human exposure to diesel exhaust: results illuminate health effects of traffic-related air pollution and inform future directions. Part Fibre Toxicol 2022; 19:11. [PMID: 35139881 PMCID: PMC8827176 DOI: 10.1186/s12989-022-00450-5] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Accepted: 01/31/2022] [Indexed: 12/03/2022] Open
Abstract
Air pollution is an issue of increasing interest due to its globally relevant impacts on morbidity and mortality. Controlled human exposure (CHE) studies are often employed to investigate the impacts of pollution on human health, with diesel exhaust (DE) commonly used as a surrogate of traffic related air pollution (TRAP). This paper will review the results derived from 104 publications of CHE to DE (CHE-DE) with respect to health outcomes. CHE-DE studies have provided mechanistic evidence supporting TRAP’s detrimental effects on related to the cardiovascular system (e.g., vasomotor dysfunction, inhibition of fibrinolysis, and impaired cardiac function) and respiratory system (e.g., airway inflammation, increased airway responsiveness, and clinical symptoms of asthma). Oxidative stress is thought to be the primary mechanism of TRAP-induced effects and has been supported by several CHE-DE studies. A historical limitation of some air pollution research is consideration of TRAP (or its components) in isolation, limiting insight into the interactions between TRAP and other environmental factors often encountered in tandem. CHE-DE studies can help to shed light on complex conditions, and several have included co-exposure to common elements such as allergens, ozone, and activity level. The ability of filters to mitigate the adverse effects of DE, by limiting exposure to the particulate fraction of polluted aerosols, has also been examined. While various biomarkers of DE exposure have been evaluated in CHE-DE studies, a definitive such endpoint has yet to be identified. In spite of the above advantages, this paradigm for TRAP is constrained to acute exposures and can only be indirectly applied to chronic exposures, despite the critical real-world impact of living long-term with TRAP. Those with significant medical conditions are often excluded from CHE-DE studies and so results derived from healthy individuals may not apply to more susceptible populations whose further study is needed to avoid potentially misleading conclusions. In spite of limitations, the contributions of CHE-DE studies have greatly advanced current understanding of the health impacts associated with TRAP exposure, especially regarding mechanisms therein, with important implications for regulation and policy.
Collapse
Affiliation(s)
- Erin Long
- Faculty of Medicine, University of British Columbia, 317 - 2194 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada
| | - Christopher Carlsten
- Division of Respiratory Medicine, Department of Medicine, University of British Columbia, 2775 Laurel Street 7th Floor, Vancouver, BC, V5Z 1M9, Canada.
| |
Collapse
|
9
|
Coric V, Milosevic I, Djukic T, Bukumiric Z, Savic-Radojevic A, Matic M, Jerotic D, Todorovic N, Asanin M, Ercegovac M, Ranin J, Stevanovic G, Pljesa-Ercegovac M, Simic T. GSTP1 and GSTM3 Variant Alleles Affect Susceptibility and Severity of COVID-19. Front Mol Biosci 2021; 8:747493. [PMID: 34988113 PMCID: PMC8721193 DOI: 10.3389/fmolb.2021.747493] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 11/09/2021] [Indexed: 12/20/2022] Open
Abstract
Based on the premise that oxidative stress plays an important role in severe acute respiratory syndrome coronavirus (SARS-CoV-2) infection, we speculated that variations in the antioxidant activities of different members of the glutathione S-transferase family of enzymes might modulate individual susceptibility towards development of clinical manifestations in COVID-19. The distribution of polymorphisms in cytosolic glutathione S-transferases GSTA1, GSTM1, GSTM3, GSTP1 (rs1695 and rs1138272), and GSTT1 were assessed in 207 COVID-19 patients and 252 matched healthy individuals, emphasizing their individual and cumulative effect in disease development and severity. GST polymorphisms were determined by appropriate PCR methods. Among six GST polymorphisms analyzed in this study, GSTP1 rs1695 and GSTM3 were found to be associated with COVID-19. Indeed, the data obtained showed that individuals carrying variant GSTP1-Val allele exhibit lower odds of COVID-19 development (p = 0.002), contrary to carriers of variant GSTM3-CC genotype which have higher odds for COVID-19 (p = 0.024). Moreover, combined GSTP1 (rs1138272 and rs1695) and GSTM3 genotype exhibited cumulative risk regarding both COVID-19 occurrence and COVID-19 severity (p = 0.001 and p = 0.025, respectively). Further studies are needed to clarify the exact roles of specific glutathione S-transferases once the SARS-CoV-2 infection is initiated in the host cell.
Collapse
Affiliation(s)
- Vesna Coric
- Faculty of Medicine, University of Belgrade, Belgrade, Serbia
- Institute of Medical and Clinical Biochemistry, Belgrade, Serbia
| | - Ivana Milosevic
- Faculty of Medicine, University of Belgrade, Belgrade, Serbia
- Clinic of Infectious and Tropical Diseases, Clinical Centre of Serbia, Belgrade, Serbia
| | - Tatjana Djukic
- Faculty of Medicine, University of Belgrade, Belgrade, Serbia
- Institute of Medical and Clinical Biochemistry, Belgrade, Serbia
| | - Zoran Bukumiric
- Faculty of Medicine, University of Belgrade, Belgrade, Serbia
- Institute of Medical Statistics and Informatics, Belgrade, Serbia
| | - Ana Savic-Radojevic
- Faculty of Medicine, University of Belgrade, Belgrade, Serbia
- Institute of Medical and Clinical Biochemistry, Belgrade, Serbia
| | - Marija Matic
- Faculty of Medicine, University of Belgrade, Belgrade, Serbia
- Institute of Medical and Clinical Biochemistry, Belgrade, Serbia
| | - Djurdja Jerotic
- Faculty of Medicine, University of Belgrade, Belgrade, Serbia
- Institute of Medical and Clinical Biochemistry, Belgrade, Serbia
| | - Nevena Todorovic
- Clinic of Infectious and Tropical Diseases, Clinical Centre of Serbia, Belgrade, Serbia
| | - Milika Asanin
- Faculty of Medicine, University of Belgrade, Belgrade, Serbia
- Clinic of Neurology, Clinical Centre of Serbia, Belgrade, Serbia
| | - Marko Ercegovac
- Faculty of Medicine, University of Belgrade, Belgrade, Serbia
- Clinic of Cardiology, Clinical Centre of Serbia, Belgrade, Serbia
| | - Jovan Ranin
- Faculty of Medicine, University of Belgrade, Belgrade, Serbia
- Clinic of Infectious and Tropical Diseases, Clinical Centre of Serbia, Belgrade, Serbia
| | - Goran Stevanovic
- Faculty of Medicine, University of Belgrade, Belgrade, Serbia
- Clinic of Infectious and Tropical Diseases, Clinical Centre of Serbia, Belgrade, Serbia
| | - Marija Pljesa-Ercegovac
- Faculty of Medicine, University of Belgrade, Belgrade, Serbia
- Institute of Medical and Clinical Biochemistry, Belgrade, Serbia
| | - Tatjana Simic
- Faculty of Medicine, University of Belgrade, Belgrade, Serbia
- Institute of Medical and Clinical Biochemistry, Belgrade, Serbia
- Serbian Academy of Sciences and Arts, Belgrade, Serbia
| |
Collapse
|
10
|
van de Wetering C, Manuel AM, Sharafi M, Aboushousha R, Qian X, Erickson C, MacPherson M, Chan G, Adcock IM, ZounematKermani N, Schleich F, Louis R, Bohrnsen E, D'Alessandro A, Wouters EF, Reynaert NL, Li J, Wolf CR, Henderson CJ, Lundblad LKA, Poynter ME, Dixon AE, Irvin CG, van der Vliet A, van der Velden JL, Janssen-Heininger YM. Glutathione-S-transferase P promotes glycolysis in asthma in association with oxidation of pyruvate kinase M2. Redox Biol 2021; 47:102160. [PMID: 34624602 PMCID: PMC8502950 DOI: 10.1016/j.redox.2021.102160] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 10/02/2021] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Interleukin-1-dependent increases in glycolysis promote allergic airways disease in mice and disruption of pyruvate kinase M2 (PKM2) activity is critical herein. Glutathione-S-transferase P (GSTP) has been implicated in asthma pathogenesis and regulates the oxidation state of proteins via S-glutathionylation. We addressed whether GSTP-dependent S-glutathionylation promotes allergic airways disease by promoting glycolytic reprogramming and whether it involves the disruption of PKM2. METHODS We used house dust mite (HDM) or interleukin-1β in C57BL6/NJ WT or mice that lack GSTP. Airway basal cells were stimulated with interleukin-1β and the selective GSTP inhibitor, TLK199. GSTP and PKM2 were evaluated in sputum samples of asthmatics and healthy controls and incorporated analysis of the U-BIOPRED severe asthma cohort database. RESULTS Ablation of Gstp decreased total S-glutathionylation and attenuated HDM-induced allergic airways disease and interleukin-1β-mediated inflammation. Gstp deletion or inhibition by TLK199 decreased the interleukin-1β-stimulated secretion of pro-inflammatory mediators and lactate by epithelial cells. 13C-glucose metabolomics showed decreased glycolysis flux at the pyruvate kinase step in response to TLK199. GSTP and PKM2 levels were increased in BAL of HDM-exposed mice as well as in sputum of asthmatics compared to controls. Sputum proteomics and transcriptomics revealed strong correlations between GSTP, PKM2, and the glycolysis pathway in asthma. CONCLUSIONS GSTP contributes to the pathogenesis of allergic airways disease in association with enhanced glycolysis and oxidative disruption of PKM2. Our findings also suggest a PKM2-GSTP-glycolysis signature in asthma that is associated with severe disease.
Collapse
Affiliation(s)
- Cheryl van de Wetering
- Department of Pathology and Laboratory Medicine, University of Vermont, Burlington, VT, USA; Department of Respiratory Medicine, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Allison M Manuel
- Department of Pathology and Laboratory Medicine, University of Vermont, Burlington, VT, USA
| | - Mona Sharafi
- Department of Chemistry, University of Vermont, Burlington, VT, USA
| | - Reem Aboushousha
- Department of Pathology and Laboratory Medicine, University of Vermont, Burlington, VT, USA
| | - Xi Qian
- Department of Pathology and Laboratory Medicine, University of Vermont, Burlington, VT, USA
| | - Cuixia Erickson
- Department of Pathology and Laboratory Medicine, University of Vermont, Burlington, VT, USA
| | - Maximilian MacPherson
- Department of Pathology and Laboratory Medicine, University of Vermont, Burlington, VT, USA
| | - Garrett Chan
- Department of Pathology and Laboratory Medicine, University of Vermont, Burlington, VT, USA
| | - Ian M Adcock
- National Heart & Lung Institute & Data Science Institute, Imperial College London, UK
| | | | - Florence Schleich
- Department of Respiratory Medicine, CHU Sart-TilmanB35, Liege, Belgium
| | - Renaud Louis
- Department of Respiratory Medicine, CHU Sart-TilmanB35, Liege, Belgium
| | - Eric Bohrnsen
- Department of Biochemistry and Molecular Genetics, University of Colorado, Anschutz Medical Campus, Aurora, CO, United States
| | - Angelo D'Alessandro
- Department of Biochemistry and Molecular Genetics, University of Colorado, Anschutz Medical Campus, Aurora, CO, United States
| | - Emiel F Wouters
- Department of Respiratory Medicine, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, Maastricht, the Netherlands; Ludwig Boltzmann Institute for Lung Health, Vienna, Austria
| | - Niki L Reynaert
- Department of Respiratory Medicine, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Jianing Li
- Department of Chemistry, University of Vermont, Burlington, VT, USA
| | - C Roland Wolf
- Systems Medicine, School of Medicine, University of Dundee, Jacqui Wood Cancer Center, Ninewells Hospital Dundee DD1 9SY, Scotland, United Kingdom
| | - Colin J Henderson
- Systems Medicine, School of Medicine, University of Dundee, Jacqui Wood Cancer Center, Ninewells Hospital Dundee DD1 9SY, Scotland, United Kingdom
| | - Lennart K A Lundblad
- Meakins-Christie Laboratories, McGill University & THORASYS Thoracic Medical Systems Inc., Montréal, QC, Canada
| | - Matthew E Poynter
- Department of Medicine, College of Medicine, University of Vermont, Burlington, VT, USA
| | - Anne E Dixon
- Department of Medicine, College of Medicine, University of Vermont, Burlington, VT, USA
| | - Charles G Irvin
- Department of Medicine, College of Medicine, University of Vermont, Burlington, VT, USA
| | - Albert van der Vliet
- Department of Pathology and Laboratory Medicine, University of Vermont, Burlington, VT, USA
| | - Jos L van der Velden
- Department of Pathology and Laboratory Medicine, University of Vermont, Burlington, VT, USA
| | | |
Collapse
|
11
|
Taniguchi A, Tsuge M, Miyahara N, Tsukahara H. Reactive Oxygen Species and Antioxidative Defense in Chronic Obstructive Pulmonary Disease. Antioxidants (Basel) 2021; 10:antiox10101537. [PMID: 34679673 PMCID: PMC8533053 DOI: 10.3390/antiox10101537] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 09/23/2021] [Accepted: 09/24/2021] [Indexed: 02/06/2023] Open
Abstract
The respiratory system is continuously exposed to endogenous and exogenous oxidants. Chronic obstructive pulmonary disease (COPD) is characterized by chronic inflammation of the airways, leading to the destruction of lung parenchyma (emphysema) and declining pulmonary function. It is increasingly obvious that reactive oxygen species (ROS) and reactive nitrogen species (RNS) contribute to the progression and amplification of the inflammatory responses related to this disease. First, we described the association between cigarette smoking, the most representative exogenous oxidant, and COPD and then presented the multiple pathophysiological aspects of ROS and antioxidative defense systems in the development and progression of COPD. Second, the relationship between nitric oxide system (endothelial) dysfunction and oxidative stress has been discussed. Third, we have provided data on the use of these biomarkers in the pathogenetic mechanisms involved in COPD and its progression and presented an overview of oxidative stress biomarkers having clinical applications in respiratory medicine, including those in exhaled breath, as per recent observations. Finally, we explained the findings of recent clinical and experimental studies evaluating the efficacy of antioxidative interventions for COPD. Future breakthroughs in antioxidative therapy may provide a promising therapeutic strategy for the prevention and treatment of COPD.
Collapse
Affiliation(s)
- Akihiko Taniguchi
- Department of Hematology, Oncology, Allergy and Respiratory Medicine, Okayama University Academic Field of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama 700-8558, Japan;
| | - Mitsuru Tsuge
- Department of Pediatrics, Okayama University Academic Field of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama 700-8558, Japan;
| | - Nobuaki Miyahara
- Department of Medical Technology, Okayama University Academic Field of Health Sciences, Okayama 700-8558, Japan;
| | - Hirokazu Tsukahara
- Department of Pediatrics, Okayama University Academic Field of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama 700-8558, Japan;
- Correspondence:
| |
Collapse
|
12
|
Sun B, Song J, Wang Y, Jiang J, An Z, Li J, Zhang Y, Wang G, Li H, Alexis NE, Jaspers I, Wu W. Associations of short-term PM 2.5 exposures with nasal oxidative stress, inflammation and lung function impairment and modification by GSTT1-null genotype: A panel study of the retired adults. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2021; 285:117215. [PMID: 33932759 DOI: 10.1016/j.envpol.2021.117215] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 04/16/2021] [Accepted: 04/20/2021] [Indexed: 06/12/2023]
Abstract
PM2.5 (particulate matter ≤2.5 μm in aerodynamic diameter) is a major urban air pollutant worldwide. Its effects on the respiratory system of the susceptible population have been less characterized. This study aimed to estimate the association of short-term PM2.5 exposure with respiratory outcomes of the retired adults, and to examine whether these associations were stronger among the subjects with GSTT-null genotype. 32 healthy subjects (55-77 years) were recruited for five follow-up examinations. Ambient concentrations of PM2.5 were monitored consecutively for 7 days prior to physical examination. Pulmonary outcomes including forced vital capacity (FVC), forced expiratory volume in 1 s (FEV1), peak expiratory flow (PEF), and fractional exhaled nitric oxide (FeNO), and nasal fluid concentrations of 8-epi-prostaglandin F2 alpha (8-epi-PGF2α), tumor necrosis factor-α (TNF-α), interleukin-8 (IL-8) and IL-1β were measured. A linear mixed-effect model was introduced to evaluate the associations of PM2.5 concentrations with respiratory outcomes. Additionally, GSTT1 genotype-based stratification was performed to characterize modification on PM2.5-related respiratory outcomes. We found that a 10 μg/m3 increase in PM2.5 was associated with decreases of 0.52 L (95% confidence interval [CI]: -1.04, -0.002), 0.64 L (95% CI: -1.13, -0.16), 0.1 (95% CI: -0.23, 0.04) and 2.87 L/s (95% CI: -5.09, -0.64) in FVC, FEV1, FEV1/FVC ratio and PEF at lag 2, respectively. Meanwhile, marked increases of 80.82% (95% CI: 5.13%, 156.50%) in IL-8, 77.14% (95% CI: 1.88%, 152.40%) in IL-1β and 67.87% (95% CI: 14.85%, 120.88%) in 8-epi-PGF2α were observed as PM2.5 concentration increased by 10 μg/m3 at lag 2. Notably, PM2.5-associated decreases in FVC and PEF and increase in FeNO were stronger among the subjects with GSTT1-null genotype. In summary, short-term exposure to PM2.5 is associated with nasal inflammation, oxidative stress and lung function reduction in the retired subjects. Lung function reduction and inflammation are stronger among the subjects with GSTT1-null genotype.
Collapse
Affiliation(s)
- Beibei Sun
- School of Public Health, Xinxiang Medical University, Xinxiang, Henan Province, 453003, China
| | - Jie Song
- School of Public Health, Xinxiang Medical University, Xinxiang, Henan Province, 453003, China
| | - Ya Wang
- School of Public Health, Xinxiang Medical University, Xinxiang, Henan Province, 453003, China
| | - Jing Jiang
- School of Public Health, Xinxiang Medical University, Xinxiang, Henan Province, 453003, China
| | - Zhen An
- School of Public Health, Xinxiang Medical University, Xinxiang, Henan Province, 453003, China
| | - Juan Li
- School of Public Health, Xinxiang Medical University, Xinxiang, Henan Province, 453003, China
| | - Yange Zhang
- School of Public Health, Xinxiang Medical University, Xinxiang, Henan Province, 453003, China
| | - Gui Wang
- School of Public Health, Xinxiang Medical University, Xinxiang, Henan Province, 453003, China
| | - Huijun Li
- School of Public Health, Xinxiang Medical University, Xinxiang, Henan Province, 453003, China
| | - Neil E Alexis
- Center for Environmental Medicine, Asthma, and Lung Biology, University of North Carolina at Chapel Hill, NC, 27599, United States
| | - Ilona Jaspers
- Center for Environmental Medicine, Asthma, and Lung Biology, University of North Carolina at Chapel Hill, NC, 27599, United States
| | - Weidong Wu
- School of Public Health, Xinxiang Medical University, Xinxiang, Henan Province, 453003, China.
| |
Collapse
|
13
|
van de Wetering C, Elko E, Berg M, Schiffers CHJ, Stylianidis V, van den Berge M, Nawijn MC, Wouters EFM, Janssen-Heininger YMW, Reynaert NL. Glutathione S-transferases and their implications in the lung diseases asthma and chronic obstructive pulmonary disease: Early life susceptibility? Redox Biol 2021; 43:101995. [PMID: 33979767 PMCID: PMC8131726 DOI: 10.1016/j.redox.2021.101995] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2020] [Revised: 04/23/2021] [Accepted: 04/24/2021] [Indexed: 01/01/2023] Open
Abstract
Our lungs are exposed daily to airborne pollutants, particulate matter, pathogens as well as lung allergens and irritants. Exposure to these substances can lead to inflammatory responses and may induce endogenous oxidant production, which can cause chronic inflammation, tissue damage and remodeling. Notably, the development of asthma and Chronic Obstructive Pulmonary Disease (COPD) is linked to the aforementioned irritants. Some inhaled foreign chemical compounds are rapidly absorbed and processed by phase I and II enzyme systems critical in the detoxification of xenobiotics including the glutathione-conjugating enzymes Glutathione S-transferases (GSTs). GSTs, and in particular genetic variants of GSTs that alter their activities, have been found to be implicated in the susceptibility to and progression of these lung diseases. Beyond their roles in phase II metabolism, evidence suggests that GSTs are also important mediators of normal lung growth. Therefore, the contribution of GSTs to the development of lung diseases in adults may already start in utero, and continues through infancy, childhood, and adult life. GSTs are also known to scavenge oxidants and affect signaling pathways by protein-protein interaction. Moreover, GSTs regulate reversible oxidative post-translational modifications of proteins, known as protein S-glutathionylation. Therefore, GSTs display an array of functions that impact the pathogenesis of asthma and COPD. In this review we will provide an overview of the specific functions of each class of mammalian cytosolic GSTs. This is followed by a comprehensive analysis of their expression profiles in the lung in healthy subjects, as well as alterations that have been described in (epithelial cells of) asthmatics and COPD patients. Particular emphasis is placed on the emerging evidence of the regulatory properties of GSTs beyond detoxification and their contribution to (un)healthy lungs throughout life. By providing a more thorough understanding, tailored therapeutic strategies can be designed to affect specific functions of particular GSTs.
Collapse
Affiliation(s)
- Cheryl van de Wetering
- Department of Respiratory Medicine, Maastricht University Medical Center+, Maastricht, the Netherlands; Department of Pathology and Laboratory Medicine, University of Vermont College of Medicine, Burlington, VT, USA
| | - Evan Elko
- Department of Pathology and Laboratory Medicine, University of Vermont College of Medicine, Burlington, VT, USA
| | - Marijn Berg
- Pathology and Medical Biology, GRIAC Research Institute, University of Groningen, University Medical Center Groningen (UMCG), Groningen, the Netherlands
| | - Caspar H J Schiffers
- Department of Respiratory Medicine, Maastricht University Medical Center+, Maastricht, the Netherlands; Department of Pathology and Laboratory Medicine, University of Vermont College of Medicine, Burlington, VT, USA
| | - Vasili Stylianidis
- Department of Respiratory Medicine, Maastricht University Medical Center+, Maastricht, the Netherlands
| | - Maarten van den Berge
- Pulmonology, GRIAC Research Institute, University of Groningen, University Medical Center Groningen (UMCG), Groningen, the Netherlands
| | - Martijn C Nawijn
- Pathology and Medical Biology, GRIAC Research Institute, University of Groningen, University Medical Center Groningen (UMCG), Groningen, the Netherlands
| | - Emiel F M Wouters
- Department of Respiratory Medicine, Maastricht University Medical Center+, Maastricht, the Netherlands; Ludwig Boltzmann Institute for Lung Health, Vienna, Austria
| | - Yvonne M W Janssen-Heininger
- Department of Pathology and Laboratory Medicine, University of Vermont College of Medicine, Burlington, VT, USA.
| | - Niki L Reynaert
- Department of Respiratory Medicine, Maastricht University Medical Center+, Maastricht, the Netherlands.
| |
Collapse
|
14
|
Orach J, Rider CF, Carlsten C. Concentration-dependent health effects of air pollution in controlled human exposures. ENVIRONMENT INTERNATIONAL 2021; 150:106424. [PMID: 33596522 DOI: 10.1016/j.envint.2021.106424] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 01/21/2021] [Accepted: 01/26/2021] [Indexed: 06/12/2023]
Abstract
BACKGROUND Air pollution is a leading contributor to premature mortality worldwide and is often represented by particulate matter (PM), a key contributor to its harmful health effects. Concentration-response relationships are useful for quantifying the effects of air pollution in relevant populations and in considering potential effect thresholds. Controlled human exposures can provide data on acute effects and concentration-response relationships that complement epidemiological studies. OBJECTIVES We examined PM concentration-responses after controlled human air pollution exposures to examine exposure-response markers, assess effect modifiers, and identify potential effect thresholds. METHODS We reviewed primary research from published controlled human exposure studies where responses were reported at multiple target PM concentrations or summarized per unit change in PM to identify concentration-dependent effects. RESULTS Of the 191 publications identified through PubMed and supplementary searches, 31 were eligible. Eligible studies collectively represented four pollutant models: concentrated ambient particles, engineered carbon nanoparticles, diesel exhaust, and woodsmoke. We identified concentration-dependent effects on oxidative stress markers, inflammation, and cardiovascular function that overlapped across different pollutants. Metabolic syndrome and glutathione s-transferase mu 1 genotype were identified as potential effect modifiers. DISCUSSION Improved understanding of concentration-response relationships is integral to biomonitoring and mitigation of health effects through impact assessment and policy. Although we identified potential concentration-response markers, thresholds, and modifiers, our conclusions on these relationships were limited by a dearth of eligible publications, considerable variability in methodology, and inconsistent reporting standards between studies. More research is required to validate these observations. We recommend that future studies harmonize estimate reporting to facilitate the identification of robust response markers across research and applied settings.
Collapse
Affiliation(s)
- Juma Orach
- Air Pollution Exposure Laboratory, Division of Respiratory Medicine, Department of Medicine, Vancouver Coastal Health Research Institute, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Christopher F Rider
- Air Pollution Exposure Laboratory, Division of Respiratory Medicine, Department of Medicine, Vancouver Coastal Health Research Institute, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Christopher Carlsten
- Air Pollution Exposure Laboratory, Division of Respiratory Medicine, Department of Medicine, Vancouver Coastal Health Research Institute, The University of British Columbia, Vancouver, British Columbia, Canada.
| |
Collapse
|
15
|
Delmond KA, Delleon H, Goveia RM, Teixeira TM, Abreu DC, Mello-Andrade F, Reis AADS, Silva DDME, Barbosa ADP, Tavares RS, Anunciação CE, Silveira-Lacerda E. Influence of genetic polymorphisms in glutathione-S-transferases gene in response to imatinib among Brazilian patients with chronic myeloid leukemia. Mol Biol Rep 2021; 48:2035-2046. [PMID: 33709282 DOI: 10.1007/s11033-020-06093-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2020] [Accepted: 12/15/2020] [Indexed: 11/26/2022]
Abstract
Polymorphism in metabolizing enzymes can influence drug response as well as the risk for adverse drug reactions. Nevertheless, there are still few studies analyzing the consequence of polymorphisms for the Glutathione-S-transferases (GST) gene to drug response in chronic myeloid leukemia (CML). This study reports, the influence of GSTP1*B and GSTT1/GSTM1null polymorphisms in response to imatinib in CML patients in a Brazilian population. One hundred thirty-nine CML patients from the Clinical Hospital of Goiânia, Goiás, Brazil, treated with imatinib were enrolled in this study. Genotyping of GSTT1 and GSTM1 genes deletions were performed by qPCR and of GSTP1 gene was performed by RFLP-PCR. The frequency of GSTP1*1B, GSTT1 and GSTM1null polymorphisms were determined for all patients. The influence of each patient's genotypes was analyzed with the patient's response to imatinib treatment. Brazilian CML patients revealed GSTT1 and GSTM1 genes deletions. GSTT1 deletion was found in 19.3% of patients and GSTM1 deletion in 48.7% of patients with CML. GSTT1/GSTM1 deletion was found in 11.7% in Brazilian CML patients. The "G allele" of GSTP1*B, is associated with later cytogenetic response in imatinib therapy. While, the gene presence combined with GG genotype (GSTM1 present/GSTPI-GG) conferred a tend to a later cytogenetic response to patients. GSTP1*B and GSTT1/GSTM1null polymorphisms influence treatment response in CML. Brazilian CML patients presenting GSTP1 AA/AG genotypes alone and in combination with GSTT1 null reach the cytogenetic response faster, while patients presenting GSTP1-GG and GSTMI positive genotypes may take longer to achieve cytogenetic response. As a result, it allows a better prognosis, with the use of an alternative therapy, other than reducing treatment cost.
Collapse
Affiliation(s)
- Kezia Aguiar Delmond
- Department of Genetics, Laboratory of Molecular Genetics and Cytogenetics, Institute of Biological Sciences, Federal University of Goiás, Avenida Esperança, s/n, Campus Samambaia (Campus II), Cx. Postal 131, Goiânia, Goiás, 74690-900, Brazil
- College of Goyazes Union, Trindade, Goiás, 75380-000, Brazil
| | - Hugo Delleon
- Department of Genetics, Laboratory of Molecular Genetics and Cytogenetics, Institute of Biological Sciences, Federal University of Goiás, Avenida Esperança, s/n, Campus Samambaia (Campus II), Cx. Postal 131, Goiânia, Goiás, 74690-900, Brazil
- Uni-Anhanguera University Center of Goias, Goiânia, Goiás, 74423-115, Brazil
| | - Rebeca Mota Goveia
- Department of Genetics, Laboratory of Molecular Genetics and Cytogenetics, Institute of Biological Sciences, Federal University of Goiás, Avenida Esperança, s/n, Campus Samambaia (Campus II), Cx. Postal 131, Goiânia, Goiás, 74690-900, Brazil
| | - Thallita Monteiro Teixeira
- Department of Genetics, Laboratory of Molecular Genetics and Cytogenetics, Institute of Biological Sciences, Federal University of Goiás, Avenida Esperança, s/n, Campus Samambaia (Campus II), Cx. Postal 131, Goiânia, Goiás, 74690-900, Brazil
| | - Davi Carvalho Abreu
- Department of Genetics, Laboratory of Molecular Genetics and Cytogenetics, Institute of Biological Sciences, Federal University of Goiás, Avenida Esperança, s/n, Campus Samambaia (Campus II), Cx. Postal 131, Goiânia, Goiás, 74690-900, Brazil
| | - Francyelli Mello-Andrade
- Department of Genetics, Laboratory of Molecular Genetics and Cytogenetics, Institute of Biological Sciences, Federal University of Goiás, Avenida Esperança, s/n, Campus Samambaia (Campus II), Cx. Postal 131, Goiânia, Goiás, 74690-900, Brazil
- Department of Chemistry, Federal Institute of Education, Science and Technology of Goiás, Goiânia, Goiás, 74055-110, Brazil
| | - Angela Adamski da Silva Reis
- Department of Biochemistry and Molecular Biology, Institute of Biological Sciences, Federal University of Goiás, Goiânia, Goiás, 74690-900, Brazil
| | - Daniela de Melo E Silva
- Department of Genetics, Institute of Biological Sciences, Federal University of Goiás, Goiânia, Goiás, 74690-900, Brazil
| | | | | | - Carlos Eduardo Anunciação
- Department of Biochemistry and Molecular Biology, Institute of Biological Sciences, Federal University of Goiás, Goiânia, Goiás, 74690-900, Brazil
| | - Elisângela Silveira-Lacerda
- Department of Genetics, Laboratory of Molecular Genetics and Cytogenetics, Institute of Biological Sciences, Federal University of Goiás, Avenida Esperança, s/n, Campus Samambaia (Campus II), Cx. Postal 131, Goiânia, Goiás, 74690-900, Brazil.
| |
Collapse
|
16
|
Cosselman KE, Allen J, Jansen KL, Stapleton P, Trenga CA, Larson TV, Kaufman JD. Acute exposure to traffic-related air pollution alters antioxidant status in healthy adults. ENVIRONMENTAL RESEARCH 2020; 191:110027. [PMID: 32810504 PMCID: PMC8568481 DOI: 10.1016/j.envres.2020.110027] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 07/20/2020] [Accepted: 07/30/2020] [Indexed: 05/27/2023]
Abstract
BACKGROUND Exposure to traffic-related air pollution is associated with an increased risk of cardiovascular and respiratory disease. Evidence suggests that inhaled pollutants precipitate these effects via multiple pathways involving oxidative stress. OBJECTIVE Postulating that a decrease in circulating antioxidant levels reflect an oxidative response, we investigated the effect of inhaled diesel exhaust (DE) on the ratio of reduced to oxidized glutathione (GSH/GSSG) in healthy adults, and whether pre-exposure antioxidant supplementation blunted this response. We also examined exposure-related changes in antioxidant/stress response leukocyte gene expression (GCLc, HMOX-1, IL-6, TGFβ) and plasma IL-6 levels. METHODS Nineteen nonsmoking adults participated in a double-blind, randomized, four-way crossover study. Each subject completed 120-min exposures to filtered air and DE (200 μg/m3), with and without antioxidant pretreatment. Antioxidant comprised 1000 mg ascorbate for 7 days and 1200 mg N-acetylcysteine 1 day prior to exposure, with 1000 mg and 600 mg, respectively, administered 2 h prior to exposure. Whole blood glutathione was measured pre- and post-exposure; plasma IL-6 and mRNA expression were quantified pre, during and post exposure. RESULTS Diesel exhaust exposure was associated with significantly decreased GSH/GSSG (p = 0.001) and a 4-fold increase in IL-6 mRNA (p = 0.01) post exposure. Antioxidant pretreatment did not significantly mediate the effect of DE exposure on GSH/GSSG, though appeared to decrease the effect of exposure on IL-6 mRNA expression. CONCLUSIONS Acute DE inhalation induced detectable oxidative effects in healthy adults, which were not significantly attenuated by the selected antioxidant pre-treatment. This finding supports the premise that oxidative stress is one mechanism underlying the adverse effects of traffic-related air pollution.
Collapse
Affiliation(s)
| | - Jason Allen
- Department of Environmental and Occupational Health Sciences, USA
| | - Karen L Jansen
- Department of Environmental and Occupational Health Sciences, USA
| | | | - Carol A Trenga
- Department of Environmental and Occupational Health Sciences, USA
| | - Timothy V Larson
- Department of Environmental and Occupational Health Sciences, USA; Department of Civil and Environmental Engineering, USA
| | - Joel D Kaufman
- Department of Environmental and Occupational Health Sciences, USA; Department of Medicine, USA; Department of Epidemiology, University of Washington, Seattle, WA, USA
| |
Collapse
|
17
|
Wang CM, Chang CB, Lee SP, W-Y Chan M, Wu SF. Differential DNA methylation profiles of peripheral blood mononuclear cells in allergic asthmatic children following dust mite immunotherapy. JOURNAL OF MICROBIOLOGY, IMMUNOLOGY, AND INFECTION = WEI MIAN YU GAN RAN ZA ZHI 2020; 53:986-995. [PMID: 32684341 DOI: 10.1016/j.jmii.2020.06.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Revised: 05/28/2020] [Accepted: 06/11/2020] [Indexed: 12/21/2022]
Abstract
BACKGROUND/PURPOSE Allergen-specific immunotherapy (SIT) is now considered curative to allergic diseases such as asthma. Mechanistically, our previous work showed DNA hypermethylation of cytokine genes, in T-helper cells, in allergic asthmatic children treated with allergen-SIT. In this study, we extended to work to assess possible changes in the DNA methylomes of peripheral blood mononuclear cells (PBMCs), isolated from mite allergen-SIT asthmatic children, to explore further the underlying methylation changes. METHODS Thirteen allergic asthmatic children who received Der p-SIT, 12 non-SIT allergic asthmatic controls, and 12 healthy controls were enrolled. Bisulfite-converted DNA from Der p-stimulated PBMCs was analyzed using Human Methylation 450 k BeadChip. Pyrosequencing and quantitative real-time PCR were used to validate the DNA methylation levels and the gene expression of individual samples. RESULTS We identified 108 significantly differentially methylated regions (DMRs) unique to Der p-treated PBMCs, with 53 probes linked to demethylated DMRs, and 55 probes linked to methylated DMRs. Three associated genes (BCL6, HSPG2, and HSP90AA1), of selected DMRs, were subjected to bisulfite pyrosequencing. Of these, BCL6 showed significant hypomethylation, while HSPG2 and HSP90AA1 were hypermethylated in SIT group, compared to the AA group. Furthermore, SIT group had significantly higher gene expression of BCL6 and lower gene expression of HSPG2. KEGG pathway analysis further revealed DMR genes involved in ECM-receptor interactions, asthma, and antigen processing and presentation pathways. CONCLUSIONS Several DNA regions showed DNA methylation altered by Der p specific immunotherapy, indicating desensitization-associated methylomes. Genes belonging to these SIT-altered pathways may represent therapeutic targets for better clinical management of asthma.
Collapse
Affiliation(s)
- Chuang-Ming Wang
- Department of Pediatrics, Ditmanson Medical Foundation Chia-Yi Christian Hospital, Chia-Yi, Taiwan; Min-Hwei Junior College of Health Care Management, Tainan, Taiwan
| | - Chia-Bin Chang
- Department of Biomedical Sciences and Institute of Molecular Biology, National Chung Cheng University, Chia-Yi, Taiwan
| | - Shiao-Pieng Lee
- Division of Oral and Maxillofacial Surgery, Department of Dentistry, School of Dentistry, Tri-Service General Hospital and National Defense Medical Center, Taipei, Taiwan
| | - Michael W-Y Chan
- Department of Biomedical Sciences and Institute of Molecular Biology, National Chung Cheng University, Chia-Yi, Taiwan
| | - Shu-Fen Wu
- Center for Innovative Research on Aging Society, National Chung Cheng University, Chia-Yi, Taiwan.
| |
Collapse
|
18
|
Song J, Zhu J, Tian G, Li H, Li H, An Z, Jiang J, Fan W, Wang G, Zhang Y, Wu W. Short time exposure to ambient ozone and associated cardiovascular effects: A panel study of healthy young adults. ENVIRONMENT INTERNATIONAL 2020; 137:105579. [PMID: 32086080 DOI: 10.1016/j.envint.2020.105579] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/02/2019] [Revised: 02/11/2020] [Accepted: 02/12/2020] [Indexed: 06/10/2023]
Abstract
The evidence that exposure to ambient ozone (O3) causes acute cardiovascular effects appears inconsistent. A repeated-measure study with 61 healthy young volunteers was conducted in Xinxiang, Central China. Real-time concentrations of O3 were monitored. Cardiovascular outcomes including blood pressure (BP), heart rate (HR), serum levels of high sensitivity C-reactive protein (hs-CRP), 8-hydroxy-2'-deoxyguanosine (8-OHdG), tissue-type plasminogen activator (t-PA), and platelet-monocyte aggregation (PMA) were repeated measured. Linear mixed-effect models were used to analyze the association of ambient O3 with these cardiovascular outcomes. Additionally, the modifying effects of glutathione S-transferase mu 1 (GSTM1) and glutathione S-transferase theta 1 (GSTT1) polymorphisms were estimated to explore the potential mechanisms and role of the association between O3 exposure and the above cardiovascular outcomes. A 10 μg/m3 increase in O3 was associated with increases of 9.2 mmHg (95% confidence interval [CI]: 2.5, 15.9), 7.2 mmHg (95% CI: 0.8, 13.6), and 21.2 bpm (95% CI: 5.8, 36.6) in diastolic BP (DBP, lag1), mean arterial BP (MABP, lag1), and HR (lag01), respectively. Meanwhile, the serum concentrations of hs-CRP, 8-OHdG, and t-PA were all increased by O3 exposure, but the PMA level was decreased. Stratification analyses showed that the estimated effects of O3 on DBP, MABP, and HR in GSTM1-sufficient subjects were significantly higher than in GSTM1-null subjects. Moreover, GSTM1-null genotype enhanced O3-induced increases, albeit insignificant, in levels of serum hs-CRP, 8-OHdG, and t-PA compared with GSTM1-sufficient genotype. Insignificant increases in hs-CRP and t-PA were also detected in GSTT1-null subjects. Taken together, our findings indicate that acute exposure to ambient O3 induces autonomic alterations, systemic inflammation, oxidative stress, and fibrinolysis in healthy young subjects. GSTM1 genotype presents the trend of modifying O3-induced cardiovascular effects.
Collapse
Affiliation(s)
- Jie Song
- Henan International Collaborative Laboratory for Health Effects and Intervention of Air Pollution, School of Public Health, Xinxiang Medical University, Xinxiang, Henan Province 453003, China
| | - Jingfang Zhu
- Henan International Collaborative Laboratory for Health Effects and Intervention of Air Pollution, School of Public Health, Xinxiang Medical University, Xinxiang, Henan Province 453003, China
| | - Ge Tian
- Henan International Collaborative Laboratory for Health Effects and Intervention of Air Pollution, School of Public Health, Xinxiang Medical University, Xinxiang, Henan Province 453003, China
| | - Haibin Li
- Henan International Collaborative Laboratory for Health Effects and Intervention of Air Pollution, School of Public Health, Xinxiang Medical University, Xinxiang, Henan Province 453003, China
| | - Huijun Li
- Henan International Collaborative Laboratory for Health Effects and Intervention of Air Pollution, School of Public Health, Xinxiang Medical University, Xinxiang, Henan Province 453003, China
| | - Zhen An
- Henan International Collaborative Laboratory for Health Effects and Intervention of Air Pollution, School of Public Health, Xinxiang Medical University, Xinxiang, Henan Province 453003, China
| | - Jing Jiang
- Henan International Collaborative Laboratory for Health Effects and Intervention of Air Pollution, School of Public Health, Xinxiang Medical University, Xinxiang, Henan Province 453003, China
| | - Wei Fan
- Henan International Collaborative Laboratory for Health Effects and Intervention of Air Pollution, School of Public Health, Xinxiang Medical University, Xinxiang, Henan Province 453003, China
| | - Gui Wang
- Henan International Collaborative Laboratory for Health Effects and Intervention of Air Pollution, School of Public Health, Xinxiang Medical University, Xinxiang, Henan Province 453003, China
| | - Yange Zhang
- Henan International Collaborative Laboratory for Health Effects and Intervention of Air Pollution, School of Public Health, Xinxiang Medical University, Xinxiang, Henan Province 453003, China
| | - Weidong Wu
- Henan International Collaborative Laboratory for Health Effects and Intervention of Air Pollution, School of Public Health, Xinxiang Medical University, Xinxiang, Henan Province 453003, China.
| |
Collapse
|
19
|
Sophonnithiprasert T, Saelee P, Pongtheerat T. GSTM1 and GSTT1 copy number variants and the risk to Thai females of hepatocellular carcinoma. J Gastrointest Oncol 2019; 10:324-329. [PMID: 31032101 DOI: 10.21037/jgo.2018.09.14] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Background Hepatocellular carcinoma (HCC) is a common malignancy found throughout the world that most often occurs in males. The cancer is associated with many risk factors such as viral infection, cirrhosis, alcohol, smoking, and fungal toxins. GSTM1 and GSTT1 are detoxification enzymes activated by the cleansing of carcinogenic compounds. Low DNA copy numbers of Glutathione S-transferases M1 and T1 result in a loss of enzyme activity, which causes carcinogenesis factors. DNA copy number variants (CNVs) were determined to compare the differences between the frequencies of GSTM1 and GSTT1 in a control group and patients. Then, the association of these genes with the pathological/survival status of HCC patients was investigated. Methods Forty-nine Thai HCC patients' DNA and the genomic DNA of 66 healthy controls were investigated for GSTM1 and GSTT1 CNVs by real-time polymerase chain reaction (PCR). Then, the correlations between GSTM1 and GSTT1 patients' CNVs, the control group, and clinico-pathological parameters were determined. Results The results show that were no differences between the CNVs of GSTM1 and GSTT1 in the controls and patients (P≥0.05). Only GSTT1 genotypes 0/0 correlated to an increase in the risk of hepatocellular carcinogenesis (OR value was 1.88). GSTM1 CNVs were associated with the gender of patients (P=0.002). However, no correlations were found between GSTT1 CNVs and any of the clinico-pathological parameters. Conclusions The results suggest that only GSTT1 CNVs are associated with increased risk factors of HCC in Thais. GSTM1 copy numbers had a dominant correlation with female HCC patients.
Collapse
Affiliation(s)
- Thanet Sophonnithiprasert
- Unit of Biochemistry, Department of Medical Sciences, Faculty of Science, Rangsit University, Patumthani, Thailand
| | - Pensri Saelee
- Research Division, National Cancer Institute, Bangkok, Thailand
| | - Tanett Pongtheerat
- Unit of Biochemistry, Department of Medical Sciences, Faculty of Science, Rangsit University, Patumthani, Thailand
| |
Collapse
|
20
|
Ding Z, Wang K, Li J, Tan Q, Tan W, Guo G. Association between glutathione S‐transferase gene M1 and T1 polymorphisms and chronic obstructive pulmonary disease risk: A meta‐analysis. Clin Genet 2018; 95:53-62. [PMID: 29704242 DOI: 10.1111/cge.13373] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2018] [Revised: 04/23/2018] [Accepted: 04/23/2018] [Indexed: 12/27/2022]
Affiliation(s)
- Z. Ding
- Department of Respiratory MedicineWeifang People's Hospital Weifang China
| | - K. Wang
- Department of Respiratory MedicineWeifang People's Hospital Weifang China
| | - J. Li
- Department of Respiratory MedicineWeifang People's Hospital Weifang China
| | - Q. Tan
- Department of Respiratory MedicineWeifang People's Hospital Weifang China
| | - W. Tan
- Department of Respiratory MedicineWeifang People's Hospital Weifang China
| | - G. Guo
- Department of Respiratory MedicineWeifang People's Hospital Weifang China
| |
Collapse
|
21
|
Kim D, Chen Z, Zhou LF, Huang SX. Air pollutants and early origins of respiratory diseases. Chronic Dis Transl Med 2018; 4:75-94. [PMID: 29988883 PMCID: PMC6033955 DOI: 10.1016/j.cdtm.2018.03.003] [Citation(s) in RCA: 96] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Indexed: 12/13/2022] Open
Abstract
Air pollution is a global health threat and causes millions of human deaths annually. The late onset of respiratory diseases in children and adults due to prenatal or perinatal exposure to air pollutants is emerging as a critical concern in human health. Pregnancy and fetal development stages are highly susceptible to environmental exposure and tend to develop a long-term impact in later life. In this review, we briefly glance at the direct impact of outdoor and indoor air pollutants on lung diseases and pregnancy disorders. We further focus on lung complications in later life with early exposure to air pollutants. Epidemiological evidence is provided to show the association of prenatal or perinatal exposure to air pollutants with various adverse birth outcomes, such as preterm birth, lower birth weight, and lung developmental defects, which further associate with respiratory diseases and reduced lung function in children and adults. Mechanistic evidence is also discussed to support that air pollutants impact various cellular and molecular targets at early life, which link to the pathogenesis and altered immune responses related to abnormal respiratory functions and lung diseases in later life.
Collapse
Affiliation(s)
- Dasom Kim
- Department of Environmental Health, University of Cincinnati College of Medicine, Cincinnati, OH 45249, USA
| | - Zi Chen
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Lin-Fu Zhou
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Shou-Xiong Huang
- Department of Environmental Health, University of Cincinnati College of Medicine, Cincinnati, OH 45249, USA
| |
Collapse
|
22
|
Cheng W, Duncan KE, Ghio AJ, Ward-Caviness C, Karoly ED, Diaz-Sanchez D, Conolly RB, Devlin RB. Changes in Metabolites Present in Lung-Lining Fluid Following Exposure of Humans to Ozone. Toxicol Sci 2018; 163:430-439. [PMID: 29471466 PMCID: PMC6348881 DOI: 10.1093/toxsci/kfy043] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Controlled human exposure to the oxidant air pollutant ozone causes decrements in lung function and increased inflammation as evidenced by neutrophil influx into the lung and increased levels of proinflammatory cytokines in the airways. Here we describe a targeted metabolomics evaluation of human bronchoalveolar lavage fluid (BALF) following controlled in vivo exposure to ozone to gain greater insight into its pulmonary effects. In a 2-arm cross-over study, each healthy adult human volunteer was randomly exposed to filtered air (FA) and to 0.3 ppm ozone for 2 h while undergoing intermittent exercise with a minimum of 4 weeks between exposures. Bronchoscopy was performed and BALF obtained at 1 (n = 9) or 24 (n = 23) h postexposure. Metabolites were detected using ultrahigh performance liquid chromatography-tandem mass spectroscopy. At 1-h postexposure, a total of 28 metabolites were differentially expressed (DE) (p < .05) following ozone exposure compared with FA-exposure. These changes were associated with increased glycolysis and antioxidant responses, suggesting rapid increased energy utilization as part of the cellular response to oxidative stress. At 24-h postexposure, 41 metabolites were DE. Many of the changes were in amino acids and linked with enhanced proteolysis. Changes associated with increased lipid membrane turnover were also observed. These later-stage changes were consistent with ongoing repair of airway tissues. There were 1.37 times as many metabolites were differentially expressed at 24 h compared with 1-h postexposure. The changes at 1 h reflect responses to oxidative stress while the changes at 24 h indicate a broader set of responses consistent with tissue repair. These results illustrate the ability of metabolomic analysis to identify mechanistic features of ozone toxicity and aspects of the subsequent tissue response.
Collapse
Affiliation(s)
- WanYun Cheng
- National Health and Environmental Effects Research Laboratory, U.S. Environmental Protection Agency, Research Triangle Park, North Carolina 2799
| | - Kelly E Duncan
- School of Medicine, University of North Carolina-Chapel Hill, Chapel Hill, North Carolina 27599
| | - Andrew J Ghio
- National Health and Environmental Effects Research Laboratory, U.S. Environmental Protection Agency, Research Triangle Park, North Carolina 2799
| | - Cavin Ward-Caviness
- National Health and Environmental Effects Research Laboratory, U.S. Environmental Protection Agency, Research Triangle Park, North Carolina 2799
| | | | - David Diaz-Sanchez
- National Health and Environmental Effects Research Laboratory, U.S. Environmental Protection Agency, Research Triangle Park, North Carolina 2799
| | - Rory B Conolly
- National Health and Environmental Effects Research Laboratory, U.S. Environmental Protection Agency, Research Triangle Park, North Carolina 2799
| | - Robert B Devlin
- National Health and Environmental Effects Research Laboratory, U.S. Environmental Protection Agency, Research Triangle Park, North Carolina 2799
| |
Collapse
|
23
|
Stankovic M, Nikolic A, Nagorni-Obradovic L, Petrovic-Stanojevic N, Radojkovic D. Gene–Gene Interactions Between Glutathione S-Transferase M1 and Matrix Metalloproteinases 1, 9, and 12 in Chronic Obstructive Pulmonary Disease in Serbians. COPD 2017; 14:581-589. [DOI: 10.1080/15412555.2017.1369022] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Marija Stankovic
- Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Belgrade, Serbia
| | - Aleksandra Nikolic
- Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Belgrade, Serbia
| | - Ljudmila Nagorni-Obradovic
- Clinic for Pulmonary Diseases, Clinical Centre of Serbia, Belgrade, Serbia
- School of Medicine, University of Belgrade, Belgrade, Serbia
| | - Natasa Petrovic-Stanojevic
- Department of Pulmonology, Zvezdara University Medical Center, Belgrade, Serbia
- School of Dentistry, University of Belgrade, Belgrade, Serbia
| | - Dragica Radojkovic
- Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Belgrade, Serbia
| |
Collapse
|
24
|
Frampton MW, Balmes JR, Bromberg PA, Stark P, Arjomandi M, Hazucha MJ, Rich DQ, Hollenbeck-Pringle D, Dagincourt N, Alexis N, Ganz P, Zareba W, Costantini MG. Multicenter Ozone Study in oldEr Subjects (MOSES): Part 1. Effects of Exposure to Low Concentrations of Ozone on Respiratory and Cardiovascular Outcomes. Res Rep Health Eff Inst 2017; 2017:1-107. [PMID: 31898880 PMCID: PMC7266375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/10/2023] Open
Abstract
INTRODUCTION Exposure to air pollution is a well-established risk factor for cardiovascular morbidity and mortality. Most of the evidence supporting an association between air pollution and adverse cardiovascular effects involves exposure to particulate matter (PM). To date, little attention has been paid to acute cardiovascular responses to ozone, in part due to the notion that ozone causes primarily local effects on lung function, which are the basis for the current ozone National Ambient Air Quality Standards (NAAQS). There is evidence from a few epidemiological studies of adverse health effects of chronic exposure to ambient ozone, including increased risk of mortality from cardiovascular disease. However, in contrast to the well-established association between ambient ozone and various nonfatal adverse respiratory effects, the observational evidence for impacts of acute (previous few days) increases in ambient ozone levels on total cardiovascular mortality and morbidity is mixed. Ozone is a prototypic oxidant gas that reacts with constituents of the respiratory tract lining fluid to generate reactive oxygen species (ROS) that can overwhelm antioxidant defenses and cause local oxidative stress. Pathways by which ozone could cause cardiovascular dysfunction include alterations in autonomic balance, systemic inflammation, and oxidative stress. These initial responses could lead ultimately to arrhythmias, endothelial dysfunction, acute arterial vasoconstriction, and procoagulant activity. Individuals with impaired antioxidant defenses, such as those with the null variant of glutathione S-transferase mu 1 (GSTM1), may be at increased risk for acute health effects. The Multicenter Ozone Study in oldEr Subjects (MOSES) was a controlled human exposure study designed to evaluate whether short-term exposure of older, healthy individuals to ambient levels of ozone induces acute cardiovascular responses. The study was designed to test the a priori hypothesis that short-term exposure to ambient levels of ozone would induce acute cardiovascular responses through the following mechanisms: autonomic imbalance, systemic inflammation, and development of a prothrombotic vascular state. We also postulated a priori the confirmatory hypothesis that exposure to ozone would induce airway inflammation, lung injury, and lung function decrements. Finally, we postulated the secondary hypotheses that ozone-induced acute cardiovascular responses would be associated with: (a) increased systemic oxidative stress and lung effects, and (b) the GSTM1-null genotype. METHODS The study was conducted at three clinical centers with a separate Data Coordinating and Analysis Center (DCAC) using a common protocol. All procedures were approved by the institutional review boards (IRBs) of the participating centers. Healthy volunteers 55 to 70 years of age were recruited. Consented participants who successfully completed the screening and training sessions were enrolled in the study. All three clinical centers adhered to common standard operating procedures (SOPs) and used common tracking and data forms. Each subject was scheduled to participate in a total of 11 visits: screening visit, training visit, and three sets of exposure visits, each consisting of the pre-exposure day, the exposure day, and the post-exposure day. The subjects spent the night in a nearby hotel the night of the pre-exposure day. On exposure days, the subjects were exposed for three hours in random order to 0 ppb ozone (clean air), 70 ppb ozone, and 120 ppm ozone, alternating 15 minutes of moderate exercise with 15 minutes of rest. A suite of cardiovascular and pulmonary endpoints was measured on the day before, the day of, and up to 22 hours after, each exposure. The endpoints included: (1) electrocardiographic changes (continuous Holter monitoring: heart rate variability [HRV], repolarization, and arrhythmia); (2) markers of inflammation and oxidative stress (C-reactive protein [CRP], interleukin-6 [IL-6], 8-isoprostane, nitrotyrosine, and P-selectin); (3) vascular function measures (blood pressure [BP], flow-mediated dilatation [FMD] of the brachial artery, and endothelin-1 [ET-1]; (4) venous blood markers of platelet activation, thrombosis, and microparticle-associated tissue factor activity (MP-TFA); (5) pulmonary function (spirometry); (6) markers of airway epithelial cell injury (increases in plasma club cell protein 16 [CC16] and sputum total protein); and (7) markers of lung inflammation in sputum (polymorphonuclear leukocytes [PMN], IL-6, interleukin-8 [IL-8], and tumor necrosis factor-alpha [TNF-α]). Sputum was collected only at 22 hours after exposure. The analyses of the continuous electrocardiographic monitoring, the brachial artery ultrasound (BAU) images, and the blood and sputum samples were carried out by core laboratories. The results of all analyses were submitted directly to the DCAC. The variables analyzed in the statistical models were represented as changes from pre-exposure to post-exposure (post-exposure minus pre-exposure). Mixed-effect linear models were used to evaluate the impact of exposure to ozone on the prespecified primary and secondary continuous outcomes. Site and time (when multiple measurements were taken) were controlled for in the models. Three separate interaction models were constructed for each outcome: ozone concentration by subject sex; ozone concentration by subject age; and ozone concentration by subject GSTM1 status (null or sufficient). Because of the issue of multiple comparisons, the statistical significance threshold was set a priori at P < 0.01. RESULTS Subject recruitment started in June 2012, and the first subject was randomized on July 25, 2012. Subject recruitment ended on December 31, 2014, and testing of all subjects was completed by April 30, 2015. A total of 87 subjects completed all three exposures. The mean age was 59.9 ± 4.5 years, 60% of the subjects were female, 88% were white, and 57% were GSTM1 null. Mean baseline body mass index (BMI), BP, cholesterol (total and low-density lipoprotein), and lung function were all within the normal range. We found no significant effects of ozone exposure on any of the primary or secondary endpoints for autonomic function, repolarization, ST segment change, or arrhythmia. Ozone exposure also did not cause significant changes in the primary endpoints for systemic inflammation (CRP) and vascular function (systolic blood pressure [SBP] and FMD) or secondary endpoints for systemic inflammation and oxidative stress (IL-6, P-selectin, and 8-isoprostane). Ozone did cause changes in two secondary endpoints: a significant increase in plasma ET-1 (P = 0.008) and a marginally significant decrease in nitrotyrosine (P = 0.017). Lastly, ozone exposure did not affect the primary prothrombotic endpoints (MP-TFA and monocyte-platelet conjugate count) or any secondary markers of prothrombotic vascular status (platelet activation, circulating microparticles [MPs], von Willebrand factor [vWF], or fibrinogen.). Although our hypothesis focused on possible acute cardiovascular effects of exposure to low levels of ozone, we recognized that the initial effects of inhaled ozone involve the lower airways. Therefore, we looked for: (a) changes in lung function, which are known to occur during exposure to ozone and are maximal at the end of exposure; and (b) markers of airway injury and inflammation. We found an increase in forced vital capacity (FVC) and forced expiratory volume in 1 second (FEV₁) after exposure to 0 ppb ozone, likely due to the effects of exercise. The FEV₁ increased significantly 15 minutes after 0 ppb exposure (85 mL; 95% confidence interval [CI], 64 to 106; P < 0.001), and remained significantly increased from pre-exposure at 22 hours (45 mL; 95% CI, 26 to 64; P < 0.001). The increase in FVC followed a similar pattern. The increase in FEV₁ and FVC were attenuated in a dose-response manner by exposure to 70 and 120 ppb ozone. We also observed a significant ozone-induced increase in the percentage of sputum PMN 22 hours after exposure at 120 ppb compared to 0 ppb exposure (P = 0.003). Plasma CC16 also increased significantly after exposure to 120 ppb (P < 0.001). Sputum IL-6, IL-8, and TNF-α concentrations were not significantly different after ozone exposure. We found no significant interactions with sex, age, or GSTM1 status regarding the effect of ozone on lung function, percentage of sputum PMN, or plasma CC16. CONCLUSIONS In this multicenter clinical study of older healthy subjects, ozone exposure caused concentration-related reductions in lung function and presented evidence for airway inflammation and injury. However, there was no convincing evidence for effects on cardiovascular function. Blood levels of the potent vasoconstrictor, ET-1, increased with ozone exposure (with marginal statistical significance), but there were no effects on BP, FMD, or other markers of vascular function. Blood levels of nitrotyrosine decreased with ozone exposure, the opposite of our hypothesis. Our study does not support acute cardiovascular effects of low-level ozone exposure in healthy older subjects. Inclusion of only healthy older individuals is a major limitation, which may affect the generalizability of our findings. We cannot exclude the possibility of effects with higher ozone exposure concentrations or more prolonged exposure, or the possibility that subjects with underlying vascular disease, such as hypertension or diabetes, would show effects under these conditions.
Collapse
Affiliation(s)
- M W Frampton
- University of Rochester Medical Center, Rochester, New York
| | | | | | - P Stark
- New England Research Institute, Watertown, Massachusetts
| | | | | | - D Q Rich
- University of Rochester Medical Center, Rochester, New York
| | | | - N Dagincourt
- New England Research Institute, Watertown, Massachusetts
| | - N Alexis
- University of North Carolina, Chapel Hill
| | - P Ganz
- University of California, San Francisco
| | - W Zareba
- University of Rochester Medical Center, Rochester, New York
| | | |
Collapse
|
25
|
Feng F, Jin Y, Duan L, Yan Z, Wang S, Li F, Liu Y, Samet JM, Wu W. Regulation of ozone-induced lung inflammation by the epidermal growth factor receptor in mice. ENVIRONMENTAL TOXICOLOGY 2016; 31:2016-2027. [PMID: 26464147 DOI: 10.1002/tox.22202] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Revised: 09/25/2015] [Accepted: 09/26/2015] [Indexed: 06/05/2023]
Abstract
Human exposure to the highly reactive oxidant gas Ozone (O3 ) is associated with inflammatory responses in the airway epithelium. The mechanisms responsible have not been fully elucidated. Epidermal growth factor receptor (EGFR) has previously been shown to play a critical role in the pathogenesis of lung inflammation. To define the role of EGFR in O3 -induced lung inflammation in mice. 40 BALB/c mice were exposed to filtered air (FA) or (0.25, 0.5, 1.00 ppm) O3 for 3 h per day for 7 consecutive days. Levels of reactive oxygen species (ROS), EGF, and transforming growth factor α (TGF-α) in the bronchoalveolar lavage fluid (BALF) of mice were measured using ELISA. BALB/c mice were intratracheally instilled with the EGFR kinase inhibitor PD153035 2 h prior to O3 exposure and every other day thereafter. Phosphorylation of EGFR (Y1068) in lung sections was determined using immunohistochemical staining and western blot 24 h after exposure. Inhalation of O3 induced pronounced lung inflammation in a dose-dependent manner. Levels of ROS, TGF-α, and total proteins and cells in the BALF of mice exposed to 0.5 ppm or 1.0 ppm of O3 were markedly elevated relative to those in the BALF of the mice exposed to FA. In addition, exposure to O3 induced EGFR(Y1068) phosphorylation in the airway epithelium. Administration of PD153035 resulted in a significantly reduced lung inflammation as well as EGFR phosphorylation induced by O3 exposure. Inhalation of O3 leads to inflammatory responses that are dependent on the activation the EGFR in the airway epithelium. © 2015 Wiley Periodicals, Inc. Environ Toxicol 31: 2016-2027, 2016.
Collapse
Affiliation(s)
- Feifei Feng
- College of Public Health, Zhengzhou University, Zhengzhou, China
| | - Yuefei Jin
- College of Public Health, Zhengzhou University, Zhengzhou, China
| | - Liju Duan
- College of Public Health, Zhengzhou University, Zhengzhou, China
| | - Zhen Yan
- College of Public Health, Zhengzhou University, Zhengzhou, China
| | - Shouying Wang
- School of Public Health, Xinxiang Medical University, China
| | - Fangfang Li
- School of Public Health, Xinxiang Medical University, China
| | - Yingying Liu
- School of Public Health, Xinxiang Medical University, China
| | - James M Samet
- Environmental Public Health Division, National Health and Environmental Effects Research Laboratory, United States Environmental Protection Agency, Chapel Hill, North Carolina
| | - Weidong Wu
- School of Public Health, Xinxiang Medical University, China
| |
Collapse
|
26
|
Bromberg PA. Mechanisms of the acute effects of inhaled ozone in humans. Biochim Biophys Acta Gen Subj 2016; 1860:2771-81. [PMID: 27451958 DOI: 10.1016/j.bbagen.2016.07.015] [Citation(s) in RCA: 99] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Revised: 07/13/2016] [Accepted: 07/18/2016] [Indexed: 12/31/2022]
Abstract
Ambient air ozone (O3) is generated photochemically from oxides of nitrogen and volatile hydrocarbons. Inhaled O3 causes remarkably reversible acute lung function changes and inflammation. Approximately 80% of inhaled O3 is deposited on the airways. O3 reacts rapidly with CC double bonds in hydrophobic airway and alveolar surfactant-associated phospholipids and cholesterol. Resultant primary ozonides further react to generate bioactive hydrophilic products that also initiate lipid peroxidation leading to eicosanoids and isoprostanes of varying electrophilicity. Airway surface liquid ascorbate and urate also scavenge O3. Thus, inhaled O3 may not interact directly with epithelial cells. Acute O3-induced lung function changes are dominated by involuntary inhibition of inspiration (rather than bronchoconstriction), mediated by stimulation of intraepithelial nociceptive vagal C-fibers via activation of transient receptor potential (TRP) A1 cation channels by electrophile (e.g., 4-oxo-nonenal) adduction of TRPA1 thiolates enhanced by PGE2-stimulated sensitization. Acute O3-induced neutrophilic airways inflammation develops more slowly than the lung function changes. Surface macrophages and epithelial cells are involved in the activation of epithelial NFkB and generation of proinflammatory mediators such as IL-6, IL-8, TNFa, IL-1b, ICAM-1, E-selectin and PGE2. O3-induced partial depolymerization of hyaluronic acid and the release of peroxiredoxin-1 activate macrophage TLR4 while oxidative epithelial cell release of EGFR ligands such as TGFa or EGFR transactivation by activated Src may also be involved. The ability of lipid ozonation to generate potent electrophiles also provides pathways for Nrf2 activation and inhibition of canonical NFkB activation. This article is part of a Special Issue entitled Air Pollution, edited by Wenjun Ding, Andrew J. Ghio and Weidong Wu.
Collapse
Affiliation(s)
- Philip A Bromberg
- Center for Environmental Medicine, Asthma and Lung Biology, and Division of Pulmonary and Critical Care Medicine, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, United States.
| |
Collapse
|
27
|
Bowatte G, Lodge CJ, Knibbs LD, Lowe AJ, Erbas B, Dennekamp M, Marks GB, Giles G, Morrison S, Thompson B, Thomas PS, Hui J, Perret JL, Abramson MJ, Walters H, Matheson MC, Dharmage SC. Traffic-related air pollution exposure is associated with allergic sensitization, asthma, and poor lung function in middle age. J Allergy Clin Immunol 2016; 139:122-129.e1. [PMID: 27372567 DOI: 10.1016/j.jaci.2016.05.008] [Citation(s) in RCA: 110] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2015] [Revised: 05/04/2016] [Accepted: 05/16/2016] [Indexed: 11/18/2022]
Abstract
BACKGROUND Traffic-related air pollution (TRAP) exposure is associated with allergic airway diseases and reduced lung function in children, but evidence concerning adults, especially in low-pollution settings, is scarce and inconsistent. OBJECTIVES We sought to determine whether exposure to TRAP in middle age is associated with allergic sensitization, current asthma, and reduced lung function in adults, and whether these associations are modified by variants in Glutathione S-Transferase genes. METHODS The study sample comprised the proband 2002 laboratory study of the Tasmanian Longitudinal Health Study. Mean annual residential nitrogen dioxide (NO2) exposure was estimated for current residential addresses using a validated land-use regression model. Associations between TRAP exposure and allergic sensitization, lung function, current wheeze, and asthma (n = 1405) were investigated using regression models. RESULTS Increased mean annual NO2 exposure was associated with increased risk of atopy (adjusted odds ratio [aOR], 1.14; 95% CI, 1.02-1.28 per 1 interquartile range increase in NO2 [2.2 ppb]) and current wheeze (aOR, 1.14; 1.02-1.28). Similarly, living less than 200 m from a major road was associated with current wheeze (aOR, 1.38; 95% CI, 1.06-1.80) and atopy (aOR, 1.26; 95% CI, 0.99-1.62), and was also associated with having significantly lower prebronchodilator and postbronchodilator FEV1 and prebronchodilator forced expiratory flow at 25% to 75% of forced vital capacity. We found evidence of interactions between living less than 200 m from a major road and GSTT1 polymorphism for atopy, asthma, and atopic asthma. Overall, carriers of the GSTT1 null genotype had an increased risk of asthma and allergic outcomes if exposed to TRAP. CONCLUSIONS Even relatively low TRAP exposures confer an increased risk of adverse respiratory and allergic outcomes in genetically susceptible individuals.
Collapse
Affiliation(s)
- Gayan Bowatte
- Allergy and Lung Health Unit, Centre for Epidemiology and Biostatistics, School of Population & Global Health, the University of Melbourne, Melbourne, Australia
| | - Caroline J Lodge
- Allergy and Lung Health Unit, Centre for Epidemiology and Biostatistics, School of Population & Global Health, the University of Melbourne, Melbourne, Australia
| | - Luke D Knibbs
- School of Public Health, the University of Queensland, Brisbane, Australia
| | - Adrian J Lowe
- Allergy and Lung Health Unit, Centre for Epidemiology and Biostatistics, School of Population & Global Health, the University of Melbourne, Melbourne, Australia
| | - Bircan Erbas
- School of Psychology & Public Health, Department of Public Health, La Trobe University, Melbourne, Australia
| | - Martine Dennekamp
- School of Public Health & Preventive Medicine, Monash University, Melbourne, Australia
| | - Guy B Marks
- Woolcock Institute of Medical Research, The University of Sydney, Sydney, Australia; South Western Sydney Clinical School, University of New South Wales, Sydney, Australia
| | - Graham Giles
- Cancer Epidemiology Centre, the Cancer Council Victoria, Melbourne, Australia
| | | | | | - Paul S Thomas
- Inflammation and Infection Research Centre, Faculty of Medicine, University of New South Wales, Sydney, Australia
| | - Jennie Hui
- Busselton Population Medical Research Institute, Perth, Australia; School of Population Health, the University of Western Australia, Perth, Australia; School of Pathology and Laboratory Medicine, the University of Western Australia, Perth, Australia; PathWest Laboratory Medicine of WA, Sir Charles Gairdner Hospital, Perth, Australia
| | - Jennifer L Perret
- Allergy and Lung Health Unit, Centre for Epidemiology and Biostatistics, School of Population & Global Health, the University of Melbourne, Melbourne, Australia
| | - Michael J Abramson
- School of Public Health & Preventive Medicine, Monash University, Melbourne, Australia
| | - Haydn Walters
- NHMRC CRE, University of Tasmania Medical School, Hobart, Australia
| | - Melanie C Matheson
- Allergy and Lung Health Unit, Centre for Epidemiology and Biostatistics, School of Population & Global Health, the University of Melbourne, Melbourne, Australia
| | - Shyamali C Dharmage
- Allergy and Lung Health Unit, Centre for Epidemiology and Biostatistics, School of Population & Global Health, the University of Melbourne, Melbourne, Australia; Murdoch Childrens Research Institute, Melbourne, Australia.
| |
Collapse
|
28
|
Saadeh R, Klaunig J. Children's Inter-Individual Variability and Asthma Development. Int J Health Sci (Qassim) 2015; 9:456-467. [PMID: 26715926 PMCID: PMC4682601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/05/2023] Open
Abstract
Children of different ages vary in their response to environmental stressors due to their continuous development and changes in their bodies' anatomy, physiology, and biochemistry. Each age group of children has special biological features that distinguish their toxicokinetic and toxicodynamic characteristics from other age groups. The variability in responses extends to include children of the same age group. These intra- and inter-group differences in biological features explains the variability in responses to environmental exposures. Based on such differences in children's responses to exposures, adverse health outcomes and diseases develop differently in children. One of these diseases that are common in children is asthma. Asthma is a complex respiratory chronic disease that is multifactorial in origin. This paper discusses how variability in certain factors among children contributes to asthma occurrence or exacerbation, and links these factors to asthma in children of different ages. The importance of this review is to provide an insight on factors affecting asthma prevalence among children. These factors are usually overlooked in clinical or public health practice, which might significantly affect asthma management, and decrease the predictability of asthma detection measures. Therefore, keeping these factors into consideration can significantly improve asthma treatment and assist in asthma prevention amongst susceptible populations.
Collapse
Affiliation(s)
- Rami Saadeh
- Department of Environmental Health, School of Public Health, Indiana University at Bloomington, Bloomington, Indiana, USA
| | - James Klaunig
- Department of Environmental Health, School of Public Health, Indiana University at Bloomington, Bloomington, Indiana, USA
| |
Collapse
|
29
|
Affiliation(s)
- D B Peden
- Center for Environmental Medicine, Asthma and Lung Biology & Department of Pediatrics, The School of Medicine, The University of North Carolina, Chapel Hill, NC, USA
| |
Collapse
|
30
|
Paradis F, Yue S, Grant JR, Stothard P, Basarab JA, Fitzsimmons C. Transcriptomic analysis by RNA sequencing reveals that hepatic interferon-induced genes may be associated with feed efficiency in beef heifers1. J Anim Sci 2015; 93:3331-41. [DOI: 10.2527/jas.2015-8975] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
31
|
Environmental effects on immune responses in patients with atopy and asthma. J Allergy Clin Immunol 2014; 134:1001-8. [PMID: 25439226 DOI: 10.1016/j.jaci.2014.07.064] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2014] [Revised: 07/25/2014] [Accepted: 07/30/2014] [Indexed: 12/13/2022]
Abstract
Despite attempts and some successes to improve air quality over the decades, current US national trends suggest that exposure to outdoor and indoor air pollution remains a significant risk factor for both the development of asthma and the triggering of asthma symptoms. Emerging science also suggests that environmental exposures during the prenatal period and early childhood years increase the risk of asthma. Multiple mechanisms mediate this risk because a wide range of deleterious air pollutants contribute to the pathogenesis of asthma across a variety of complex asthma phenotypes. In this review we will consider the role of altered innate and adaptive immune responses, gene-environment interactions, epigenetic regulation, and possibly gene-environment-epigene interactions. Gaining a greater understanding of the mechanisms that underlie the effect of exposure to air pollution on asthma, allergies, and other airway diseases can identify targets for therapy. Such interventions will include pollutant source reduction among those most exposed and most vulnerable and novel pharmaceutical strategies to reduce asthma morbidity.
Collapse
|
32
|
Ather JL, Martin RA, Ckless K, Poynter ME. Inflammasome Activity in Non-Microbial Lung Inflammation. JOURNAL OF ENVIRONMENTAL IMMUNOLOGY AND TOXICOLOGY 2014; 1:108-117. [PMID: 25642415 PMCID: PMC4308734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
The understanding of interleukin-1 (IL-1) family cytokines in inflammatory disease has rapidly developed, due in part to the discovery and characterization of inflammasomes, which are multi-subunit intracellular protein scaffolds principally enabling recognition of a myriad of cellular stimuli, leading to the activation of caspase-1 and the processing of IL-1β and IL-18. Studies continue to elucidate the role of inflammasomes in immune responses induced by both microbes and environmental factors. This review focuses on the current understanding of inflammasome activity in the lung, with particular focus on the non-microbial instigators of inflammasome activation, including inhaled antigens, oxidants, cigarette smoke, diesel exhaust particles, mineral fibers, and engineered nanomaterials, as well as exposure to trauma and pre-existing inflammatory conditions such as metabolic syndrome. Inflammasome activity in these sterile inflammatory states contribute to diseases including asthma, chronic obstructive disease, acute lung injury, ventilator-induced lung injury, pulmonary fibrosis, and lung cancer.
Collapse
Affiliation(s)
- Jennifer L. Ather
- Vermont Lung Center, Division of Pulmonary Disease and Critical Care, Department of Medicine, University of Vermont, Burlington, USA
| | - Rebecca A. Martin
- Vermont Lung Center, Division of Pulmonary Disease and Critical Care, Department of Medicine, University of Vermont, Burlington, USA
| | - Karina Ckless
- Chemistry Department, State University of New York at Plattsburgh, Plattsburgh, USA
| | - Matthew E. Poynter
- Vermont Lung Center, Division of Pulmonary Disease and Critical Care, Department of Medicine, University of Vermont, Burlington, USA
| |
Collapse
|
33
|
Tong H, Rappold AG, Caughey M, Hinderliter AL, Graff DW, Berntsen JH, Cascio WE, Devlin RB, Samet JM. Cardiovascular effects caused by increasing concentrations of diesel exhaust in middle-aged healthy GSTM1 null human volunteers. Inhal Toxicol 2014; 26:319-26. [PMID: 24655088 DOI: 10.3109/08958378.2014.889257] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
CONTEXT Epidemiological studies have shown an association between the incidence of adverse cardiovascular effects and exposure to ambient particulate matter (PM). Diesel exhaust (DE) is a major contributor to ambient PM and gaseous emissions in urban areas. OBJECTIVE This was a pilot study designed to evaluate concentration-dependent effects of short-term exposure to whole DE on the cardiovascular system in order to identify a threshold concentration that can elicit biological responses in healthy human volunteers. MATERIALS AND METHODS Six healthy middle-aged participants with glutathione-S-transferase-Mu 1 (GSTM1) null genotype underwent sequential exposures to 100 µg/m(3), 200 µg/m(3), and 300 µg/m(3) whole DE generated in real time using an idling diesel truck engine. Exposures were separated by 14 d and each was 2 h in duration. RESULTS We report concentration-dependent effects of exposure to DE, with 100 µg/m(3) concentration causing minimal cardiovascular effects, while exposure to 300 µg/m(3) DE for 2 h resulted in a borderline significant reduction of baseline brachial artery diameter (3.34 ± 0.27 mm pre- versus 3.23 ± 0.25 mm post-exposure; p = 0.08). Exposure to the highest concentration of DE also resulted in increases of 5 mmHg in diastolic blood pressure as well as a decrease in indices of the frequency domain of heart rate variability (HRV). DISCUSSION AND CONCLUSIONS These findings demonstrate that acute exposure to relatively high concentrations of DE produces cardiovascular changes in middle-aged GSTM1 null individuals. This study therefore suggests that arterial vasoconstriction and changes in HRV are responses through which traffic-related air pollution increases the risk of adverse cardiovascular outcomes.
Collapse
Affiliation(s)
- Haiyan Tong
- Environmental Public Health Division, National Health and Environmental Effects Research Laboratory, US Environmental Protection Agency, Research Triangle Park , NC , USA
| | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Shevelkin AV, Ihenatu C, Pletnikov MV. Pre-clinical models of neurodevelopmental disorders: focus on the cerebellum. Rev Neurosci 2014; 25:177-94. [PMID: 24523305 PMCID: PMC4052755 DOI: 10.1515/revneuro-2013-0049] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2013] [Accepted: 12/31/2013] [Indexed: 12/24/2022]
Abstract
Recent studies have advanced our understanding of the role of the cerebellum in non-motor behaviors. Abnormalities in the cerebellar structure have been demonstrated to produce changes in emotional, cognitive, and social behaviors resembling clinical manifestations observed in patients with autism spectrum disorders (ASD) and schizophrenia. Several animal models have been used to evaluate the effects of relevant environmental and genetic risk factors on the cerebellum development and function. However, very few models of ASD and schizophrenia selectively target the cerebellum and/or specific cell types within this structure. In this review, we critically evaluate the strength and weaknesses of these models. We will propose that the future progress in this field will require time- and cell type-specific manipulations of disease-relevant genes, not only selectively in the cerebellum, but also in frontal brain areas connected with the cerebellum. Such information can advance our knowledge of the cerebellar contribution to non-motor behaviors in mental health and disease.
Collapse
|
35
|
Wang T, Wang B. Association between Glutathione S-transferase M1/Glutathione S-transferase T1 polymorphisms and Parkinson's disease: a meta-analysis. J Neurol Sci 2013; 338:65-70. [PMID: 24382428 DOI: 10.1016/j.jns.2013.12.018] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2013] [Revised: 11/03/2013] [Accepted: 12/10/2013] [Indexed: 01/28/2023]
Abstract
The Glutathione S-transferase M1 (GSTM1) and Glutathione S-transferase T1 (GSTT1) genes have been studied extensively as potential candidate genes for the risk of Parkinson's disease (PD). However, direct evidence from genetic association studies remains inconclusive. In order to address this issue, we performed an updated and refined meta-analysis to determine the effect of GSTM1 and GSTT1 polymorphisms on Parkinson's disease. A fixed-effect model was utilized to calculate the combined odds ratio (OR), OR of different ethnicities, and 95% confidence intervals (CIs). Potential publication bias was estimated. Homogeneity of the included studies was also evaluated. The pooled OR was 1.13 [95% CI (1.03, 1.24)] and 0.96 [95% CI (0.82, 1.12)] for GSTM1 and GSTT1 polymorphisms, respectively. Analysis according to different races found no association between GSTM1/GSTT1 polymorphisms and PD risks except for GSTM1 variant in Caucasians, which showed a weak correlation (OR 1.16 [95% CI (1.04, 1.29), I squared=6.2%, p=0.384]). Neither publication bias nor heterogeneity was found among the included studies. The results of this meta-analysis suggest that GSTM1 polymorphism is weakly associated with the risk of PD in Caucasians whereas GSTT1 polymorphism is not a PD risk factor.
Collapse
Affiliation(s)
- Tengfei Wang
- Department of Pharmacology, University of Tennessee Health Science Center, Memphis, TN, USA.
| | - Bin Wang
- Department of Pharmacology, Shaanxi University of Traditional Chinese Medicine, Xianyang, Shaanxi, PR China.
| |
Collapse
|
36
|
Weichenthal SA, Godri-Pollitt K, Villeneuve PJ. PM2.5, oxidant defence and cardiorespiratory health: a review. Environ Health 2013; 12:40. [PMID: 23641908 PMCID: PMC3652795 DOI: 10.1186/1476-069x-12-40] [Citation(s) in RCA: 101] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2012] [Accepted: 05/01/2013] [Indexed: 05/20/2023]
Abstract
Airborne fine particle mass concentrations (PM2.5) are used for ambient air quality management worldwide based in part on known cardiorespiratory health effects. While oxidative stress is generally thought to be an important mechanism in determining these effects, relatively few studies have specifically examined how oxidant defence may impact susceptibility to particulate air pollution. Here we review studies that explore the impact of polymorphisms in anti-oxidant related genes or anti-oxidant supplementation on PM2.5-induced cardiorespiratory outcomes in an effort to summarize existing evidence related to oxidative stress defence and the health effects of PM2.5. Recent studies of PM-oxidative burden were also examined. In total, nine studies were identified and reviewed and existing evidence generally suggests that oxidant defence may modify the impact of PM2.5 exposure on various health outcomes, particularly heart rate variability (a measure of autonomic function) which was the most common outcome examined in the studies reviewed. Few studies examined interactions between PM2.5 and oxidant defence for respiratory outcomes, and in general studies focused primarily on acute health effects. Therefore, further evaluation of the potential modifying role of oxidant defence in PM2.5-induced health effects is required, particularly for chronic outcomes. Similarly, while an exposure metric that captures the ability of PM2.5 to cause oxidative stress may offer advantages over traditional mass concentration measurements, little epidemiological evidence is currently available to evaluate the potential benefits of such an approach. Therefore, further evaluation is required to determine how this metric may be incorporated in ambient air quality management.
Collapse
Affiliation(s)
| | - Krystal Godri-Pollitt
- University of Toronto, Southern Ontario Centre for Atmospheric Aerosol Research, Toronto, Canada
| | | |
Collapse
|
37
|
Wu W, Peden DB, McConnell R, Fruin S, Diaz-Sanchez D. Glutathione-S-transferase M1 regulation of diesel exhaust particle-induced pro-inflammatory mediator expression in normal human bronchial epithelial cells. Part Fibre Toxicol 2012; 9:31. [PMID: 22867088 PMCID: PMC3480908 DOI: 10.1186/1743-8977-9-31] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2011] [Accepted: 08/01/2012] [Indexed: 12/11/2022] Open
Abstract
Background Diesel exhaust particles (DEP) contribute substantially to ambient particulate matter (PM) air pollution in urban areas. Inhalation of PM has been associated with increased incidence of lung disease in susceptible populations. We have demonstrated that the glutathione S-transferase M1 (GSTM1) null genotype could aggravate DEP-induced airway inflammation in human subjects. Given the critical role airway epithelial cells play in the pathogenesis of airway inflammation, we established the GSTM1 deficiency condition in primary bronchial epithelial cells from human volunteers with GSTM1 sufficient genotype (GSTM1+) using GSTM1 shRNA to determine whether GSTM1 deficiency could exaggerate DEP-induced expression of interleukin-8 (IL-8) and IL-1β proteins. Furthermore, the mechanisms underlying GSTM1 regulation of DEP-induced IL-8 and IL-1β expression were also investigated. Methods IL-8 and IL-1β protein levels were measured using enzyme-linked immunosorbent assay. GSTM1 deficiency in primary human bronchial epithelial cells was achieved using lentiviral GSTM1 shRNA particles and verified using real-time polymerase chain reaction and immunoblotting. Intracellular reactive oxygen species (ROS) production was evaluated using flow cytometry. Phosphorylation of protein kinases was detected using immunoblotting. Results Exposure of primary human bronchial epithelial cells (GSTM1+) to 25-100 μg/ml DEP for 24 h significantly increased IL-8 and IL-1β protein expression. Knockdown of GSTM1 in these cells further elevated DEP-induced IL-8 and IL-1β expression, implying that GSTM1 deficiency aggravated DEP-induced pro-inflammatory response. DEP stimulation induced the phosphorylation of extracellular signal-regulated kinase (ERK) and Akt, the downstream kinase of phosphoinositide 3-kinase (PI3K), in GSTM1+ bronchial epithelial cells. Pharmacological inhibition of ERK kinase and PI3K activity blocked DEP-induced IL-8 and IL-1β expression. DEP-induced ERK and Akt phosphorylation could be increased by GSTM1 knockdown. In addition, pretreatment of HBEC with the antioxidant N-acetyl cysteine significantly inhibited DEP-induced ERK and Akt phosphorylation, and subsequent IL-8 and IL-1β expression. Conclusion GSTM1 regulates DEP-induced IL-8 and IL-1β expression in primary human bronchial epithelial cells by modulation of ROS, ERK and Akt signaling.
Collapse
Affiliation(s)
- Weidong Wu
- Center for Environmental Medicine, Asthma, and Lung Biology, University of North Carolina, Chapel Hill, NC 27599, USA.
| | | | | | | | | |
Collapse
|